Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Zhonghua Bing Li Xue Za Zhi ; 47(5): 354-359, 2018 May 08.
Artículo en Zh | MEDLINE | ID: mdl-29783802

RESUMEN

Objective: To investigate the clinicopathologic and molecular features of the rare cribriform morular variant of papillary thyroid carcinoma (CMV-PTC). Methods: The clinicopathologic data of 10 patients with CMV-PTC were retrospectively reviewed. Immunohistochemical (IHC) staining was done using LSAB method. DNA sequencing for APC were applied using Sanger method. BRAF V600E mutation was examined using ARMS method. The cytological, morphological, IHC and molecular features were analyzed. Results: All patients were female at an average age of 27 years old. The tumors were mostly located in the right lobe of thyroid. Fine needle aspiration cytology was performed in three patients; two were diagnosed as suspicious for PTC and one as PTC. Nine tumors presented as solitary nodule and two as multiple nodules in both lobes. Infiltration was demonstrated in three cases. The average size was 2.6 cm. The neoplastic cells were arranged in papillary, cribriform, solid and glandular patterns, with rare or without colloid inside the lumen. The number of morula varied, ranging from zero to many. The neoplastic cells were variably enlarged, showing round, oval or spindle shape. Nuclear irregularity was identified as irregular membrane, nuclear grooves or pseudoinclusion, but no typical ground glass feature. Peculiar nuclear clearing could be observed in the morular cells. IHC staining showed the neoplastic cells were negative for thyroglobulin and p63, but positive for TTF1, cytokeratin 19 and estrogen receptor. Diffuse staining with cytokeratin was seen in the neoplastic cells and the morula. Specific cytoplasmic and nuclear staining of ß-catenin was seen in the neoplastic cells but not the morula. Ki-67 proliferation index was 1%-30%. No recurrence or metastasis was observed. One patient was demonstrated to harbor both somatic and germline mutations of the APC gene, who was found to have adenomatous polyposis and her mother died of colonic carcinoma. No BRAF V600E mutation was detected. Conclusions: CMV-PTC is rare and shows atypical cytological and clinicopathological features, and it is easily misdiagnosed.TG, TTF1, ER and ß-catenin are specific IHC markers for CMV-PTC. The morula is negative for cytokeratin 19, in contrast to squamous metaplasia. Although CMV-PTC has indolent clinical behavior, a definite diagnosis is necessary to rule out the possibility of APC gene mutation and related extra-thyroidal neoplasm, such as FAP and Gardner syndrome.


Asunto(s)
Carcinoma Papilar/genética , Carcinoma Papilar/patología , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Poliposis Adenomatosa del Colon , Adulto , Biomarcadores de Tumor/metabolismo , Biopsia con Aguja Fina , Carcinoma Papilar/metabolismo , Núcleo Celular , Femenino , Humanos , Queratina-19/metabolismo , Mutación , Recurrencia Local de Neoplasia , Estudios Retrospectivos , Tiroglobulina/metabolismo , Cáncer Papilar Tiroideo , Neoplasias de la Tiroides/metabolismo , Carga Tumoral , beta Catenina/metabolismo
2.
Acta Otolaryngol ; 128(7): 732-8, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18568513

RESUMEN

CONCLUSIONS: Significant difference in the incidence of mitochondrial DNA (mtDNA) mutations was found between the Chinese and USA populations. The identification of the mtDNA A1555G mutation in a large proportion of Chinese probands with nonsyndromic sensorineural hearing loss (NSHL) provides a molecular explanation for the high prevalence of aminoglycoside-induced deafness in China. OBJECTIVE: The aim was to characterize the audiological and genetic features of NSHL due to mutations in mtDNA. SUBJECTS AND METHODS: The mtDNA and audiogram analyses were performed in 498 NSHL patients (290 from China and 208 from the USA) with and without history of aminoglycoside exposure. A PCR and restriction enzyme digestion protocol was used for mutational screening and the European Workshop on Genetic Hearing Loss criteria were applied for audiological classification. RESULTS: All Chinese probands (15.5%) with mtDNA mutation were found to carry the homoplasmic mtDNA A1555G mutation, whereas four probands (1.9%) from the USA were found to carry the mtDNA A1555G and two (1%) had mtDNA G7444A. Approximately 63% of the probands with mtDNA mutations had post-lingual hearing loss and 56.8% of them had a medical history of exposure to aminoglycosides. Hearing losses are bilateral, sensorineural, and symmetric. The main audiogram shapes found were sloping.


