Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Audiol Neurootol ; 28(5): 327-337, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37121227

RESUMEN

BACKGROUND: Low-frequency non-syndromic hearing loss (LFNSHL) is a rare form of hearing loss (HL). It is defined as HL at low frequencies (≤2,000 Hz) resulting in a characteristic ascending audiogram. LFNSHL is usually diagnosed postlingually and is progressive, leading to HL affecting other frequencies as well. Sometimes it occurs with tinnitus. Around half of the diagnosed prelingual HL cases have a genetic cause and it is usually inherited in an autosomal recessive mode. Postlingual HL caused by genetic changes generally has an autosomal dominant pattern of inheritance and its incidence remains unknown. SUMMARY: To date, only a handful of genes have been found as causing LFNSHL: well-established WFS1 and, reported in some cases, DIAPH1, MYO7A, TNC, and CCDC50 (respectively, responsible for DFNA6/14/38, DFNA1, DFNA11, DFNA56, and DFNA44). In this review, we set out audiological phenotypes, causative genetic changes, and molecular mechanisms leading to the development of LFNSHL. KEY MESSAGES: LFNSHL is most commonly caused by pathogenic variants in the WFS1 gene, but it is also important to consider changes in other HL genes, which may result in similar audiological phenotype.


Asunto(s)
Sordera , Pérdida Auditiva Sensorineural , Pérdida Auditiva , Humanos , Linaje , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva/genética , Forminas/genética
2.
Eur Arch Otorhinolaryngol ; 280(10): 4361-4369, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37004521

RESUMEN

PURPOSE: If before cochlear implantation it was possible to assay biomarkers of neuroplasticity, we might be able to identify those children with congenital deafness who, later on, were at risk of poor speech and language rehabilitation outcomes. METHODS: A group of 40 children aged up to 2 years with DFNB1-related congenital deafness was observed in this prospective cohort study over three follow-up intervals (0, 8, and 18 months) after cochlear implant (CI) activation. Children were assessed for auditory development using the LittlEARS Questionnaire (LEAQ) score, and at the same time, measurements were made of matrix metalloproteinase-9 (MMP-9) plasma levels. RESULTS: There were significant negative correlations between plasma levels of MMP-9 at 8-month follow-up and LEAQ score at cochlear implantation (p = 0.04) and LEAQ score at 18-month follow-up (p = 0.02) and between MMP-9 plasma levels at 18-month follow-up and LEAQ score at cochlear implantation (p = 0.04). As already reported, we confirmed a significant negative correlation between MMP-9 plasma level at cochlear implantation and LEAQ score at 18-month follow-up (p = 0.005). Based on this latter correlation, two clusters of good and poor CI performers could be isolated. CONCLUSIONS: The study shows that children born deaf who have an MMP-9 plasma level of less than 150 ng/ml at cochlear implantation have a good chance of attaining a high LEAQ score after 18 months of speech and language rehabilitation. This indicates that MMP-9 plasma level at cochlear implantation is a good prognostic marker for CI outcome.


Asunto(s)
Implantación Coclear , Sordera , Niño , Humanos , Metaloproteinasa 9 de la Matriz , Estudios de Cohortes , Estudios Prospectivos , Sordera/cirugía , Sordera/rehabilitación , Biomarcadores
3.
Int J Mol Sci ; 24(4)2023 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-36835126

RESUMEN

Congenitally deaf children who undergo cochlear implantation before 1 year of age develop their auditory skills faster than children who are implanted later. In this longitudinal study, a cohort of 59 implanted children were divided into two subgroups according to their ages at implantation-below or above 1 year old-and the plasma levels of matrix metalloproteinase-9 (MMP-9), brain-derived neurotrophic factor (BDNF), and pro-BDNF were measured at 0, 8, and 18 months after cochlear implant activation, while auditory development was simultaneously evaluated using the LittlEARs Questionnaire (LEAQ). A control group consisted of 49 age-matched healthy children. We identified statistically higher BDNF levels at 0 months and at the 18-month follow-ups in the younger subgroup compared to the older one and lower LEAQ scores at 0 months in the younger subgroup. Between the subgroups, there were significant differences in the changes in BDNF levels from 0 to 8 months and in LEAQ scores from 0 to 18 months. The MMP-9 levels significantly decreased from 0 to 18 months and from 0 to 8 months in both subgroups and from 8 to 18 months only in the older one. For all measured protein concentrations, significant differences were identified between the older study subgroup and the age-matched control group.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Implantación Coclear , Sordera , Metaloproteinasa 9 de la Matriz , Niño , Humanos , Lactante , Factor Neurotrófico Derivado del Encéfalo/sangre , Factor Neurotrófico Derivado del Encéfalo/química , Sordera/terapia , Estudios Longitudinales , Metaloproteinasa 9 de la Matriz/sangre , Metaloproteinasa 9 de la Matriz/química
4.
Int J Mol Sci ; 24(24)2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38139324

