Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Eur Arch Otorhinolaryngol ; 281(7): 3577-3586, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38400873

ABSTRACT

PURPOSE: Hearing loss (HL) is often monogenic. The clinical importance of genetic testing in HL may further increase when gene therapy products become available. Diagnoses are, however, complicated by a high genetic and allelic heterogeneity, particularly of autosomal dominant (AD) HL. This work aimed to characterize the mutational spectrum of AD HL in Austria. METHODS: In an ongoing prospective study, 27 consecutive index patients clinically diagnosed with non-syndromic AD HL, including 18 previously unpublished cases, were analyzed using whole-exome sequencing (WES) and gene panels. Novel variants were characterized using literature and bioinformatic means. Two additional Austrian medical centers provided AD HL mutational data obtained with in-house pipelines. Other Austrian cases of AD HL were gathered from literature. RESULTS: The solve rate (variants graded as likely pathogenic (LP) or pathogenic (P)) within our cohort amounted to 59.26% (16/27). MYO6 variants were the most common cause. One third of LP/P variants were truncating variants in haploinsufficiency genes. Ten novel variants in HL genes were identified, including six graded as LP or P. In one cohort case and one external case, the analysis uncovered previously unrecognized syndromic presentations. CONCLUSION: More than half of AD HL cases analyzed at our center were solved with WES. Our data demonstrate the importance of genetic testing, especially for the diagnosis of syndromic presentations, enhance the molecular knowledge of genetic HL, and support other laboratories in the interpretation of variants.


Subject(s)
Exome Sequencing , Mutation , Humans , Austria , Male , Female , Prospective Studies , Adult , Child , Adolescent , Child, Preschool , Middle Aged , Young Adult , Genetic Testing/methods , Genes, Dominant , Aged , Hearing Loss/genetics , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/diagnosis , Infant
2.
Hum Genet ; 142(8): 1077-1089, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36445457

ABSTRACT

Loss-of-function variants in AP3D1 have been linked to Hermansky-Pudlak syndrome (HPS) 10, a severe multisystem disorder characterized by oculocutaneous albinism, immunodeficiency, neurodevelopmental delay, hearing loss (HL), and neurological abnormalities, fatal in early childhood. Here, we report a consanguineous family who presented with presumably isolated autosomal recessive (AR) HL. Whole-exome sequencing was performed on all core family members, and selected patients were screened using array-based copy-number analysis and karyotyping. Candidate variants were validated by Sanger sequencing and assessed in silico. A homozygous, likely pathogenic p.V711I missense variant in AP3D1 segregated with the HL. The family was characterized by thorough medical and laboratory examination. The HL was consistent across patients and accompanied by neurological manifestations in two brothers. The sole female patient was diagnosed with premature ovarian failure. Further findings, including mild neutropenia and reduced NK-cell cytotoxicity in some as well as brain alterations in all homozygous patients, were reminiscent of HPS10, though milder and lacking the characteristic albinism. Previously unrecognized, milder, isolated HL was identified in all heterozygous carriers. A protein model indicates that the variant interferes with protein-protein interactions. These results suggest that a missense variant alters inner-ear-specific functions leading to HL with mild HPS10-like symptoms of variable penetrance. Milder HL in heterozygous carriers may point towards semi-dominant inheritance of this trait. Since all previously reported HPS10 cases were pediatric, it is unknown whether the observed primary ovarian insufficiency recapitulates the subfertility in Ap3d1-deficient mice.


Subject(s)
Deafness , Hearing Loss, Sensorineural , Hermanski-Pudlak Syndrome , Male , Humans , Child, Preschool , Female , Animals , Mice , Hermanski-Pudlak Syndrome/diagnosis , Hermanski-Pudlak Syndrome/pathology , Mutation, Missense , Hearing Loss, Sensorineural/genetics , Carrier Proteins , Homozygote , Adaptor Protein Complex 3 , Adaptor Protein Complex delta Subunits , Adaptor Protein Complex beta Subunits
3.
Clin Otolaryngol ; 46(5): 1044-1049, 2021 09.
Article in English | MEDLINE | ID: mdl-33851515