Asunto(s)
ADN Mitocondrial/genética , Pérdida Auditiva Sensorineural/fisiopatología , Audición/fisiología , Mutación , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Aminoglicósidos/efectos adversos , Pueblo Asiatico , Audiometría de Tonos Puros , Niño , China/epidemiología , Sordera/inducido químicamente , Sordera/genética , Sordera/fisiopatología , Frecuencia de los Genes , Pérdida Auditiva Sensorineural/epidemiología , Pérdida Auditiva Sensorineural/genética , Humanos , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Estados Unidos/epidemiología , Población Blanca
3.
J Mol Biol ; 357(3): 755-64, 2006 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-16464467

RESUMEN

The driving forces for the regulation of cell morphology are the Rho family GTPases that coordinate the assembly of the actin cytoskeleton. This dynamic feature is a result of tight coupling between the cytoskeleton and signal transduction and is facilitated by actin-binding proteins (ABPs). Mutations in the actin bundling and PDZ domain-containing protein harmonin are the causes of Usher syndrome type 1C (USH1C), a syndrome of congenital deafness and progressive blindness, as well as certain forms of non-syndromic deafness. Here, we have used the yeast two-hybrid assay to isolate molecular partners of harmonin and identified DOCK4, an unconventional guanine exchange factor for the Rho family of guanosine triphosphatases (Rho GEF GTPases), as a protein interacting with harmonin. Detailed molecular analysis revealed that a novel DOCK4 isoform (DOCK4-Ex49) is expressed in the brain, eye and inner ear tissues. We have further provided evidence that the DOCK4-Ex49 binds to nucleotide free Rac as effectively as DOCK2 and DOCK4 and it is a potent Rac activator. By immunostaining using a peptide antibody specific to DOCK4-Ex49, we showed its localization in the inner ear within the hair bundles along the stereocilia (SC). Together, our data indicate a possible Rac-DOCK4-ABP harmonin-activated signaling pathway in regulating actin cytoskeleton organization in stereocilia.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/metabolismo , Oído Interno/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Actinas/metabolismo , Animales , Proteínas de Ciclo Celular , Línea Celular , Cilios/enzimología , Cilios/metabolismo , Proteínas del Citoesqueleto , Citoesqueleto/metabolismo , Oído Interno/enzimología , Exones , Proteínas Activadoras de GTPasa/inmunología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Ratones , Isoformas de Proteínas/metabolismo , Transducción de Señal/fisiología , Técnicas del Sistema de Dos Híbridos , Proteínas de Unión al GTP rac/metabolismo
4.
J Med Genet ; 43(2): 170-4, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15958501

RESUMEN

BACKGROUND: Non-syndromic hearing loss is among the most genetically heterogeneous traits known in humans. To date, at least 50 loci for autosomal dominant non-syndromic sensorineural hearing loss (ADNSSHL) have been identified by linkage analysis. OBJECTIVE: To report the mapping of a novel autosomal dominant deafness locus on the long arm of chromosome 14 at 14q11.2-q12, DFNA53, in a large multigenerational Chinese family with post-lingual, high frequency hearing loss that progresses to involve all frequencies. RESULTS: A maximum multipoint LOD score of 5.4 was obtained for marker D14S1280. The analysis of recombinant haplotypes mapped DFNA53 to a 9.6 cM region interval between markers D14S581 and D14S1021. Four deafness loci (DFNA9, DFNA23, DFNB5, and DFNB35) have previously been mapped to the long arm of chromosome 14. The critical region for DFNA53 contains the gene for DFNA9 but does not overlap with the regions for DFNB5, DFNA23, or DFNB35. Screening of the COCH gene (DFNA9), BOCT, EFS, and HSPC156 within the DFNA53 interval did not identify the cause for deafness in this family. CONCLUSIONS: Identifying the DFNA53 locus is the first step in isolating the gene responsible for hearing loss in this large multigeneration Chinese family.


Asunto(s)
Cromosomas Humanos Par 14/genética , Proteínas de Unión al ADN/genética , Genes Dominantes/genética , Pérdida Auditiva/genética , Mapeo Físico de Cromosoma , Factores de Transcripción/genética , Adolescente , Adulto , Anciano , Femenino , Haplotipos , Humanos , Escala de Lod , Masculino , Persona de Mediana Edad , Linaje , Fenotipo
6.
Clin Genet ; 72(5): 471-7, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17868390

RESUMEN

We report here the clinical, genetic, and molecular characteristics of a large Chinese family exhibiting non-syndromic, late-onset autosomal dominant sensorineural hearing loss. Clinical evaluation revealed variable phenotypes of hearing loss in terms of severity and age-at-onset of disease in these subjects. Genome-wide linkage analysis mapped the disease gene to the DFNA5 locus with a maximum two-point log odds score of 5.39 at [theta] = 0 for marker D7S2457. DNA sequencing of DFNA5 revealed a novel heterozygous IVS8+4 A>G substitution in the splice donor site of intron 8. Reverse transcriptase-polymerase chain reaction (RT-PCR) showed skipping of exon 8 in the mutant transcript. This mutation faithfully cosegregated with hearing loss in the family. In addition, the mutation was absent in 100 unrelated control DNA samples of Chinese origin. The IVS8+4 A>G mutation is predicted to create a shift in the reading frame and introduce a stop codon at position 372, thereby resulting in a prematurely truncated DFNA5 protein. Up to date, a total of four mutations in DFNA5 have been reported to lead to hearing impairment, all of them result in skipping of exon 8 at the mRNA level. Our findings provide further support for the hypothesis that DFNA5-associated hearing loss is caused by a very specific gain-of-function mutation.


Asunto(s)
Familia , Pérdida Auditiva/genética , Intrones , Mutación , Sitios de Empalme de ARN/genética , Receptores de Estrógenos/genética , Edad de Inicio , Secuencia de Bases , China , Análisis Mutacional de ADN , Femenino , Ligamiento Genético , Humanos , Masculino , Linaje
7.
Clin Genet ; 63(2): 150-3, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12630964

RESUMEN

Usher syndrome (USH) is characterized by the associated findings of hearing loss and retinitis pigmentosa (RP), leading to progressive loss of vision. Three forms of USH can be distinguished clinically. In the most severe form, USH1, profound congenital deafness is associated with vestibular dysfunction and RP. To determine the frequency of USH1C mutations as a cause for USH1, 128 probands with Usher syndrome type 1 including seven from Acadian and 121 from non-Acadian populations were systematically screened for mutations in USH1C using a combined single-strand conformational polymorphisms (SSCP)/heteroduplex and sequencing method. All seven Acadian USH1 patients were found to be homozygous for both the 216G>A mutation and the 9-repeat VNTR which characterizes the Acadian allele, confirming previous evidence for a founder effect by haplotype analysis. However, USH1C mutations were identified in only two non-Acadian USH1 probands (1.65%) including one from Pakistan who was homozygous for a 238-239insC mutation and one from Canada was also homozygous for the Acadian allele. The low prevalence of USH1C mutations in the present study suggests that the high prevalence of the 238-239insC in Germany may reflect a founder effect. Comparison of the affected haplotypes in the Canadian patient with the Acadian USH1 patients yielded evidence for a founder effect. Our data suggest that USH1C is a relatively rare form of USH1 in non-Acadian populations and that in addition to the 216G>A Acadian mutation, the 238-239insC mutation appears to be common in some populations.


Asunto(s)
Proteínas Portadoras/genética , Efecto Fundador , Pérdida Auditiva/genética , Retinitis Pigmentosa/genética , Proteínas Adaptadoras Transductoras de Señales , Alelos , Proteínas de Ciclo Celular , Mapeo Cromosómico , Proteínas del Citoesqueleto , Análisis Mutacional de ADN , Pérdida Auditiva/complicaciones , Pérdida Auditiva/epidemiología , Humanos , Louisiana/epidemiología , Polimorfismo Conformacional Retorcido-Simple , Quebec/etnología , Retinitis Pigmentosa/complicaciones , Retinitis Pigmentosa/epidemiología
8.
Clin Genet ; 65(4): 288-93, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15025721

RESUMEN

Usher syndrome type II is an autosomal recessive disorder characterized by moderate to severe hearing impairment and progressive visual loss due to retinitis pigmentosa (RP). We carried out a mutation screening of the USH2A gene in 88 probands with Usher syndrome type II to determine the frequency of USH2A mutations as a cause for USH2. Six mutations, including 2299delG, 921-922insCAGC, R334W, N346H, R626X, and N357T were identified, with 2299delG mutation being the most frequent (16.5% of alleles), accounting for 77.5% of the pathologic alleles. Thirty-five percent (31/88) of the probands had a USH2A mutation. Nine of them carried two pathogenic mutations: six cases were homozygotes and three were compound heterozygotes. Twenty-two probands (25%) were found to carry only single USH2A mutations. One new missense mutation (N357T) occuring within the laminin N-terminal (type VI) domain of usherin was identified. Eight polymorphisms were found, five of which are novel. Our data support the view that the 2299delG is the most common mutation in USH2A.


Asunto(s)
Proteínas de la Matriz Extracelular/genética , Pérdida Auditiva/genética , Mutación , Retinitis Pigmentosa/genética , Estudios de Casos y Controles , Análisis Mutacional de ADN , Frecuencia de los Genes , Humanos , América del Norte , Polimorfismo Genético , Síndrome , Reino Unido
9.
Hum Mol Genet ; 10(25): 2945-51, 2001 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-11741837

RESUMEN

Mutations in four members of the connexin gene family have been shown to underlie distinct genetic forms of deafness, including GJB2 [connexin 26 (Cx26)], GJB3 (Cx31), GJB6 (Cx30) and GJB1 (Cx32). We have found that alterations in a fifth member of this family, GJA1 (Cx43), appear to cause a common form of deafness in African Americans. We identified two different GJA1 mutations in four of 26 African American probands. Three were homozygous for a Leu-->Phe substitution in the absolutely conserved codon 11, whereas the other was homozygous for a Val-->Ala transversion at the highly conserved codon 24. Neither mutation was detected in DNA from 100 control subjects without deafness. Cx43 is expressed in the cochlea, as is demonstrated by PCR amplification from human fetal cochlear cDNA and by RT-PCR of mouse cochlear tissues. Immunohistochemical staining of mouse cochlear preparations showed immunostaining for Cx43 in non-sensory epithelial cells and in fibrocytes of the spiral ligament and the spiral limbus. To our knowledge this is the first alpha connexin gene to be associated with non-syndromic deafness. Cx43 must also play a critical role in the physiology of hearing, presumably by participating in the recycling of potassium to the cochlear endolymph.


Asunto(s)
Cóclea/metabolismo , Conexina 43/genética , Sordera/genética , Mutación/genética , Secuencia de Aminoácidos , Animales , Conexina 26 , Conexina 43/metabolismo , Conexinas , Análisis Mutacional de ADN , Cartilla de ADN/química , Femenino , Humanos , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Endogámicos CBA , Datos de Secuencia Molecular , Linaje , Fenotipo , Reacción en Cadena de la Polimerasa , Polimorfismo Conformacional Retorcido-Simple , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Síndrome
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda