Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 17(3): e0261530, 2022.
Article in English | MEDLINE | ID: mdl-35235570

ABSTRACT

Tasmanian devil (tde) mice are deaf and exhibit circling behaviour. Sensory hair cells of mutants show disorganised hair bundles with abnormally thin stereocilia. The origin of this mutation is the insertion of a transgene which disrupts expression of the Grxcr1 (glutaredoxin cysteine rich 1) gene. We report here that Grxcr1 exons and transcript sequences are not affected by the transgene insertion in tde homozygous (tde/tde) mice. Furthermore, 5'RACE PCR experiments showed the presence of two different transcripts of the Grxcr1 gene, expressed in both tde/tde and in wild-type controls. However, quantitative analysis of Grxcr1 transcripts revealed a significantly decreased mRNA level in tde/tde mice. The key stereociliary proteins ESPN, MYO7A, EPS8 and PTPRQ were distributed in hair bundles of homozygous tde mutants in a similar pattern compared with control mice. We found that the abnormal morphology of the stereociliary bundle was associated with a reduction in the size and Ca2+-sensitivity of the mechanoelectrical transducer (MET) current. We propose that GRXCR1 is key for the normal growth of the stereociliary bundle prior to the onset of hearing, and in its absence hair cells are unable to mature into fully functional sensory receptors.


Subject(s)
Hair Cells, Auditory
2.
Rheumatology (Oxford) ; 56(11): 2004-2014, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28968858

ABSTRACT

Objectives: RA patients receiving TNF inhibitors (TNFi) usually maintain their initial doses. The aim of the Optimizing Treatment with Tumour Necrosis Factor Inhibitors in Rheumatoid Arthritis trial was to evaluate whether tapering TNFi doses causes loss of clinical response. Methods: We enrolled RA patients receiving etanercept or adalimumab and a DMARD with DAS28 under 3.2 for over 3 months. Initially (months 0-6) patients were randomized to control (constant TNFi) or two experimental groups (tapering TNFi by 33 or 66%). Subsequently (months 6-12) control subjects were randomized to taper TNFi by 33 or 66%. Disease flares (DAS28 increasing ⩾0.6 with at least one additional swollen joint) were the primary outcome. Results: Two hundred and forty-four patients were screened, 103 randomized and 97 treated. In months 0-6 there were 8/50 (16%) flares in controls, 3/26 (12%) with 33% tapering and 6/21 (29%) with 66% tapering. Multivariate Cox analysis showed time to flare was unchanged with 33% tapering but was reduced with 66% tapering compared with controls (adjusted hazard ratio 2.81, 95% CI: 0.99, 7.94; P = 0.051). Analysing all tapered patients after controls were re-randomized (months 6-12) showed differences between groups: there were 6/48 (13%) flares with 33% tapering and 14/39 (36%) with 66% tapering. Multivariate Cox analysis showed 66% tapering reduced time to flare (adjusted hazard ratio 3.47, 95% CI: 1.26, 9.58; P = 0.016). Conclusion: Tapering TNFi by 33% has no impact on disease flares and appears practical in patients in sustained remission and low disease activity states. Trail registration: EudraCT, https://www.clinicaltrialsregister.eu, 2010-020738-24; ISRCTN registry, https://www.isrctn.com, 28955701.


Subject(s)
Adalimumab/administration & dosage , Antirheumatic Agents/administration & dosage , Arthritis, Rheumatoid/drug therapy , Etanercept/administration & dosage , Aged , Arthritis, Rheumatoid/physiopathology , Disease Progression , Female , Humans , Maintenance Chemotherapy , Male , Middle Aged , Multivariate Analysis , Proportional Hazards Models , Remission Induction , Treatment Outcome , Tumor Necrosis Factor-alpha/antagonists & inhibitors
3.
Hum Mol Genet ; 23(23): 6407-18, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25060954

ABSTRACT

Hearing function is known to be heritable, but few significant and reproducible associations of genetic variants have been identified to date in the adult population. In this study, genome-wide association results of hearing function from the G-EAR consortium and TwinsUK were used for meta-analysis. Hearing ability in eight population samples of Northern and Southern European ancestry (n = 4591) and the Silk Road (n = 348) was measured using pure-tone audiometry and summarized using principal component (PC) analysis. Genome-wide association analyses for PC1-3 were conducted separately in each sample assuming an additive model adjusted for age, sex and relatedness of subjects. Meta-analysis was performed using 2.3 million single-nucleotide polymorphisms (SNPs) tested against each of the three PCs of hearing ability in 4939 individuals. A single SNP lying in intron 6 of the salt-inducible kinase 3 (SIK3) gene was found to be associated with hearing PC2 (P = 3.7×10(-8)) and further supported by whole-genome sequence in a subset. To determine the relevance of this gene in the ear, expression of the Sik3 protein was studied in mouse cochlea of different ages. Sik3 was expressed in murine hair cells during early development and in cells of the spiral ganglion during early development and adulthood. Our results suggest a developmental role of Sik3 in hearing and may be required for the maintenance of adult auditory function.


Subject(s)
Hearing/genetics , Protein Kinases/genetics , Protein Kinases/metabolism , Age Factors , Animals , Cochlea/growth & development , Cochlea/metabolism , Genome-Wide Association Study , Humans , Mice, Inbred C57BL , Polymorphism, Single Nucleotide , White People
4.
PLoS One ; 9(1): e85352, 2014.
Article in English | MEDLINE | ID: mdl-24454846

ABSTRACT

Considerable progress has been made in identifying deafness genes, but still little is known about the genetic basis of normal variation in hearing function. We recently carried out a Genome Wide Association Study (GWAS) of quantitative hearing traits in southern European populations and found several SNPs with suggestive but none with significant association. In the current study, we followed up these SNPs to investigate which of them might show a genuine association with auditory function using alternative approaches. Firstly, we generated a shortlist of 19 genes from the published GWAS results. Secondly, we carried out immunocytochemistry to examine expression of these 19 genes in the mouse inner ear. Twelve of them showed distinctive cochlear expression patterns. Four showed expression restricted to sensory hair cells (Csmd1, Arsg, Slc16a6 and Gabrg3), one only in marginal cells of the stria vascularis (Dclk1) while the others (Ptprd, Grm8, GlyBP, Evi5, Rimbp2, Ank2, Cdh13) in multiple cochlear cell types. In the third step, we tested these 12 genes for replication of association in an independent set of samples from the Caucasus and Central Asia. Nine out of them showed nominally significant association (p<0.05). In particular, 4 were replicated at the same SNP and with the same effect direction while the remaining 5 showed a significant association in a gene-based test. Finally, to look for genotype-phenotype relationship, the audiometric profiles of the three genotypes of the most strongly associated gene variants were analyzed. Seven out of the 9 replicated genes (CDH13, GRM8, ANK2, SLC16A6, ARSG, RIMBP2 and DCLK1) showed an audiometric pattern with differences between different genotypes further supporting their role in hearing function. These data demonstrate the usefulness of this multistep approach in providing new insights into the molecular basis of hearing and may suggest new targets for treatment and prevention of hearing impairment.


Subject(s)
DNA Replication , Gene Expression Profiling , Genome-Wide Association Study , Hearing/genetics , Adult , Animals , Genotype , Hair Cells, Auditory/metabolism , Humans , Mice , Phenotype , Polymorphism, Single Nucleotide , Stria Vascularis/metabolism
5.
Dis Model Mech ; 6(2): 434-42, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23065636

