Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep ; 19(2): 307-320, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28402854

ABSTRACT

Hearing loss is widespread and persistent because mature mammalian auditory hair cells (HCs) are nonregenerative. In mice, the ability to regenerate HCs from surrounding supporting cells (SCs) declines abruptly after postnatal maturation. We find that combining p27Kip1 deletion with ectopic ATOH1 expression surmounts this age-related decline, leading to conversion of SCs to HCs in mature mouse cochleae and after noise damage. p27Kip1 deletion, independent of canonical effects on Rb-family proteins, upregulated GATA3, a co-factor for ATOH1 that is lost from SCs with age. Co-activation of GATA3 or POU4F3 and ATOH1 promoted conversion of SCs to HCs in adult mice. Activation of POU4F3 alone also converted mature SCs to HCs in vivo. These data illuminate a genetic pathway that initiates auditory HC regeneration and suggest p27Kip1, GATA3, and POU4F3 as additional therapeutic targets for ATOH1-mediated HC regeneration.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Cyclin-Dependent Kinase Inhibitor p27/genetics , GATA3 Transcription Factor/biosynthesis , Hearing Loss/genetics , Homeodomain Proteins/biosynthesis , Transcription Factor Brn-3C/biosynthesis , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Proliferation/genetics , Cochlea/growth & development , Cochlea/pathology , GATA3 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Hearing Loss/pathology , Homeodomain Proteins/genetics , Humans , Mice , Regeneration/genetics , Signal Transduction/genetics , Transcription Factor Brn-3C/genetics
2.
J Neurosci ; 37(5): 1320-1339, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28039372

ABSTRACT

The mammalian inner ear (IE) subserves auditory and vestibular sensations via highly specialized cells and proteins. Sensory receptor hair cells (HCs) are necessary for transducing mechanical inputs and stimulating sensory neurons by using a host of known and as yet unknown protein machinery. To understand the protein composition of these unique postmitotic cells, in which irreversible protein degradation or damage can lead to impaired hearing and balance, we analyzed IE samples by tandem mass spectrometry to generate an unbiased, shotgun-proteomics view of protein identities and abundances. By using Pou4f3/eGFP-transgenic mice in which HCs express GFP driven by Pou4f3, we FACS purified a population of HCs to analyze and compare the HC proteome with other IE subproteomes from sensory epithelia and whole IE. We show that the mammalian HC proteome comprises hundreds of uniquely or highly expressed proteins. Our global proteomic analysis of purified HCs extends the existing HC transcriptome, revealing previously undetected gene products and isoform-specific protein expression. Comparison of our proteomic data with mouse and human databases of genetic auditory/vestibular impairments confirms the critical role of the HC proteome for normal IE function, providing a cell-specific pool of candidates for novel, important HC genes. Several proteins identified exclusively in HCs by proteomics and verified by immunohistochemistry map to human genetic deafness loci, potentially representing new deafness genes. SIGNIFICANCE STATEMENT: Hearing and balance rely on specialized sensory hair cells (HCs) in the inner ear (IE) to convey information about sound, acceleration, and orientation to the brain. Genetically and environmentally induced perturbations to HC proteins can result in deafness and severe imbalance. We used transgenic mice with GFP-expressing HCs, coupled with FACS sorting and tandem mass spectrometry, to define the most complete HC and IE proteome to date. We show that hundreds of proteins are uniquely identified or enriched in HCs, extending previous gene expression analyses to reveal novel HC proteins and isoforms. Importantly, deafness-linked proteins were significantly enriched in HCs, suggesting that this in-depth proteomic analysis of IE sensory cells may hold potential for deafness gene discovery.


Subject(s)
Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Hair Cells, Auditory, Inner/metabolism , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Proteomics , Amino Acid Sequence , Animals , Chromosome Mapping , Female , Hair Cells, Auditory, Inner/chemistry , Hearing Disorders/genetics , Hearing Disorders/pathology , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/chemistry , Tandem Mass Spectrometry , Transcription Factor Brn-3C/biosynthesis , Transcription Factor Brn-3C/genetics , Transcriptome , Vestibular Diseases/genetics , Vestibular Diseases/pathology
3.
J Comp Neurol ; 520(4): 742-55, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-21935940