ABSTRACT

OBJECTIVE: Identification of variations in tumour suppressor genes encoding the tetrameric succinate dehydrogenase (SDHx) mitochondrial enzyme complex may lead to personalised therapeutic concepts for the orphan disease, familial paraganglioma (PGL) type 1-5. We undertook to determine the causative variation in a family suffering from idiopathic early-onset (22 ± 2 years) head and neck PGL by PCR and Sanger sequencing. DESIGN: Prospective genetic study. SETTING: Tertiary Referral Otolaryngology Centre. PARTICIPANTS: Twelve family members. MAIN OUTCOME MEASURES: Main outcomes were clinical analysis and SDH genotyping RESULTS AND CONCLUSIONS: A novel heterozygous c.298delA frameshift variation in exon 3 of SDH subunit D (SDHD) was associated with a paternal transmission pattern of PGL in affected family members available to the study. Family history over five generations in adulthood indicated a variable penetrance for PGL inheritance in older generations. The c.298delA variant would cause translation of a 34-residue C-terminus distal to lysine residue 99 in the predicted transmembrane domain II of the full-length sequence p.(Thr100LeufsTer35) and would affect the translation products of all protein-coding SDHD isoforms containing transmembrane topologies required for positional integration in the inner mitochondrial membrane and complex formation. These results underly the importance of genetic screening for PGL also in cases of unclear inheritance, and variation carriers should benefit from screening and lifelong follow-up.


Subject(s)
Head and Neck Neoplasms/genetics , Paraganglioma/genetics , Succinate Dehydrogenase/genetics , Adult , Age of Onset , Aged, 80 and over , Austria , Exons , Female , Frameshift Mutation , Genetic Testing , Head and Neck Neoplasms/diagnostic imaging , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Paraganglioma/diagnostic imaging , Pedigree , Penetrance , Phenotype , Prospective Studies , Young Adult
4.
eNeurologicalSci ; 23: 100337, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33898793

ABSTRACT

"High" vagus nerve lesions are rare and refer to the region of the nerve from the jugular foramen through the branching of the auricular (Arnold's branch) and the pharyngeal branch. Rapid onset of vagus nerve palsy is observed predominantly in trauma, and rarely in inflammation. An insidious onset points to a neoplastic cause. The acute "high" vagus nerve lesion is characterized by a unilateral paralysis of the recurrent laryngeal nerve, an incomplete paresis of the soft palate and a transient inability to swallow. This is a case description of a 79-year-old woman who presented with painful swelling of the left ear and occipital headache, followed by inability to swallow for 3 weeks. A markedly elevated Varicella Zoster titer suggested a herpes virus infection.

5.
Otol Neurotol ; 42(6): e648-e657, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33710140

ABSTRACT

INTRODUCTION: Genetic hearing loss (HL) is often monogenic. Whereas more than half of autosomal recessive (AR) cases in Austria are caused by mutations in a single gene, no disproportionately frequent contributing genetic factor has been identified in cases of autosomal dominant (AD) HL. The genetic characterization of HL continues to improve diagnosis, genetic counseling, and lays a foundation for the development of personalized medicine approaches. METHODS: Diagnostic HL panel screening was performed in an Austrian multiplex family with AD HL, and segregation was tested with polymerase chain reaction and Sanger sequencing. In an independent approach, 18 unrelated patients with AD HL were screened for causative variants in all known HL genes to date and segregation was tested if additional family members were available. The pathogenicity of novel variants was assessed based on previous literature and bioinformatic tools such as prediction software and protein modeling. RESULTS: In six of the 19 families under study, candidate pathogenic variants were identified in MYO6, including three novel variants (p.Gln441Pro, p.Ser612Tyr, and p.Gln650ValfsTer7). Some patients carried more than one likely pathogenic variant in known deafness genes. CONCLUSION: These results suggest a potential high prevalence of MYO6 variants in Austrian cases of AD HL. The presence of multiple rare HL variants in some patients highlights the relevance of considering multiple-hit diagnoses for genetic counseling and targeted therapy design.


Subject(s)
Deafness , Hearing Loss , Austria/epidemiology , Humans , Mutation , Pedigree , Prevalence
6.
Exp Eye Res ; 205: 108497, 2021 04.
Article in English | MEDLINE | ID: mdl-33596443

ABSTRACT

Nanophthalmos-4 is a rare autosomal dominant disorder caused by two known variations in TMEM98. An Austrian Caucasian pedigree was identified suffering from nanophthalmos and late onset angle-closure glaucoma and premature loss of visual acuity. Whole exome sequencing identified segregation of a c.602G > C transversion in TMEM98 (p.Arg201Pro) as potentially causative. A protein homology model generated showed a TMEM98 structure comprising α4, α5/6, α7 and α8 antiparallel helix bundles and two predicted transmembrane domains in α1 and α7 that have been confirmed in vitro. Both p.Arg201Pro and the two missense variations representing proline insertions identified previously to cause nanophthalmos-4 (p.Ala193Pro and p.His196Pro) are located in the charge polarized helix α8 (p.183-p210). Stability of the C-terminal alpha helical structure of TMEM98 is therefore essential to prevent the development of human nanophthalmos-4. Precise molecular diagnosis could lead to the development of tailored therapies for patients with orphan ocular disease.


Subject(s)
Glaucoma, Angle-Closure/genetics , Hyperopia/genetics , Membrane Proteins/genetics , Microphthalmos/genetics , Mutation, Missense , Vision Disorders/genetics , Visual Acuity/physiology , Adult , Aged, 80 and over , Amino Acid Substitution , Arginine , Female , Filtering Surgery , Glaucoma, Angle-Closure/physiopathology , Glaucoma, Angle-Closure/surgery , Humans , Hyperopia/physiopathology , Hyperopia/surgery , Lens Implantation, Intraocular , Male , Microphthalmos/physiopathology , Microphthalmos/surgery , Microscopy, Acoustic , Middle Aged , Pedigree , Phacoemulsification , Proline , Protein Conformation, alpha-Helical/genetics , Slit Lamp Microscopy , Vision Disorders/physiopathology , Exome Sequencing
7.
Front Cell Neurosci ; 14: 585669, 2020.
Article in English | MEDLINE | ID: mdl-33281559

ABSTRACT

Background: Hereditary hearing loss is a disorder with high genetic and allelic heterogeneity. Diagnostic screening of candidate genes commonly yields novel variants of unknown clinical significance. TBC1D24 is a pleiotropic gene associated with recessive DOORS syndrome, epileptic encephalopathy, myoclonic epilepsy, and both recessive and dominant hearing impairment. Genotype-phenotype correlations have not been established to date but could facilitate diagnostic variant assessment and elucidation of pathomechanisms. Methods and Results: Whole-exome and gene panel screening identified a novel (c.919A>C; p.Asn307His) causative variant in TBC1D24 in two unrelated Caucasian families with Autosomal dominant (AD) nonsyndromic late-onset hearing loss. Protein modeling on the Drosophila TBC1D24 ortholog Skywalker crystal structure showed close interhelix proximity (6.8Å) between the highly conserved residue p.Asn307 in α18 and the position of the single known pathogenic dominant variation (p.Ser178Leu) in α11 that causes a form of deafness with similar clinical characteristics. Conclusion: Genetic variants affecting two polar hydrophilic residues in neighboring helices of TBC1D24 cause AD nonsyndromic late-onset hearing loss. The spatial proximity of the affected residues suggests the first genotype-phenotype association in TBC1D24-related disorders. Three conserved residues in α18 contribute to the formation of a functionally relevant cationic phosphoinositide binding pocket that regulates synaptic vesicle trafficking which may be involved in the molecular mechanism of disease.

8.
Clin Exp Otorhinolaryngol ; 12(4): 405-411, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31220907

ABSTRACT

OBJECTIVES: Hereditary hemorrhagic telangiectasia (HHT) is a rare autosomal dominant genetic disorder characterized by pathogenic blood vessel development and maintenance. HHT type 1 (HHT1) and type 2 (HHT2) are caused by variants in endoglin (ENG) and activin receptor-like kinase-1 (ACVRL1), respectively. The aim of this study was to identify the spectrum of pathogenic variants in ENG and ACVRL1 in Austrian HHT families. METHODS: In this prospective study, eight Austrian HHT families were screened for variants in ENG and ACVRL1 by polymerase chain reaction amplification and sequencing of DNA isolated from peripheral blood. RESULTS: Heterozygous variants were identified in all families under study. HHT1 was caused by a novel c.816+1G>A splice donor variant, a novel c.1479C>A nonsense (p.Cys493X) variant and a published c.1306C>T nonsense (p.Gln436X) variant in ENG. Variants found in ACVRL1 were novel c.200G>C (p.Arg67Pro) and known c.772G>A (p.Gly258Ser) missense variants in highly conserved residues, a known heterozygous c.100dupT frameshift (p.Cys34Leufs*4) and the known c.1204G>A missense (p.Gly402Ser) and c.1435C>T nonsense (p.Arg479X) variants as causes of HHT2. CONCLUSION: Novel and published variants in ENG (37.5%) and ACVRL1 (62.5%) were exclusively identified as the cause of HHT in an Austrian patient cohort. Identification of novel causative genetics variants should facilitate the development of tailored therapeutical applications in the future treatment of autosomal dominant HHT.

9.
Wien Klin Wochenschr ; 130(9-10): 299-306, 2018 May.
Article in English | MEDLINE | ID: mdl-28733840

ABSTRACT

BACKGROUND: Non-syndromic autosomal dominant hearing impairment is characteristically postlingual in onset. Genetic diagnostics are essential for genetic counselling, disease prognosis and understanding of the molecular mechanisms of disease. To date, 36 causative genes have been identified, many in only individual families. Gene selection for genetic screening by traditional methods and genetic diagnosis in autosomal dominant patients has therefore been fraught with difficulty. Whole-exome sequencing provides a powerful tool to analyze all protein-coding genomic regions in parallel, thus allowing the comprehensive screening of all known genes and associated alterations. METHODS: In this study, a previously undiagnosed late-onset progressive autosomal dominant hearing loss in an Austrian family was investigated by means of whole-exome sequencing. Results were confirmed by Sanger sequencing. RESULTS: A previously described c.151C>T missense (p.Pro51Ser) mutation in the LCCL (limulus factor C, cochlin, late gestation lung protein Lgl1) domain of the cochlin gene (COCH) was identified as causative and segregated with disease in five members of the family. Molecular diagnostics led to the decision to perform cochlear implantation in an index patient who subsequently showed excellent postoperative auditory performance. The c.151C>T mutation was not found in 18 screened Austrian families with autosomal dominant hearing loss but was represented alongside other known pathogenic mutant COCH alleles in the Genome Aggregation Database (gnomAD) in European populations. A combined allele frequency of 0.000128 implies an orphan disease frequency for COCH-induced hearing loss of 1:3900 in Europe. CONCLUSIONS: Exome sequencing successfully resolved the genetic diagnosis in a family suffering from autosomal dominant hearing impairment and allowed prediction of purported auditory outcome after cochlear implantation in an index patient. Personalized treatment approaches based on the molecular mechanisms of disease may become increasingly important in the future.


Subject(s)
Exome Sequencing , Hearing Loss, Sensorineural , Austria , Europe , Exome , Extracellular Matrix Proteins , Female , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/rehabilitation , Humans , Male , Mutation , Pedigree , Exome Sequencing/methods
10.
Eur Arch Otorhinolaryngol ; 274(10): 3619-3625, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28821934

ABSTRACT

Bi-allelic variations in the gap junction protein beta-2 (GJB2) gene cause up to 50% of cases of newborn hearing loss. Heterozygous pathogenic GJB2 variations are also fivefold overrepresented in idiopathic patient groups compared to the normal-hearing population. Whether hearing loss in this group is due to unidentified additional variations within GJB2 or variations in other deafness genes is unknown in most cases. Whole-exome sequencing offers an effective approach in the search for causative variations in patients with Mendelian diseases. In this prospective genetic cohort study, we initially investigated a family of Turkish origin suffering from congenital autosomal recessive hearing loss. An index patient and his normal-hearing father, both bearing a single heterozygous pathogenic c.262G>T (p.Ala88Ser) GJB2 transversion as well as the normal-hearing mother were investigated by means of whole-exome sequencing. Subsequently the genetic screening was extended to a hearing-impaired cohort of 24 families of Turkish origin. A homozygous missense c.5492G>T transversion (p.Gly1831Val) in the Myosin 15a gene, previously linked to deafness, was identified as causative in the index family. This very rare variant is not listed in any population in the Genome Aggregation Database. Subsequent screening of index patients from additional families of Turkish origin with recessive hearing loss identified the c.5492G>T variation in an additional family. Whole-exome sequencing may effectively identify the causes of idiopathic hearing loss in patients bearing heterozygous GJB2 variations.


Subject(s)
Connexins/genetics , Hearing Loss, Sensorineural , Adult , Austria/epidemiology , Connexin 26 , Female , Genetic Testing/methods , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/epidemiology , Hearing Loss, Sensorineural/genetics , Humans , Infant, Newborn , Male , Mutation , Prospective Studies , Turkey , Exome Sequencing/methods
11.
Int Forum Allergy Rhinol ; 7(3): 293-299, 2017 03.
Article in English | MEDLINE | ID: mdl-28297555

ABSTRACT

BACKGROUND: Use of reliable grading scores to measure epistaxis severity in hereditary hemorrhagic telangiectasia (HHT) is essential in clinical routine and for scientific purposes. For practical reasons, visual analog scale (VAS) scoring and the Epistaxis Severity Score (ESS) are widely used. VAS scores are purely subjective, and a potential shortcoming of the ESS is that it is based on self-reported anamnestic bleeding data. The aim of this study was to validate the level of correlation between VAS scores, the ESS, and actual bleeding events, based on detailed epistaxis diaries of patients. METHODS: Records from daily epistaxis diaries maintained by 16 HHT patients over 112 consecutive days were compared with the monthly ESS and daily VAS scores in the corresponding time period. The Spearman rank correlation coefficient, analysis of variance models, and multiple R2 measures were used for statistical analysis. RESULTS: Although the ESS and VAS scores generally showed a high degree of correlation with actual bleeding events, mild events were underrepresented in both scores. CONCLUSIONS: Our results highlight the usefulness of the ESS as a standard epistaxis score in cohorts with moderate to severe degrees of epistaxis. The use of detailed epistaxis diaries should be considered when monitoring patients and cohorts with mild forms of HHT.


Subject(s)
Epistaxis/pathology , Medical Records , Telangiectasia, Hereditary Hemorrhagic/pathology , Adult , Aged , Epistaxis/etiology , Female , Humans , Male , Middle Aged , Severity of Illness Index , Telangiectasia, Hereditary Hemorrhagic/complications , Visual Analog Scale
12.
Otol Neurotol ; 38(2): 173-179, 2017 02.
Article in English | MEDLINE | ID: mdl-27861301

ABSTRACT

BACKGROUND: Heterozygous mutations in GJB2 (MIM: 121011) encoding the gap junction protein connexin 26 are overrepresented in patient groups suffering from nonsyndromic sensorineural hearing impairment (HI) implying the involvement of additional genetic factors. Mutations in SLC26A4 (MIM: 605646), encoding the protein pendrin can cause both Pendred syndrome and autosomal recessive, nonsyndromic HI locus 4 type sensorineural HI (MIM: 600791). OBJECTIVES: Aim of this study was to investigate the role of SLC26A4 coding mutations in a nonsyndromic hearing impairment (NSHI) patient group bearing heterozygous GJB2 35delG mutations. DESIGN: We analyzed the 20 coding exons of SLC26A4 in a group of patients (n = 15) bearing heterozygous 35delG mutations and exclusively suffering from congenital HI. RESULTS: In a case of bilateral congenital hearing loss we identified a rare, novel SLC26A4 exon 2 splice donor mutation (c.164+1delG) predicted to truncate pendrin in the first cytoplasmic domain, as a compound heterozygote with the pathogenic missense mutation c.1061T>C (p.354F>S; rs111033243). CONCLUSIONS: Screening for SLC26A4 mutations may identify the genetic causes of hearing loss in patients bearing heterozygous mutations in GJB2. HYPOTHESIS: SLC26A4 coding mutations are genetic causes for nonsyndromic HI in patients bearing heterozygous GJB2 35delG mutations.


Subject(s)
Hearing Loss, Bilateral/genetics , Hearing Loss, Sensorineural/genetics , Membrane Transport Proteins/genetics , Adult , Child , Child, Preschool , Connexin 26 , Connexins/genetics , Exons , Female , Heterozygote , Humans , Infant , Male , Middle Aged , Mutation , Sulfate Transporters , Young Adult
13.
Acta Otolaryngol ; 137(4): 356-360, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27827000

