Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
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
3.
Biomed Res Int ; 2018: 5370802, 2018.
Article in English | MEDLINE | ID: mdl-29850532

ABSTRACT

Hereditary nonsyndromic hearing loss is extremely heterogeneous. Mutations in the POU class 4 transcription factor 3 (POU4F3) are known to cause autosomal dominant nonsyndromic hearing loss linked to the loci of DFNA15. In this study, we describe a pathogenic missense mutation in POU4F3 in a four-generation Chinese family (6126) with midfrequency, progressive, and postlingual autosomal dominant nonsyndromic hearing loss (ADNSHL). By combining targeted capture of 129 known deafness genes, next-generation sequencing, and bioinformatic analysis, we identified POU4F3 c.602T>C (p.Leu201Pro) as the disease-causing variant. This variant cosegregated with hearing loss in other family members but was not detected in 580 normal controls or the ExAC database and could be classified as a "pathogenic variant" according to the American College of Medical Genetics and Genomics guidelines. We conclude that POU4F3 c.602T>C (p.Leu201Pro) is related to midfrequency hearing loss in this family. Routine examination of POU4F3 is necessary for the genetic diagnosis of midfrequency hearing loss.


Subject(s)
Asian People/genetics , Hearing Loss, Sensorineural/genetics , Homeodomain Proteins/genetics , Mutation, Missense/genetics , Transcription Factor Brn-3C/genetics , Adult , Aged , Amino Acid Sequence , Base Sequence , Child, Preschool , DNA Mutational Analysis , Family , Female , Homeodomain Proteins/chemistry , Humans , Middle Aged , Pedigree , Transcription Factor Brn-3C/chemistry
4.
Hum Mutat ; 29(4): 545-54, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18228599

ABSTRACT

In a Dutch pedigree suffering from autosomal dominant nonsyndromic hearing impairment (ADNSHI), linkage was found to the locus for DFNA15, with a two-point logarithm of the odds (LOD) score of 5.1. Sequence analysis of the POU4F3 gene that is involved in DFNA15 revealed the presence of a missense mutation (c.865C>T), segregating with the deafness in this family. The mutation is predicted to result in the substitution of a phenylalanine residue for a leucine residue (p.L289F) in the POU homeodomain of the transcription factor POU4F3. Mutation analysis of the POU4F3 gene in 30 patients suffering from dominantly inherited hearing impairment revealed a second novel missense mutation (c.668T>C), resulting in the substitution of a proline for a leucine residue (p.L223P) within the POU-specific DNA-binding domain of the protein. In a computer model describing the structure of the two DNA-binding domains, the alterations are predicted to affect the tertiary structure of these domains. Transient transfection studies showed that whereas the wild-type POU4F3 is located almost exclusively in the nucleus, part of the mutant proteins was also present in the cytoplasm. In addition, both mutant proteins showed greatly reduced capability for binding to DNA as well as transcriptionally activating reporter gene expression. Together, our results describe the identification of the first missense mutations in POU4F3 causing DFNA15. Furthermore, mutations in this gene do not seem to be a rare cause of hearing impairment in the Dutch population, and the POU4F3 gene may thus be suitable for implementation in diagnostic testing.


Subject(s)
Hearing Loss/genetics , Hearing Loss/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mutation, Missense , Transcription Factor Brn-3C/genetics , Transcription Factor Brn-3C/metabolism , Adult , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , Binding Sites/genetics , COS Cells , Chlorocebus aethiops , DNA/metabolism , DNA Primers/genetics , Female , Genes, Dominant , Genetic Linkage , Homeodomain Proteins/chemistry , Humans , Male , Middle Aged , Models, Molecular , Molecular Sequence Data , Pedigree , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Subcellular Fractions/metabolism , Transcription Factor Brn-3C/chemistry , Transcription, Genetic , Transfection
5.
Int J Comput Biol Drug Des ; 1(3): 295-301, 2008.
Article in English | MEDLINE | ID: mdl-20054994

ABSTRACT

The POU4F3 transcription factor is expressed in the cochlear and vestibular hair cells of the inner ear and its targeted deletion results in a loss of inner ear hair cells. The DFNA15 truncation mutation has been demonstrated to result in a loss of transcriptional activity, but an increase in the stability of the protein. Molecular modelling is utilised to propose a mechanism of stability enhancement, via an interaction between the truncated POU(HD) domain and the POU(S) domain of the transcription factor.


Subject(s)
Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Transcription Factor Brn-3C/chemistry , Transcription Factor Brn-3C/genetics , Computational Biology , Computer Simulation , Databases, Protein , Half-Life , Homeodomain Proteins/metabolism , Humans , In Vitro Techniques , Models, Molecular , Protein Interaction Domains and Motifs , Protein Stability , Sequence Deletion , Static Electricity , Structural Homology, Protein , Thermodynamics , Transcription Factor Brn-3C/metabolism
6.
Brain Res Mol Brain Res ; 141(2): 174-80, 2005 Nov 30.
Article in English | MEDLINE | ID: mdl-16226339

ABSTRACT

Barhl1 and Brn-3c have been identified as transcription factors that are essential for survival and maintenance of hair cells of the inner ear. Little is known about the mechanism of how Brn-3c or Barhl1 may regulate transcription in the inner ear. In this study, the transcriptional function of both Brn-3c and Barhl1 was investigated in the organ-of-Corti-derived cell lines, OC-1 and OC-2. We examined regulatory domains in these transcription factors by linking regions of Barhl1 and Brn-3c to the DNA binding domain of the heterologous transcription factor GAL4 and assayed their effect on a heterologous promoter containing GAL4 DNA binding sites by co-transfection into OC-1 and OC-2 cell lines. Brn-3c was found to contain an independent N-terminal activation domain that is sufficient to activate gene transcription in the organ of corti derived cell lines. Barhl1 on the other hand was found to act as a transcriptional repressor with repressive activity not restricted to a particular domain of Barhl1. In addition, we analyzed the effect of Barhl1 on the promoters of the neurotrophin genes NT-3 and BDNF in OC-1 and OC-2 cell lines. However, Barhl1 was not found to directly regulate neurotrophin promoter constructs in these cells.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Gene Expression Regulation/physiology , Homeodomain Proteins/physiology , Nerve Tissue Proteins/physiology , Neurotrophin 3/genetics , Organ of Corti/cytology , Repressor Proteins/physiology , Transcription Factor Brn-3C/physiology , Transcription, Genetic/physiology , Animals , Binding Sites , Brain-Derived Neurotrophic Factor/biosynthesis , Cell Line , Genes, Reporter , Genes, Synthetic , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Mice , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurotrophin 3/biosynthesis , Promoter Regions, Genetic , Protein Structure, Tertiary , Recombinant Fusion Proteins/physiology , Repressor Proteins/chemistry , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor Brn-3C/chemistry , Transcription Factor Brn-3C/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...