ABSTRACT

Several subtypes of melanopsin-expressing, intrinsically photosensitive retinal ganglion cells (ipRGCs) have been reported. The M1 type of ipRGCs exhibit distinct properties compared with the remaining (non-M1) cells. They differ not only in their soma size and dendritic arbor, but also in their physiological properties, projection patterns, and functions. However, it is not known how these differences arise. We tested the hypothesis that M1 and non-M1 cells express Brn3 transcription factors differentially. The Brn3 family of class IV POU-domain transcription factors (Brn3a, Brn3b, and Brn3c) is involved in the regulation of differentiation, dendritic stratification, and axonal projection of retinal ganglion cells during development. By using double immunofluorescence for Brn3 transcription factors and melanopsin, and with elaborate morphometric analyses, we show in mouse retina that neither Brn3a nor Brn3c are expressed in ipRGCs. However, Brn3b is expressed in a subset of ipRGCs, particularly those with larger somas and lower melanopsin levels, suggesting that Brn3b is expressed preferentially in the non-M1 cells. By using dendritic stratification to distinguish M1 from non-M1 cells, we found that whereas nearly all non-M1 cells expressed Brn3b, a vast majority of the M1 cells were negative for Brn3b. Interestingly, in the small proportion of the M1 cells that did express Brn3b, the expression level of Brn3b was significantly lower than in the non-M1 cells. These results provide insights about how expression of specific molecules in a ganglion cell could be linked to its role in visual function.


Subject(s)
Retinal Ganglion Cells/physiology , Transcription Factor Brn-3/biosynthesis , Transcription Factor Brn-3/genetics , Animals , Cell Count , Dendrites/metabolism , Fluorescent Antibody Technique, Indirect , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Image Processing, Computer-Assisted , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Fluorescence , Retinal Ganglion Cells/metabolism , Rod Opsins/metabolism , Transcription Factor Brn-3A/biosynthesis , Transcription Factor Brn-3A/genetics , Transcription Factor Brn-3B/biosynthesis , Transcription Factor Brn-3B/genetics , Transcription Factor Brn-3C/biosynthesis , Transcription Factor Brn-3C/genetics
4.
Neuroscience ; 197: 48-64, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21958861

ABSTRACT

The POU-domain transcription POU4F3 is expressed in the sensory cells of the inner ear. Expression begins shortly after commitment to the hair cell (HC) fate, and continues throughout life. It is required for terminal HC differentiation and survival. To explore regulation of the murine Pou4f3 gene, we linked enhanced green fluorescent protein (eGFP) to 8.5 kb of genomic sequence 5' to the start codon in transgenic mice. eGFP was uniformly present in all embryonic and neonatal HCs. Expression of eGFP was also observed in developing Merkel cells and olfactory neurons as well as adult inner and vestibular HCs, mimicking the normal expression pattern of POU4F3 protein, with the exception of adult outer HCs. Apparently ectopic expression was observed in developing inner ear neurons. On a Pou4f3 null background, the transgene produced expression in embryonic HCs which faded soon after birth both in vivo and in vitro. Pou4f3 null HCs treated with caspase 3 and 9 inhibitors survived longer than untreated HCs, but still showed reduced expression of eGFP. The results suggest the existence of separate enhancers for different HC types, as well as strong autoregulation of the Pou4f3 gene. Bioinformatic analysis of four divergent mammalian species revealed three highly conserved regions within the transgene: 400 bp immediately 5' to the Pou4f3 ATG, a short sequence at -1.3 kb, and a longer region at -8.2 to -8.5 kb. The latter contained E-box motifs that bind basic helix-loop-helix (bHLH) transcription factors, including motifs activated by ATOH1. Cotransfection of HEK293 or VOT-E36 cells with ATOH1 and the transgene as a reporter enhanced eGFP expression when compared with the transgene alone. Chromatin immunoprecipitation of the three highly conserved regions revealed binding of ATOH1 to the distal-most conserved region. The results are consistent with regulation of Pou4f3 in HCs by ATOH1 at a distal enhancer.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , E-Box Elements/genetics , Gene Expression Regulation/genetics , Hair Cells, Auditory, Inner , Homeodomain Proteins/genetics , Transcription Factor Brn-3C/genetics , Animals , Cell Line , Chromatin Immunoprecipitation , DNA/genetics , Homeodomain Proteins/biosynthesis , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Transcription Factor Brn-3C/biosynthesis , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...