ABSTRACT

CONCLUSION: Alterations within a novel putative Exon 1a within the gap junction beta 2 (GJB2) gene may play a role in the development of genetic hearing impairment in Austria. OBJECTIVES: Mutations in the GJB2 gene are the most common cause of hereditary sensorineural deafness. Genome-wide screening for alternative transcriptional start sites in the human genome has revealed the presence of an additional GJB2 exon (E1a). This study tested the hypothesis of whether alternative GJB2 transcription involving E1a may play a role in the development of congenital sensorineural deafness in Austria. METHODS: GJB2 E1a and flanking regions were sequenced in randomized normal hearing control subjects and three different patient groups with non-syndromic hearing impairment (NSHI), and bioinformatic analysis was performed. Statistical analysis of disease association was carried out using the Cochran-Armitage test for trend. RESULTS: A single change 2410 bp proximal to the translational start site (c.-2410T > C, rs7994748, NM_004004.5:c.-23 + 792T > C) was found to be significantly associated with the common c.35delG GJB2 mutation (p = .009). c.35delG in combination with c.-2410CC occurred at a 6.9-fold increased frequency compared to the control group. Additionally, one patient with idiopathic congenital hearing loss was found to be homozygous c.-2410CC.


Subject(s)
Connexins/genetics , Hearing Loss, Sensorineural/genetics , Alternative Splicing , Austria , Base Sequence , Case-Control Studies , Connexin 26 , Exons , Gene Frequency , Genetic Testing , Hearing Loss, Sensorineural/congenital , Hearing Loss, Sensorineural/diagnosis , Heterozygote , Humans , Mass Screening , Polymorphism, Genetic
14.
Head Neck ; 38 Suppl 1: E1515-20, 2016 04.
Article in English | MEDLINE | ID: mdl-26669822

ABSTRACT

BACKGROUND: Primary total laryngopharyngectomy is the treatment of choice in many cases of locally advanced hypopharyngeal and laryngeal cancer. Development of pharyngocutaneous fistulae is the most common postoperative complication. A recent Danish study showed significantly increased rates of anastomosal leakage after colorectal resection in patients receiving diclofenac treatment. METHODS: We retrospectively analyzed 67 patients after primary total laryngopharyngectomy to determine whether diclofenac increases the risk for development of pharyngocutaneous fistula analogously to leakage in the colorectal area. RESULTS: The fistula rate in the total study population (n = 67) was 19.4%. In the group receiving diclofenac postoperatively (n = 31), the fistula rate was 25.8%. In the patient group not receiving diclofenac (n = 36), the fistula rate was 13.9% (p = .219). CONCLUSION: Our results suggest that cyclooxygenase-2 (COX-2) selective nonsteroidal anti-inflammatory drugs (NSAIDs) should be administered with caution after laryngopharyngectomy. Additional studies on larger cohorts are required to further evaluate our findings. © 2015 Wiley Periodicals, Inc. Head Neck 38: E1515-E1520, 2016.


Subject(s)
Cutaneous Fistula/prevention & control , Diclofenac/therapeutic use , Laryngectomy/adverse effects , Pharyngectomy/adverse effects , Cutaneous Fistula/drug therapy , Female , Humans , Laryngeal Neoplasms/surgery , Male , Middle Aged , Pharyngeal Neoplasms/surgery , Postoperative Care , Postoperative Complications , Retrospective Studies
15.
Eur Arch Otorhinolaryngol ; 272(1): 229-32, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25085637

ABSTRACT

The objective of this study was to investigate the relevance of routine assessment of c.-259C>T in the Austrian newborn screening program. Homozygous and compound heterozygous mutations in the coding region of the human gene encoding gap junction protein GJB2 (Connexin 26) cause up to 50 % of neonatal autosomal recessive non-syndromic hearing impairment identified in Caucasian newborn screening programs. More recently, a null mutation in the GC box of the GJB2 basal promoter c.-259C>T has been described which causes hearing impairment by completely suppressing GJB2 promoter activity. We determined the occurrence of c.-259C>T in cases of non-syndromic hearing impairment lacking known pathogenic alterations in GJB2 (n = 43), a non-syndromic hearing impaired patient group (n = 15) bearing the heterozygous GJB2 mutations c.35delG, c.[79G>A];[341A>G] (p. [V27I];[E114G]), c.109G>A (p.V37I), c.154G>C (p.V52L), c.262G>T (p.A88S), c.269T>C (p.L90P) and c.551G>C (p.R184P) and in a normal hearing group lacking alterations in GJB2 (n = 50). In the analyzed groups, no occurrence of c.-259C>T was found. The c.-259C>T mutation, previously described as -3438C>T, is not a common cause of non-syndromic hearing impairment alone or together with heterozygous pathogenic GJB2 mutations that are statistically overrepresented in non-syndromic hearing impaired patient groups. Screening of newborns for c.-259C>T is therefore unlikely to be commonly found in Austrian NSHI patients but could make a significant contribution to non-syndromic hearing impairment in other populations.


Subject(s)
Connexins/genetics , DNA/genetics , Hearing Loss/genetics , Mutation , Austria/epidemiology , Connexin 26 , Connexins/metabolism , Female , Hearing Loss/epidemiology , Humans , Infant, Newborn , Male , Polymerase Chain Reaction , Prevalence , Promoter Regions, Genetic
16.
Wien Klin Wochenschr ; 127(11-12): 440-4, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25409950

ABSTRACT

BACKGROUND: Hearing loss is the most common sensory disorder in developed countries and leads to a severe reduction in quality of life. In this uncontrolled case series, we evaluated the auditory development in patients suffering from congenital nonsyndromic hearing impairment related to preterm birth. METHODS: Six patients delivered preterm (25th-35th gestational weeks) suffering from mild to profound congenital nonsyndromic hearing impairment, descending from healthy, nonconsanguineous parents and were evaluated by otoacoustic emissions, tympanometry, brainstem-evoked response audiometry, and genetic testing. All patients were treated with hearing aids, and one patient required cochlear implantation. RESULTS: One preterm infant (32nd gestational week) initially presented with a 70 dB hearing loss, accompanied by negative otoacoustic emissions and normal tympanometric findings. The patient was treated with hearing aids and displayed a gradual improvement in bilateral hearing that completely normalized by 14 months of age accompanied by the development of otoacoustic emission responses. Conclusions We present here for the first time a fully documented preterm patient with delayed auditory pathway maturation and normalization of hearing within 14 months of birth. Although rare, postpartum development of the auditory system should, therefore, be considered in the initial stages for treating preterm hearing impaired patients.


Subject(s)
Correction of Hearing Impairment/instrumentation , Correction of Hearing Impairment/methods , Developmental Disabilities/diagnosis , Developmental Disabilities/rehabilitation , Hearing Disorders/congenital , Hearing Disorders/diagnosis , Female , Hearing Aids , Humans , Infant , Infant, Newborn , Infant, Premature , Male , Treatment Outcome
17.
Audiol Neurootol ; 19(3): 203-9, 2014.
Article in English | MEDLINE | ID: mdl-24801666

ABSTRACT

Norrie disease is a rare, X-linked genetic syndrome characterized by combined congenital blindness and progressive hearing impairment. Norrie disease is caused by alterations in the NDP gene encoding the growth factor norrin that plays a key role in vascular development and stabilization of the eye, inner ear and brain. We identified a family with 3 affected deafblind males and a single female carrier presenting with a serous retinal detachment but normal hearing. Genetic analysis revealed a novel c.277T>C missense mutation causing the substitution of a hydrophobic cysteine to a hydrophilic arginine [p.(Cys93Arg)] within the highly conserved cysteine knot domain of the norrin protein. These results should expand the scope for amniocentesis and genetic testing for Norrie disease which is gaining in importance due to novel postnatal therapeutic concepts to alleviate the devastating retinal symptoms of Norrie disease.


Subject(s)
Blindness/congenital , Eye Proteins/genetics , Mutation, Missense , Nerve Tissue Proteins/genetics , Nervous System Diseases/genetics , Spasms, Infantile/genetics , Blindness/genetics , Family , Female , Genetic Diseases, X-Linked , Genetic Testing , Humans , Male , Pedigree , Retinal Degeneration
18.
Int J Pediatr Otorhinolaryngol ; 78(5): 837-42, 2014 May.
Article in English | MEDLINE | ID: mdl-24636747

ABSTRACT

OBJECTIVES: Similar to other zona pellucida mutations in the alpha-tectorin (TECTA) gene, the p.Y1870C alteration in DFNA8/12 causes prelingual, nonsyndromic, autosomal dominant hearing loss. Here we investigated the effect of p.Y1870C on reverse transduction by audiometric studies in the family. METHODS: Pure tone audiometry, brainstem evoked response audiometry, the Freiburger test for speech understanding and transient evoked and distortion product otoacoustic emissions were assessed in three available affected members bearing p.Y1870C. RESULTS: Pure tone audiometry showed U-shaped curves with moderate to severe degrees of hearing impairment confirmed by brainstem evoked response audiometry. Transient evoked and distortion product otoacoustic emissions were completely absent in all affected family members whereas word recognition scores were up to 95%. CONCLUSIONS: Although the missense p.Y1870C TECTA mutation leads to complete failure of the cochlear amplifier in humans, very high speech perception scores can be achieved with appropriate therapy.