RESUMEN

Leber hereditary optic neuropathy (LHON) is a rare disorder causing a sudden painless loss of visual acuity in one or both eyes, affecting young males in their second to third decade of life. The molecular background of the LHON is up to 90%, genetically defined by a point mutation in mitochondrial DNA. Recently, an autosomal recessive form of LHON (LHONAR1, arLHON) has been discovered, caused by biallelic variants in the DNAJC30 gene. This study provides the results of the DNAJC30 gene analysis in a large group of 46 Polish patients diagnosed with LHON, together with the clinical characterization of the disease. The c.152A>G (p.Tyr51Cys) substitution in the DNAJC30 gene was detected in all the patients as homozygote or compound heterozygote. Moreover, we identified one novel variant, c.293A>G, p.(Tyr98Cys), as well as two ultra-rare DNAJC30 variants: c.293A>C, p.(Tyr98Ser), identified to date only in one individual affected with LHONAR1, and c.130_131delTC (p.Ser44ValfsTer8), previously described only in two patients with Leigh syndrome. The patients presented here represent the largest group of subjects with DNAJC30 gene mutations described to date. Based on our data, the autosomal recessive form of LHON caused by DNAJC30 gene mutations is more frequent than the mitochondrial form in Polish patients. The results of our study suggest that Sanger sequencing of the single-exon DNAJC30 gene should be a method of choice applied to identify a molecular background of clinically confirmed LHON in Polish patients. This approach will help to reduce the costs of molecular testing.


Asunto(s)
Proteínas del Choque Térmico HSP40 , Atrofia Óptica Hereditaria de Leber , Humanos , Masculino , ADN Mitocondrial/genética , Mitocondrias/genética , Mutación , Atrofia Óptica Hereditaria de Leber/genética , Polonia , Enfermedades Raras/genética , Proteínas del Choque Térmico HSP40/genética
5.
Hum Genet ; 141(3-4): 445-453, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35254497

RESUMEN

Novel hearing loss (HL) genes are constantly being discovered, and evidence from independent studies is essential to strengthen their position as causes of hereditary HL. To address this issue, we searched our genetic data of families with autosomal dominant HL (ADHL) who had been tested with high-throughput DNA sequencing methods. For CD164, only one pathogenic variant in one family has so far been reported. For LMX1A, just two previous studies have revealed its involvement in ADHL. In this study we found two families with the same pathogenic variant in CD164 and one family with a novel variant in LMX1A (c.686C>A; p.(Ala229Asp)) that impairs its transcriptional activity. Our data show recurrence of the same CD164 variant in two HL families of different geographic origin, which strongly suggests it is a mutational hotspot. We also provide further evidence for haploinsufficiency as the pathogenic mechanism underlying LMX1A-related ADHL.


Asunto(s)
Sordera , Endolina , Pérdida Auditiva Sensorineural , Pérdida Auditiva , Proteínas con Homeodominio LIM , Factores de Transcripción , Humanos , Sordera/genética , Endolina/genética , Genes Dominantes , Pérdida Auditiva/genética , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/patología , Proteínas con Homeodominio LIM/genética , Mutación , Linaje , Factores de Transcripción/genética
6.
Int J Mol Sci ; 23(11)2022 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-35682719

RESUMEN

Hearing is an important human sense for communicating and connecting with others. Partial deafness (PD) is a common hearing problem, in which there is a down-sloping audiogram. In this study, we apply a practical system for classifying PD patients, used for treatment purposes, to distinguish two groups of patients: one with almost normal hearing thresholds at low frequencies (PDT-EC, n = 20), and a second group with poorer thresholds at those same low frequencies (PDT-EAS, n = 20). After performing comprehensive genetic testing with a panel of 237 genes, we found that genetic factors can explain a significant proportion of both PDT-EC and PDT-EAS hearing losses, accounting, respectively, for approx. one-fifth and one-half of all the cases in our cohort. Most of the causative variants were located in dominant and recessive genes previously linked to PD, but more than half of the variants were novel. Among the contributors to PDT-EC we identified OSBPL2 and SYNE4, two relatively new hereditary hearing loss genes with a low publication profile. Our study revealed that, for all PD patients, a postlingual hearing loss more severe in the low-frequency range is associated with a higher detection rate of causative variants. Isolating a genetic cause of PD is important in terms of prognosis, therapeutic effectiveness, and risk of recurrence.