ABSTRACT

Mutations in the ATP6V0A4 gene lead to autosomal recessive distal renal tubular acidosis in patients, who often show sensorineural hearing impairment. A first Atp6v0a4 knockout mouse model that recapitulates the loss of H(+)-ATPase function seen in humans has been generated and recently reported (Norgett et al., 2012). Here, we present the first detailed analysis of the structure and function of the auditory system in Atp6v0a4(-/-) knockout mice. Measurements of the auditory brainstem response (ABR) showed significantly elevated thresholds in homozygous mutant mice, which indicate severe hearing impairment. Heterozygote thresholds were normal. Analysis of paint-filled inner ears and sections from E16.5 embryos revealed a marked expansion of cochlear and endolymphatic ducts in Atp6v0a4(-/-) mice. A regulatory link between Atp6v0a4, Foxi1 and Pds has been reported and we found that the endolymphatic sac of Atp6v0a4(-/-) mice expresses both Foxi1 and Pds, which suggests a downstream position of Atp6v0a4. These mutants also showed a lack of endocochlear potential, suggesting a functional defect of the stria vascularis on the lateral wall of the cochlear duct. However, the main K(+) channels involved in the generation of endocochlear potential, Kcnj10 and Kcnq1, are strongly expressed in Atp6v0a4(-/-) mice. Our results lead to a better understanding of the role of this proton pump in hearing function.


Subject(s)
Ear, Inner/enzymology , Ear, Inner/pathology , Endolymph/enzymology , Hearing Loss/enzymology , Hearing Loss/pathology , Protein Subunits/deficiency , Proton-Translocating ATPases/deficiency , Animals , Animals, Newborn , Anion Transport Proteins/metabolism , Ear, Inner/physiopathology , Endolymphatic Sac/pathology , Endolymphatic Sac/physiopathology , Epithelium/metabolism , Epithelium/pathology , Evoked Potentials, Auditory , Forkhead Transcription Factors/metabolism , Hair Cells, Auditory, Outer/metabolism , Hair Cells, Auditory, Outer/pathology , Hair Cells, Auditory, Outer/ultrastructure , Hearing Loss/physiopathology , Humans , KCNQ1 Potassium Channel/metabolism , Mice , Mice, Knockout , Mutation/genetics , Phenotype , Potassium Channels, Inwardly Rectifying/metabolism , Protein Subunits/metabolism , Proton-Translocating ATPases/metabolism , Stria Vascularis/metabolism , Stria Vascularis/pathology , Sulfate Transporters , Vacuolar Proton-Translocating ATPases
6.
PLoS One ; 7(11): e51065, 2012.
Article in English | MEDLINE | ID: mdl-23226461

ABSTRACT

Mutanlallemand (mtl) and Belly Spot and Deafness (bsd) are two new spontaneous alleles of the Lmx1a gene in mice. Homozygous mutants show head tossing and circling behaviour, indicative of vestibular defects, and they have short tails and white belly patches of variable size. The analysis of auditory brainstem responses (ABR) showed that mtl and bsd homozygotes are deaf, whereas heterozygous and wildtype littermates have normal hearing. Paint-filled inner ears at E16.5 revealed that mtl and bsd homozygotes lack endolymphatic ducts and semicircular canals and have short cochlear ducts. These new alleles show similarities with dreher (Lmx1a) mutants. Complementation tests between mtl and dreher and between mtl and bsd suggest that mtl and bsd are new mutant alleles of the Lmx1a gene. To determine the Lmx1a mutation in mtl and bsd mutant mice we performed PCR followed by sequencing of genomic DNA and cDNA. The mtl mutation is a single point mutation in the 3' splice site of exon 4 leading to an exon extension and the activation of a cryptic splice site 44 base pairs downstream, whereas the bsd mutation is a genomic deletion that includes exon 3. Both mutations lead to a truncated LMX1A protein affecting the homeodomain (mtl) or LIM2-domain (bsd), which is critical for LMX1A protein function. Moreover, the levels of Lmx1a transcript in mtl and bsd mutants are significantly down-regulated. Hmx2/3 and Pax2 expression are also down-regulated in mtl and bsd mutants, suggesting a role of Lmx1a upstream of these transcription factors in early inner ear morphogenesis. We have found that these mutants develop sensory patches although they are misshapen. The characterization of these two new Lmx1a alleles highlights the critical role of this gene in the development of the cochlea and vestibular system.


Subject(s)
Alleles , Cochlea/pathology , Deafness/genetics , LIM-Homeodomain Proteins/genetics , Point Mutation/genetics , Transcription Factors/genetics , Vestibule, Labyrinth/pathology , Animals , Base Sequence , Cochlea/abnormalities , Cochlea/embryology , Cochlea/physiopathology , Deafness/pathology , Deafness/physiopathology , Embryonic Stem Cells/metabolism , Exons/genetics , Fibroblast Growth Factor 9/genetics , Fibroblast Growth Factor 9/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Gene Transfer Techniques , Genetic Complementation Test , Hearing/physiology , Homozygote , LIM-Homeodomain Proteins/metabolism , Mice , Mice, Mutant Strains , Phenotype , RNA Splicing/genetics , Transcription Factors/metabolism , Vestibule, Labyrinth/abnormalities , Vestibule, Labyrinth/embryology , Vestibule, Labyrinth/physiopathology
7.
Proc Natl Acad Sci U S A ; 109(34): 13775-80, 2012 Aug 21.
Article in English | MEDLINE | ID: mdl-22872862

ABSTRACT

Autosomal recessive distal renal tubular acidosis (dRTA) is a severe disorder of acid-base homeostasis, often accompanied by sensorineural deafness. We and others have previously shown that mutations in the tissue-restricted a4 and B1 subunits of the H(+)-ATPase underlie this syndrome. Here, we describe an Atp6v0a4 knockout mouse, which lacks the a4 subunit. Using ß-galactosidase as a reporter for the null gene, developmental a4 expression was detected in developing bone, nose, eye, and skin, in addition to that expected in kidney and inner ear. By the time of weaning, Atp6v0a4(-/-) mice demonstrated severe metabolic acidosis, hypokalemia, and early nephrocalcinosis. Null mice were hypocitraturic, but hypercalciuria was absent. They were severely hearing-impaired, as shown by elevated auditory brainstem response thresholds and absent endocochlear potential. They died rapidly unless alkalinized. If they survived weaning with alkali supplementation, treatment could later be withdrawn, but -/- animals remained acidotic with alkaline urine. They also had an impaired sense of smell. Heterozygous animals were biochemically normal until acid-challenged, when they became more acidotic than +/+ animals. This mouse model recapitulates the loss of H(+)-ATPase function seen in human disease and can provide additional insights into dRTA and the physiology of the a4 subunit.


Subject(s)
Acidosis, Renal Tubular/genetics , Acidosis, Renal Tubular/physiopathology , Hearing Loss/genetics , Proton-Translocating ATPases/genetics , Proton-Translocating ATPases/physiology , Animals , Disease Models, Animal , Ear, Inner/physiopathology , Female , Gene Expression Regulation, Developmental , Genotype , Heterozygote , Hydrogen-Ion Concentration , Male , Mice , Mice, Knockout , Nephrocalcinosis/genetics , Phenotype , Proton Pumps , Vacuolar Proton-Translocating ATPases
8.
Dev Biol ; 361(1): 12-26, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22019302

ABSTRACT

The interpeduncular nucleus (IP) is a key limbic structure, highly conserved evolutionarily among vertebrates. The IP receives indirect input from limbic areas of the telencephalon, relayed by the habenula via the fasciculus retroflexus. The function of the habenulo-IP complex is poorly understood, although there is evidence that in rodents it modulates behaviors such as learning and memory, avoidance, reward and affective states. The IP has been an important subject of interest for neuroscientists, and there are multiple studies about the adult structure, chemoarchitecture and its connectivity, with complex results, due to the presence of multiple cell types across a variety of subnuclei. However, the ontogenetic origins of these populations have not been examined, and there is some controversy about its location in the midbrain-anterior hindbrain area. To address these issues, we first investigated the anteroposterior (AP) origin of the IP complex by fate-mapping its neuromeric origin in the chick, discovering that the IP develops strictly within isthmus and rhombomere 1. Next, we studied the dorsoventral (DV) positional identity of subpopulations of the IP complex. Our results indicate that there are at least four IP progenitor domains along the DV axis. These specific domains give rise to distinct subtypes of cell populations that target the IP with variable subnuclear specificity. Interestingly, these populations can be characterized by differential expression of the transcription factors Pax7, Nkx6.1, Otp, and Otx2. Each of these subpopulations follows a specific route of migration from its source, and all reach the IP roughly at the same stage. Remarkably, IP progenitor domains were found both in the alar and basal plates. Some IP populations showed rostrocaudal restriction in their origins (isthmus versus anterior or posterior r1 regions). A tentative developmental model of the structure of the avian IP is proposed. The IP emerges as a plurisegmental and developmentally heterogeneous formation that forms ventromedially within the isthmus and r1. These findings are relevant since they help to understand the highly complex chemoarchitecture, hodology and functions of this important brainstem structure.


Subject(s)
Cell Lineage/physiology , Cell Movement/physiology , Gene Expression Regulation, Developmental/physiology , Limbic System/cytology , Limbic System/embryology , Models, Biological , Animals , Chick Embryo , Gene Expression Regulation, Developmental/genetics , Immunohistochemistry , In Situ Hybridization , PAX7 Transcription Factor/metabolism
9.
J Comp Neurol ; 496(6): 802-18, 2006 Jun 20.
Article in English | MEDLINE | ID: mdl-16628617

ABSTRACT

We investigated in the mouse and chick the neuroepithelial origin and development of the locus coeruleus (LoC), the most important noradrenergic neuronal population in the brain. We first studied the topography of the developing LoC in the hindbrain, using as markers the key noradrenergic marker gene Dbh and the transcription factors Phox2a and Phox2b (upstream of Dbh). In both mouse and chicken, LoC neurons first appear arranged linearly along the middle one-third of the alar plate of rhombomere 1 (r1), collinear to a reference ventricular longitudinal band that early on expresses Phox2a and Phox2b in the alar plate of r2 and later expands to r1. Double-labeling experiments with LoC markers (Dbh or Phox2a) and either alar (Pax7 and Rnx3) or basal (Otp) genetic markers suggested that LoC cells migrate from their origin in the alar plate to a final position in the lateral basal plate. To corroborate these suggestions experimentally and determine the precise origin of the LoC, we fate mapped the LoC in the chick at stage HH11 by using quail-chick homotopic grafts. The experimental results confirmed that the LoC originates in the alar plate throughout the rostrocaudal extent of r1 and ruled out a rostrocaudal translocation. They also corroborated a ventralward tangential migration of LoC cells into the lateral basal plate, where the postmigratory LoC primordium is located. Comparisons with neighboring alar r1-derived cell populations established that LoC neurons originate outside the cerebellum, in a matrix area intercalated dorsoventrally between the sources of the prospective vestibular and trigeminal columns.


Subject(s)
Locus Coeruleus/cytology , Neurons/physiology , Animals , Biomarkers/metabolism , Cell Movement , Chick Embryo , Dopamine beta-Hydroxylase/metabolism , Embryonic Development , Homeodomain Proteins/metabolism , Immunohistochemistry , Locus Coeruleus/embryology , Mice , Nerve Tissue Proteins/metabolism , Neuroepithelial Cells/physiology , PAX7 Transcription Factor/metabolism , Quail , Rhombencephalon/embryology , Rhombencephalon/metabolism , Rhombencephalon/transplantation , Species Specificity , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...