Subject(s)
Evoked Potentials, Auditory, Brain Stem/genetics , Extracellular Matrix Proteins/genetics , Hearing Loss, Sensorineural/diagnosis , Otoacoustic Emissions, Spontaneous/genetics , Adult , Audiometry, Pure-Tone/methods , Austria , Female , GPI-Linked Proteins/genetics , Hearing Loss, Sensorineural/genetics , Heterozygote , Humans , Male , Middle Aged , Mutation, Missense , Pedigree , Speech Perception , Young Adult
19.
Otol Neurotol ; 34(4): 650-6, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23640091

ABSTRACT

HYPOTHESIS: Additional genetic changes in the regulatory region of the human GJB2 gene encoding the gap junction protein (Connexin 26) may contribute to sensorineural hearing loss. BACKGROUND: Mutations in GJB2 cause up to 50% of autosomal recessive nonsyndromic hearing impairment (NSHI). METHODS: In the present study, we screened the putative 5' GJB2 regulatory region for novel alterations. RESULTS: In idiopathic familial cases of NSHI lacking known pathogenic alterations in GJB2, we identified a T→C transition (refSNP: rs117685390) in a putative transcription factor binding sequence 228 bp proximal to the transcriptional start site at a homozygous frequency of 0.125 (n = 40), significantly overrepresented in comparison to the homozygous allele frequencies of 0.043 in the normal-hearing Caucasian population (n = 211; p < 0.001). In a NSHI family, inheritance of the rs117685390 C allele segregated on independent chromosomes with NSHI in conjunction with heterozygous inheritance of c.35delG, the most common Caucasian mutation in the GJB2 coding region. In a patient group (n = 32) bearing heterozygous pathogenic c.35delG mutations, - rs117685390 C allele homozygosity was also highly overrepresented (0.25; p < 0.001) and not exclusively linked to the c.35delG mutation in cis in patients homozygous for c.35delG. However, in the majority of NSHI homozygous c.35delG chromosomes examined (91/94), c.35delG homozygosity was linked to the rs117685390 C allele in cis. CONCLUSION: These results suggest that the rs117685390 C allele could represent a biomarker for the development of NSHI in Caucasian populations and may be included in risk assessment for the development of NSHI.


Subject(s)
Connexins/genetics , Hearing Loss/genetics , Polymorphism, Single Nucleotide , Adult , Alleles , Connexin 26 , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Hearing Loss, Sensorineural/genetics , Homozygote , Humans , Male , Mutation
20.
Wien Klin Wochenschr ; 123(7-8): 241-4, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21479651

ABSTRACT

INTRODUCTION: Tonsillectomy is one of the most common surgical procedures in Otorhinolaryngology and approximately 500 tonsillectomies are performed annually at the University Hospital of Vienna. Substantial postoperative bleeding is observed in 1.8% of cases, which is comparable to frequencies of 2-4% reported in other studies. Currently, routine pre-surgical coagulation investigations to predict bleeding status do not include the analysis of individual coagulation factors. MATERIAL AND METHODS: Within 2007 complete coagulation diagnostics were carried out in three patients presenting with recurrent (2-7 times) post-tonsillectomy hemorrhage but normal pre-surgical coagulation status parameters. RESULTS: After the appearance of recurrent late bleeding, single factor diagnostic analysis revealed the causes to be factor XIIa deficiency, reduced factor XIIIa or von Willebrand disease. Recurrent late bleeding was stopped by the application of coagulation active plasma. CONCLUSION: This study shows that, even if routine diagnostics show normal pre-surgical coagulation findings, the investigation of single factors in cases of post-surgical late bleeding may identify coagulation disorders allowing the application of tailored therapy. Including such analysis in pre-operative diagnostics could therefore significantly help to limit postoperative bleeding.


Subject(s)
Blood Coagulation Disorders/blood , Blood Coagulation Factors/analysis , Hemorrhage/blood , Hemorrhage/etiology , Tonsillectomy/adverse effects , Adult , Blood Coagulation Disorders/etiology , Female , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...