Asunto(s)
Implantación Coclear , Sordera , Pérdida Auditiva Sensorineural , Pérdida Auditiva , Receptores de Esteroides , Sordera/genética , Genes Recesivos , Pérdida Auditiva/genética , Pérdida Auditiva Sensorineural/genética , Humanos , Receptores de Esteroides/genética
7.
Int J Mol Sci ; 23(21)2022 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-36362125

RESUMEN

Non-canonical splice site variants are increasingly recognized as a relevant cause of the USH2A-associated diseases, non-syndromic autosomal recessive retinitis pigmentosa and Usher syndrome type 2. Many non-canonical splice site variants have been reported in public databases, but an effect on pre-mRNA splicing has only been functionally verified for a subset of these variants. In this study, we aimed to extend the knowledge regarding splicing events by assessing a selected set of USH2A non-canonical splice site variants and to study their potential pathogenicity. Eleven non-canonical splice site variants were selected based on four splice prediction tools. Ten different USH2A constructs were generated and minigene splice assays were performed in HEK293T cells. An effect on pre-mRNA splicing was observed for all 11 variants. Various events, such as exon skipping, dual exon skipping and partial exon skipping were observed and eight of the tested variants had a full effect on splicing as no conventionally spliced mRNA was detected. We demonstrated that non-canonical splice site variants in USH2A are an important contributor to the genetic etiology of the associated disorders. This type of variant generally should not be neglected in genetic screening, both in USH2A-associated disease as well as other hereditary disorders. In addition, cases with these specific variants may now receive a conclusive genetic diagnosis.


Asunto(s)
Síndromes de Usher , Humanos , Síndromes de Usher/genética , Células HEK293 , Precursores del ARN , Proteínas de la Matriz Extracelular/genética , Mutación , Sitios de Empalme de ARN/genética
8.
Audiol Neurootol ; 26(4): 226-235, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33352548

RESUMEN

BACKGROUND: Genetically determined prelingual hearing loss (HL) may occur in an isolated or syndromic form. OBJECTIVE: The aim of the study was to unravel the genetic cause of medical problems in a 21-year-old woman, whose phenotypic presentation extended beyond Stickler syndrome and included enlarged vestibular aqueduct (EVA) and persistent microhematuria. METHODS AND RESULTS: After sequencing of clinical exome, a known de novo COL2A1 pathogenic variant (c.1833+1G>A, p.?) causative for Stickler syndrome and one paternally inherited pathogenic change in COL4A5 (c.1871G>A, p.Gly624Asp) causative for X-linked Alport syndrome were found. No pathogenic variants, including those within the SLC26A4 5' region (Caucasian EVA haplotype), explaining the development of EVA, were identified. CONCLUSIONS: The study reveals a multilocus genomic variation in one individual and provides a molecular diagnosis of two HL syndromes that co-occur in the proband independent of each other. For the third entity, EVA, no etiological factor was identified. Our data emphasize the relevance of detailed clinical phenotyping for accurate genotype interpretation. Focus on broadening the phenotypic spectrum of known genetic syndromes may actually obscure patients with multiple molecular diagnoses.


Asunto(s)
Pérdida Auditiva Sensorineural , Pérdida Auditiva , Acueducto Vestibular , Adulto , Femenino , Pruebas Genéticas , Pérdida Auditiva/diagnóstico , Pérdida Auditiva/genética , Pérdida Auditiva Sensorineural/diagnóstico , Pérdida Auditiva Sensorineural/genética , Humanos , Proteínas de Transporte de Membrana/genética , Mutación , Transportadores de Sulfato , Síndrome , Adulto Joven
9.
Int J Mol Sci ; 22(12)2021 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-34203967

RESUMEN

A substantial proportion of subjects with autosomal recessive retinitis pigmentosa (arRP) or Usher syndrome type II (USH2) lacks a genetic diagnosis due to incomplete USH2A screening in the early days of genetic testing. These cases lack eligibility for optimal genetic counseling and future therapy. USH2A defects are the most frequent cause of USH2 and are also causative in individuals with arRP. Therefore, USH2A is an important target for genetic screening. The aim of this study was to assess unscreened or incompletely screened and unexplained USH2 and arRP cases for (likely) pathogenic USH2A variants. Molecular inversion probe (MIP)-based sequencing was performed for the USH2A exons and their flanking regions, as well as published deep-intronic variants. This was done to identify single nucleotide variants (SNVs) and copy number variants (CNVs) in 29 unscreened or partially pre-screened USH2 and 11 partially pre-screened arRP subjects. In 29 out of these 40 cases, two (likely) pathogenic variants were successfully identified. Four of the identified SNVs and one CNV were novel. One previously identified synonymous variant was demonstrated to affect pre-mRNA splicing. In conclusion, genetic diagnoses were obtained for a majority of cases, which confirms that MIP-based sequencing is an effective screening tool for USH2A. Seven unexplained cases were selected for future analysis with whole genome sequencing.


Asunto(s)
Análisis Costo-Beneficio , Exones/genética , Proteínas de la Matriz Extracelular/genética , Sondas Moleculares/metabolismo , Sitios de Empalme de ARN/genética , Retinitis Pigmentosa/genética , Análisis de Secuencia de ADN , Síndromes de Usher/genética , Secuencia de Bases , Variaciones en el Número de Copia de ADN/genética , Eliminación de Gen , Humanos , Polimorfismo de Nucleótido Simple/genética , Retinitis Pigmentosa/economía , Síndromes de Usher/economía
10.
Genet Med ; 22(7): 1235-1246, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32307445

RESUMEN

PURPOSE: Missing heritability in human diseases represents a major challenge, and this is particularly true for ABCA4-associated Stargardt disease (STGD1). We aimed to elucidate the genomic and transcriptomic variation in 1054 unsolved STGD and STGD-like probands. METHODS: Sequencing of the complete 128-kb ABCA4 gene was performed using single-molecule molecular inversion probes (smMIPs), based on a semiautomated and cost-effective method. Structural variants (SVs) were identified using relative read coverage analyses and putative splice defects were studied using in vitro assays. RESULTS: In 448 biallelic probands 14 known and 13 novel deep-intronic variants were found, resulting in pseudoexon (PE) insertions or exon elongations in 105 alleles. Intriguingly, intron 13 variants c.1938-621G>A and c.1938-514G>A resulted in dual PE insertions consisting of the same upstream, but different downstream PEs. The intron 44 variant c.6148-84A>T resulted in two PE insertions and flanking exon deletions. Eleven distinct large deletions were found, two of which contained small inverted segments. Uniparental isodisomy of chromosome 1 was identified in one proband. CONCLUSION: Deep sequencing of ABCA4 and midigene-based splice assays allowed the identification of SVs and causal deep-intronic variants in 25% of biallelic STGD1 cases, which represents a model study that can be applied to other inherited diseases.


Asunto(s)
Degeneración Macular , Transcriptoma , Transportadoras de Casetes de Unión a ATP/genética , Genómica , Humanos , Intrones , Degeneración Macular/genética , Mutación , Linaje , Enfermedad de Stargardt
11.
BMC Nephrol ; 21(1): 380, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32873246

RESUMEN

BACKGROUND: Despite its established association with chronic kidney disease (CKD) the role of myosin-9 (MYH9) gene variation on transplanted kidney function remains unknown. This study aimed at evaluating the effect of donor MYH9 nephrogenic variants on renal allograft function within the first post transplantation year. METHODS: In the longitudinal kidney transplant study 207 deceased donors were genotyped for previously known risk MYH9 single nucleotide polymorphisms (SNPs). The predictor was MYH9 high-risk variants status. The primary outcome was mean eGFR found in low vs. high risk MYH9 genotypes between third and twelfth post-transplant month, the secondary outcome was the risk of proteinuria. RESULTS: Distribution of genotypes remained in Hardy-Weinberg equilibrium. The T allele of rs3752462 (dominant model, TT or TC vs. CC) was associated with higher filtration rate (P = 0.05) in a multivariate analysis after adjusting for delayed graft function and donor sex. Two G alleles of rs136211 (recessive model, GG vs. GA or AA) resulted in doubling the risk of proteinuria (OR = 2.22; 95% CI = 1.18-4.37, P = 0.017) after adjusting for donor and recipient sex. CONCLUSION: Deceased donor kidneys of European descent harboring MYH9 SNPs rs3752462 T allele show significantly superior estimated filtration rate while those of rs136211 GG genotype excessive risk of proteinuria. These findings, if replicated, may further inform and improve individualization of allocation and treatment policies.


Asunto(s)
Tasa de Filtración Glomerular , Fallo Renal Crónico/cirugía , Trasplante de Riñón , Cadenas Pesadas de Miosina/genética , Complicaciones Posoperatorias/genética , Proteinuria/genética , Insuficiencia Renal/genética , Adolescente , Adulto , Anciano , Cadáver , Femenino , Genotipo , Humanos , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Complicaciones Posoperatorias/epidemiología , Proteinuria/epidemiología , Insuficiencia Renal/epidemiología , Donantes de Tejidos , Adulto Joven
12.
Metab Brain Dis ; 35(8): 1317-1327, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32740724

RESUMEN

Leber hereditary optic neuropathy (LHON) is a mitochondrial disorder with symptoms limited to a single tissue, optic nerve, resulting in vision loss. In the majority of cases it is caused by one of three point mutations in mitochondrial DNA (mtDNA) but their presence is not sufficient for disease development, since ~50% of men and ~10% women who carry them are affected. Thus additional modifying factors must exist. In this study, we use next generation sequencing to investigate the role of whole mtDNA variation in male Polish patients with LHON and m.11778G > A, the most frequent LHON mutation. We present a possible association between mtDNA haplogroup K and variants in its background, a combination of m.3480A > G, m.9055G > A, m.11299 T > C and m.14167C > T, and LHON mutation. These variants may have a negative effect on m.11778G > A increasing its penetrance and the risk of LHON in the Polish population. Surprisingly, we did not observe associations previously reported for m.11778G > A and LHON in European populations, particularly for haplogroup J as a risk factor, implying that mtDNA variation is much more complex. Our results indicate possible contribution of novel combination of mtDNA genetic factors to the LHON phenotype.


Asunto(s)
ADN Mitocondrial/genética , Variación Genética/genética , Genoma Mitocondrial/genética , Mutación/genética , Atrofia Óptica Hereditaria de Leber/epidemiología , Atrofia Óptica Hereditaria de Leber/genética , Adulto , Anciano , Anciano de 80 o más Años , Humanos , Masculino , Persona de Mediana Edad , Atrofia Óptica Hereditaria de Leber/diagnóstico , Polonia/epidemiología , Adulto Joven
13.
Int J Mol Sci ; 21(10)2020 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-32413971

RESUMEN

ABCA4 gene mutations are the cause of a spectrum of ABCA4 retinopathies, and the most common juvenile macular degeneration is called Stargardt disease. ABCA4 has previously been observed almost exclusively in the retina. Therefore, studying the functional consequences of ABCA4 variants has required advanced molecular analysis techniques. The aim of the present study was to evaluate whether human hair follicles may be used for molecular analysis of the ABCA4 gene splice-site variants in patients with ABCA4 retinopathies. We assessed ABCA4 expression in hair follicles and skin at mRNA and protein levels by means of real-time PCR and Western blot analyses, respectively. We performed cDNA sequencing to reveal the presence of full-length ABCA4 transcripts and analyzed ABCA4 transcripts from three patients with Stargardt disease carrying different splice-site ABCA4 variants: c.5312+1G>A, c.5312+2T>G and c.5836-3C>A. cDNA analysis revealed that c.5312+1G>A, c.5312+2T>G variants led to the skipping of exon 37, and the c.5836-3C>A variant resulted in the insertion of 30 nucleotides into the transcript. Our results strongly argue for the use of hair follicles as a model for the molecular analysis of the pathogenicity of ABCA4 variants in patients with ABCA4 retinopathies.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Folículo Piloso/metabolismo , Enfermedades de la Retina/genética , Enfermedad de Stargardt/genética , Análisis Mutacional de ADN , Exones/genética , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica/genética , Folículo Piloso/patología , Humanos , Queratinocitos/metabolismo , Queratinocitos/patología , Degeneración Macular/genética , Degeneración Macular/patología , Masculino , Melanocitos/metabolismo , Melanocitos/patología , Mutación/genética , Linaje , Cultivo Primario de Células , Sitios de Empalme de ARN/genética , Retina/metabolismo , Retina/patología , Enfermedades de la Retina/patología , Enfermedad de Stargardt/patología
14.
J Transl Med ; 17(1): 269, 2019 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-31412945

RESUMEN

BACKGROUND: Hearing loss (HL) is the most common disability of human senses characterized by a great allelic heterogeneity. GJB2 and TMPRSS3 are two well-known HL genes typically underlying its monogenic form. Recently, TMPRSS3/GJB2 digenic inheritance has been proposed. As results of genetic testing can be easily overinterpreted, we aimed to verify the hypothesis. METHODS: From genetic database of HL patients with at least one TMPRSS3 pathogenic variants we have selected individuals with additional GJB2 pathogenic variants. All of the available family members were recruited for the study. Segregation analysis of the respective TMPRSS3 and GJB2 pathogenic variants was performed within the families. RESULTS: The strategy has allowed to identify four individuals who were double heterozygous for known pathogenic TMPRSS3 and GJB2 variants. Two individuals from different families had GJB2 c.35delG and TMPRSS3 c.208delC and in two other individuals from one family GJB2 c.35delG together with TMPRSS3 c.1343T>C variants were found. None of these subjects has ever reported hearing problems and their hearing status was normal. CONCLUSIONS: Our data provide evidence against TMPRSS3/GJB2 digenic inheritance of HL. As high throughput sequencing is increasingly used for genetic testing, particular caution should be taken to provide the patients with accurate genetic counseling.


Asunto(s)
Conexina 26/genética , Genética Médica , Pérdida Auditiva/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Proteínas de la Membrana/genética , Herencia Multifactorial/genética , Proteínas de Neoplasias/genética , Serina Endopeptidasas/genética , Conexina 26/metabolismo , Regulación de la Expresión Génica , Audición/genética , Pérdida Auditiva/fisiopatología , Humanos , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Serina Endopeptidasas/metabolismo
15.
J Transl Med ; 17(1): 351, 2019 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-31655630

RESUMEN

BACKGROUND: Biallelic PTPRQ pathogenic variants have been previously reported as causative for autosomal recessive non-syndromic hearing loss. In 2018 the first heterozygous PTPRQ variant has been implicated in the development of autosomal dominant non-syndromic hearing loss (ADNSHL) in a German family. The study presented the only, so far known, PTPRQ pathogenic variant (c.6881G>A) in ADNSHL. It is located in the last PTPRQ coding exon and introduces a premature stop codon (p.Trp2294*). METHODS: A five-generation Polish family with ADNSHL was recruited for the study (n = 14). Thorough audiological, neurotological and imaging studies were carried out to precisely define the phenotype. Genomic DNA was isolated from peripheral blood samples or buccal swabs of available family members. Clinical exome sequencing was conducted for the proband. Family segregation analysis of the identified variants was performed using Sanger sequencing. Single nucleotide polymorphism array on DNA samples from the Polish and the original German family was used for genome-wide linkage analysis. RESULTS: Combining clinical exome sequencing and family segregation analysis, we have identified the same (NM_001145026.2:c.6881G>A, NP_001138498.1:p.Trp2294*) PTPRQ alteration in the Polish ADNSHL family. Using genome-wide linkage analysis, we found that the studied family and the original German family derive from a common ancestor. Deep phenotyping of the affected individuals showed that in contrast to the recessive form, the PTPRQ-related ADNSHL is not associated with vestibular dysfunction. In both families ADNSHL was progressive, affected mainly high frequencies and had a variable age of onset. CONCLUSION: Our data provide the first confirmation of PTPRQ involvement in ADNSHL. The finding strongly reinforces the inclusion of PTPRQ to the small set of genes leading to both autosomal recessive and dominant hearing loss.


Asunto(s)
Pérdida Auditiva Sensorineural/genética , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética , Adolescente , Adulto , Edad de Inicio , Niño , Femenino , Genes Dominantes , Pérdida Auditiva Sensorineural/fisiopatología , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/fisiología , Mutación , Linaje , Terminación de la Cadena Péptídica Traduccional/genética , Fenotipo , Polonia , Polimorfismo de Nucleótido Simple , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/química , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/fisiología , Investigación Biomédica Traslacional , Adulto Joven
16.
Exp Dermatol ; 28(2): 107-112, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30548893

RESUMEN

Keratinocyte culture is a necessary and widely used tool in a variety of experimental dermatological and biomedical studies. Literature search has revealed a variety of different protocols of human primary keratinocyte isolation and culture. Therefore, the aim of this paper was to review and summarize current trends in human primary keratinocyte culture techniques. We present data on the most popular and effective methods of human keratinocyte isolation and cultivation obtained from screening of 945 papers published during the last 10 years.


Asunto(s)
Técnicas de Cultivo de Célula , Queratinocitos/citología , Células 3T3 , Animales , Proliferación Celular , Separación Celular/métodos , Células Cultivadas , Técnicas de Cocultivo , Colágeno/química , Medios de Cultivo/química , Medio de Cultivo Libre de Suero , Dermatología/métodos , Humanos , Ratones , Piel/patología , Tripsina/farmacología
17.
Graefes Arch Clin Exp Ophthalmol ; 256(11): 2127-2134, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30084067

RESUMEN

PURPOSE: Schnyder corneal dystrophy (SCD) is a rare inherited disease that leads to gradual vision loss by the deposition of lipids in the corneal stroma. The aim of this study is to report a novel pathogenic variant in the UBIAD1 gene and present clinical and molecular findings in Polish patients with SCD. METHODS: Individuals (n = 37) originating from four Polish SCD families were subjected for a complete ophthalmological check-up and genetic testing. Corneal changes were visualized by slit-lamp examination, anterior segment optical coherent tomography (AS-OCT), and in vivo confocal microscopy (IVCM). RESULTS: In a proband with primarily mild SCD that progressed rapidly at the end of the fifth decade of life, a novel missense pathogenic variant in UBIAD1 (p.Thr120Arg) was identified. The other studied SCD family represents the second family reported worldwide with the UBIAD1 p.Asp112Asn variant. SCD in the remaining two families resulted from a frequently identified p.Asn102Ser pathogenic variant. All affected subjects presented a crystalline form of SCD. The severity of corneal changes was age-dependent, and their morphology and localization are described in detail. CONCLUSION: The novel p.Thr120Arg is the fourth SCD-causing variant lying within the FARM motif of the UBIAD1 protein, which underlines a high importance of this motif for SCD pathogenesis. The current study provides independent evidence for the pathogenic potential of UBIAD1 p.Asp112Asn and new information useful for clinicians.


Asunto(s)
Distrofias Hereditarias de la Córnea/genética , Dimetilaliltranstransferasa/genética , Mutación Missense , Polimorfismo de Nucleótido Simple , Adolescente , Adulto , Anciano , Distrofias Hereditarias de la Córnea/diagnóstico , Femenino , Humanos , Masculino , Microscopía Confocal , Persona de Mediana Edad , Linaje , Reacción en Cadena de la Polimerasa , Tomografía de Coherencia Óptica , Agudeza Visual/fisiología , Adulto Joven
18.
Eur Arch Otorhinolaryngol ; 275(8): 1979-1985, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29936625

RESUMEN

PURPOSE: Tinnitus described as individual perception of phantom sound constitutes a significant medical problem and has become an essential subject of many studies conducted worldwide. In the study, we aimed to examine the prevalence of tinnitus among Polish hearing loss (HL) patients with identified mitochondrial DNA (mtDNA) variants. METHODS: Among the selected group of unrelated HL patients with known mtDNA pathogenic variants, two questionnaires were conducted, i.e. Tinnitus Handicap Inventory translated into Polish (THI-POL) and Visual Analogue Scale (VAS) for measuring subjectively perceived tinnitus loudness, distress, annoyance and possibility of coping with this condition (VASs). Pathogenic mtDNA variants were detected with real-time PCR and sequencing of the whole mtDNA. RESULTS: This is the first extensive tinnitus characterization using THI-POL and VASs questionnaires in HL patients due to mtDNA variants. We have established the prevalence of tinnitus among the studied group at 23.5%. We found that there are no statistically significant differences in the prevalence of tinnitus and its characteristic features between HL patients with known HL mtDNA variants and the general Polish population. In Polish HL patients with tinnitus, m.7511T>C was significantly more frequent than in patients without tinnitus. We observed that the prevalence of tinnitus is lower in Polish patients with m.1555A>G as compared to other available data. CONCLUSIONS: Our data suggest that the mtDNA variants causative of HL may affect tinnitus development but this effect seems to be ethnic-specific.


Asunto(s)
ADN Mitocondrial/genética , Pérdida Auditiva/genética , Acúfeno/epidemiología , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Polonia , Prevalencia , Encuestas y Cuestionarios , Escala Visual Analógica , Adulto Joven
19.
J Transl Med ; 15(1): 25, 2017 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-28178980

RESUMEN

BACKGROUND: Hearing loss and ovarian dysfunction are key features of Perrault syndrome (PRLTS) but the clinical and pathophysiological features of hearing impairment in PRLTS individuals have not been addressed. Mutations in one of five different genes HSD17B4, HARS2, LARS2, CLPP or TWNK (previous symbol C10orf2) cause the autosomal recessive disorder but they are found only in about half of the patients. METHODS: We report on two siblings with a clinical picture resembling a severe, neurological type of PRLTS. For an exhaustive characterisation of the phenotype neuroimaging with volumetric measurements and objective measures of cochlear hair cell and auditory nerve function (otoacustic emissions and auditory brainstem responses) were used. Whole exome sequencing was applied to identify the genetic cause of the disorder. Co-segregation of the detected mutations with the phenotype was confirmed by Sanger sequencing. In silico analysis including 3D protein structure modelling was used to predict the deleterious effects of the detected variants on protein function. RESULTS: We found two rare biallelic mutations in TWNK, encoding Twinkle, an essential mitochondrial helicase. Mutation c.1196A>G (p.Asn399Ser) recurred for the first time in a patient with PRLTS and the second mutation c.1802G>A (p.Arg601Gln) was novel for the disorder. In both patients neuroimaging studies showed diminished cervical enlargement of the spinal cord and for the first time in PRLTS partial atrophy of the vestibulocochlear nerves and decreased grey and increased white matter volumes of the cerebellum. Morphological changes in the auditory nerves, their desynchronized activity and partial cochlear dysfunction underlay the complex mechanism of hearing impairment in the patients. CONCLUSIONS: Our study unveils novel features on the phenotypic landscape of PRLTS and provides further evidence that the newly identified for PRLTS TWNK gene is involved in its pathogenesis.


Asunto(s)
Audiometría de Tonos Puros , ADN Helicasas/genética , Disgenesia Gonadal 46 XX/genética , Pérdida Auditiva Sensorineural/genética , Proteínas Mitocondriales/genética , Sistema Nervioso/patología , Adulto , Secuencia de Aminoácidos , Niño , Preescolar , ADN Helicasas/química , Demografía , Femenino , Heterocigoto , Humanos , Imagen por Resonancia Magnética , Masculino , Proteínas Mitocondriales/química , Mutación/genética , Linaje , Alineación de Secuencia , Adulto Joven
20.
BMC Med Genet ; 18(1): 142, 2017 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-29197352

RESUMEN

BACKGROUND: Implementation of whole exome sequencing has provided unique opportunity for a wide screening of causative variants in genetically heterogeneous diseases, including nonsyndromic hearing impairment. TRIOBP in the inner ear is responsible for proper structure and function of stereocilia and is necessary for sound transduction. METHODS: Whole exome sequencing followed by Sanger sequencing was conducted on patients derived from Polish hearing loss family. RESULTS: Based on whole exome analysis, we identified two TRIOBP pathogenic variants (c.802_805delCAGG, p.Gln268Leufs*610 and c.5014G>T, p.Gly1672*, the first of which was novel) causative of nonsyndromic, peri- to postlingual, moderate-to-severe hearing loss in three siblings from a Polish family. Typically, TRIOBP pathogenic variants lead to prelingual, severe-to-profound hearing loss, thus the onset and degree of hearing impairment in our patients represent a distinct phenotypic manifestation caused by TRIOBP variants. The pathogenic variant p.Gln268Leufs*610 disrupts the TRIOBP-4 and TRIOBP-5 isoforms (both expressed exclusively in the inner ear and retina) whereas the second pathogenic variant c.514G>T, p.Gly1672* affects only TRIOBP-5. CONCLUSIONS: The onset and degree of hearing impairment, characteristic for our patients, represent a unique phenotypic manifestation caused by TRIOBP pathogenic variants. Although TRIOBP alterations are not a frequent cause of hearing impairment, this gene should be thoroughly analyzed especially in patients with a postlingual hearing loss. A delayed onset of hearing impairment due to TRIOBP pathogenic variants creates a potential therapeutic window for future targeted therapies.


Asunto(s)
Pérdida Auditiva Sensorineural/genética , Proteínas de Microfilamentos/genética , Mutación , Edad de Inicio , Niño , Preescolar , Exoma/genética , Femenino , Humanos , Masculino , Linaje , Polonia , Isoformas de Proteínas/genética , Análisis de Secuencia de ADN , Secuenciación del Exoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA