Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mamm Genome ; 35(1): 1-12, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38351344

RESUMEN

Wolfram syndrome (OMIM 222300) is a rare autosomal recessive disease with a devastating array of symptoms, including diabetes mellitus, optic nerve atrophy, diabetes insipidus, hearing loss, and neurological dysfunction. The discovery of the causative gene, WFS1, has propelled research on this disease. However, a comprehensive understanding of the function of WFS1 remains unknown, making the development of effective treatment a pressing challenge. To bridge these knowledge gaps, disease models for Wolfram syndrome are indispensable, and understanding the characteristics of each model is critical. This review will provide a summary of the current knowledge regarding WFS1 function and offer a comprehensive overview of established disease models for Wolfram syndrome, covering animal models such as mice, rats, flies, and zebrafish, along with induced pluripotent stem cell (iPSC)-derived human cellular models. These models replicate key aspects of Wolfram syndrome, contributing to a deeper understanding of its pathogenesis and providing a platform for discovering potential therapeutic approaches.


Asunto(s)
Atrofia Óptica , Síndrome de Wolfram , Humanos , Ratas , Ratones , Animales , Síndrome de Wolfram/genética , Síndrome de Wolfram/terapia , Síndrome de Wolfram/diagnóstico , Pez Cebra , Atrofia Óptica/diagnóstico , Atrofia Óptica/genética , Mutación , Proteínas de Unión a Calmodulina/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo
2.
J Hum Genet ; 67(10): 607-611, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35606504

RESUMEN

Silver-Russel syndrome (SRS) is a representative imprinting disorder (ID) characterized by growth failure and diagnosed by clinical features. Recently, international consensus has recommended using the Netchine-Harbison clinical scoring system (NH-CSS) as clinical diagnostic criteria. Loss of methylation of H19/IGF2:intergenic differentially methylated region (H19LOM) and maternal uniparental disomy chromosome 7 (UPD(7)mat) are common etiologies of SRS; however, other IDs, pathogenic variants (PVs) of genes, and pathogenic copy number variants (PCNVs) have been reported in patients meeting NH-CSS. To clarify the frequency and clinical characteristics of each etiology, we conducted (epi)genetic analysis in 173 patients satisfying NH-CSS. H19LOM and UPD(7)mat were identified in 34.1%. PCNVs, other IDs, and PVs were in 15.0%. Patients with all six NH-CSS items were most frequently observed with H19LOM and UPD(7)mat. This study confirmed the suitability of NH-CSS as clinical diagnostic criteria, the (epi)genetic heterogeneity of SRS, and showed the necessity of further discussion regarding the "SRS spectrum".


Asunto(s)
Síndrome de Silver-Russell , Variaciones en el Número de Copia de ADN , Metilación de ADN , Impresión Genómica , Humanos , Fenotipo , Síndrome de Silver-Russell/diagnóstico , Síndrome de Silver-Russell/genética , Disomía Uniparental/genética
3.
J Med Genet ; 58(6): 427-432, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32576657

RESUMEN

BACKGROUND: ZNF597, encoding a zinc-finger protein, is the human-specific maternally expressed imprinted gene located on 16p13.3. The parent-of-origin expression of ZNF597 is regulated by the ZNF597:TSS-DMR, of which only the paternal allele acquires methylation during postimplantation period. Overexpression of ZNF597 may contribute to some of the phenotypes associated with maternal uniparental disomy of chromosome 16 (UPD(16)mat), and some patients with UPD(16)mat presenting with Silver-Russell syndrome (SRS) phenotype have recently been reported. METHODS: A 6-year-old boy presented with prenatal growth restriction, macrocephaly at birth, forehead protrusion in infancy and clinodactyly of the fifth finger. Methylation, expression, microsatellite marker, single nucleotide polymorphism array and trio whole-exome sequencing analyses were conducted. RESULTS: Isolated hypomethylation of the ZNF597:TSS-DMR and subsequent loss of imprinting and overexpression of ZNF597 were confirmed in the patient. Epigenetic alterations, such as UPD including UPD(16)mat and other methylation defects, were excluded. Pathogenic sequence or copy number variants affecting his phenotypes were not identified, indicating that primary epimutation occurred postzygotically. CONCLUSION: We report the first case of isolated ZNF597 imprinting defect, showing phenotypic overlap with SRS despite not satisfying the clinical SRS criteria. A novel imprinting disorder entity involving the ZNF597 imprinted domain can be speculated.


Asunto(s)
Anomalías Múltiples/genética , Retardo del Crecimiento Fetal/genética , Impresión Genómica , Factores de Transcripción/genética , Niño , Metilación de ADN , Humanos , Masculino , Síndrome de Silver-Russell/genética
4.
Pediatr Transplant ; 25(5): e13971, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33470490

RESUMEN

BACKGROUND: ILNEB constitute an autosomal recessive disorder caused by homozygous or compound heterozygous mutation of the gene for the ITGA3. To date, 8 ILNEB patients have been reported, but all 6 neonatal-onset ILNEB patients suffered early death within 2 years. The most common cause of death among previously reported ILNEB patients was exacerbation of the respiratory condition. METHODS: In this study, we describe a case of ILNEB with neonatal onset in a female patient and the genetic and histopathological testing performed. RESULTS: Our patient had a compound heterozygous mutation in ITGA3. Compared to previously reported patients, this patient exhibited milder clinical and histopathological characteristics. After experiencing a life-threatening respiratory infection at 8 months old, the patient started periodic subcutaneous immunoglobulin treatment once every 1-2 weeks for nephrotic-range proteinuria-induced secondary hypogammaglobulinemia. At the age of 3 years, proteinuria gradually increased with severe edema despite strict internal management. Therefore, our patient underwent unilateral nephrectomy and insertion of a peritoneal dialysis catheter followed by another unilateral nephrectomy. One month later, she underwent an ABO-compatible living-donor kidney transplantation at the age of 4 years. CONCLUSIONS: Our patient is a neonatal-onset ILNEB patient who survived for more than 2 years and underwent successful kidney transplantation.


Asunto(s)
Epidermólisis Ampollosa de la Unión/cirugía , Trasplante de Riñón , Enfermedades Pulmonares Intersticiales/cirugía , Síndrome Nefrótico/cirugía , Epidermólisis Ampollosa de la Unión/genética , Femenino , Marcadores Genéticos , Humanos , Recién Nacido , Integrina alfa3/genética , Enfermedades Pulmonares Intersticiales/congénito , Enfermedades Pulmonares Intersticiales/genética , Mutación , Nefrectomía , Síndrome Nefrótico/congénito , Síndrome Nefrótico/genética , Síndrome
5.
Gan To Kagaku Ryoho ; 48(13): 1640-1642, 2021 Dec.
Artículo en Japonés | MEDLINE | ID: mdl-35046282

RESUMEN

The patient is a 40-year-old male. He was referred to our department because, after a thorough examination, he was diagnosed with rectal cancer. Preoperative imaging showed a tumor in the rectum at the level of the seminal vesicles, and left lateral invasion was suspected. In addition, lymph node metastases in the left lateral area were suspected. We performed a robot-assisted low anterior resection plus bilateral lateral dissection plus covering ileostomy for this patient after neoadjuvant chemotherapy. The operation time was 495 minutes, and the blood loss was 50 g. The histopathological diagnosis was pT3, N3(#263), M0, pStage Ⅲc, PM0, DM0, RM0, R0, Cur A. In Japan, robotic-assisted surgery for rectal cancer has been covered by insurance since April 2018, and in our department, robotic surgery is the first option for any stage or type of surgery for rectal cancer. We believe that the greatest advantages of robotic surgery for rectal cancer are in lateral dissection, ie, the better understanding of how blood vessels and nerves travel around the internal iliac vessels and the associated anatomy of pelvic organs that comes from reliable lateral dissection. We have experienced a case of safe robotic-assisted radical resection of laterally invasive rectal cancer, which is considered to be relatively difficult, and we hereby report the usefulness of the robotic-assisted modality.


Asunto(s)
Laparoscopía , Proctectomía , Neoplasias del Recto , Procedimientos Quirúrgicos Robotizados , Adulto , Humanos , Escisión del Ganglio Linfático , Masculino , Terapia Neoadyuvante , Neoplasias del Recto/tratamiento farmacológico , Neoplasias del Recto/cirugía , Recto , Resultado del Tratamiento
6.
Gan To Kagaku Ryoho ; 48(13): 1706-1708, 2021 Dec.
Artículo en Japonés | MEDLINE | ID: mdl-35046304

RESUMEN

The patient was a 57-year-old male. He was diagnosed with locally advanced rectal cancer infiltrating the left levator ani muscle. Chemotherapy(S-1 plus L-OHP plus bevacizumab regimen)was started for the purpose of obtaining a negative circumferential radial margin. After the second course, he presented with perforation of the sigmoid colon for which an emergency operation was performed. The perforation was located 5 centimeters above the tumor in the sigmoid colon. We performed partial resection of the sigmoid colon to repair the perforation and create a sigmoid colostomy. CT, after the initial S-1 plus L-OHP plus bevacizumab chemotherapy regimen, revealed tumor shrinkage. Following 2 more courses of chemotherapy( S-1 plus L-OHP regimen), we performed transanal total mesenteric excision(taTME)as curative surgery. R0 resection was achieved. The combined transanal and laparoscopic approach was highly effective for a patient with pan-peritonitis.


Asunto(s)
Laparoscopía , Proctectomía , Neoplasias del Recto , Colostomía , Humanos , Masculino , Mesenterio , Persona de Mediana Edad , Neoplasias del Recto/tratamiento farmacológico , Neoplasias del Recto/cirugía , Recto
7.
Am J Med Genet A ; 182(4): 735-739, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31880411

RESUMEN

We present two unrelated Japanese pedigrees with achondrogenesis type 1b (ACG1B), characterized by prenatally lethal fetal hydrops and severe micromelia. The affected members in these pedigrees carried a common homozygous missense point mutation in solute carrier family 26 member 2 (SLC26A2), a gene associated with ACG1B (NM_000112:c.1987G>A). This loss-of-function point mutation causes substitution of glycine 663 with arginine in a highly conserved loop domain of SLC26A2. Interestingly, only a few cases of this mutation have been registered in Japanese genomic databases, and there are no reports of this mutation in any major genomic databases outside Japan. Furthermore, we confirmed the presence of a homozygous stretch of approximately 75 kb surrounding the pathogenic variant. Our findings suggest that this missense point mutation in SLC26A2, which is likely the cause of the ACG1B phenotypes in these unrelated fetuses, is distributed exclusively in Japan.


Asunto(s)
Acondroplasia/patología , Mutación , Transportadores de Sulfato/genética , Acondroplasia/genética , Adulto , Femenino , Humanos , Japón , Masculino , Linaje , Fenotipo
8.
J Med Genet ; 56(6): 413-418, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30242100

RESUMEN

BACKGROUND: Recently, a patient with maternal uniparental disomy of chromosome 16 (UPD(16)mat) presenting with Silver-Russell syndrome (SRS) phenotype was reported. SRS is characterised by growth failure and dysmorphic features. OBJECTIVE: To clarify the prevalence of UPD(16)mat in aetiology-unknown patients with SRS phenotype and phenotypic differences between UPD(16)mat and SRS. METHODS: We studied 94 patients with SRS phenotype of unknown aetiology. Sixty-three satisfied the Netchine-Harbison clinical scoring system (NH-CSS) criteria, and 25 out of 63 patients showed both protruding forehead and relative macrocephaly (clinical SRS). The remaining 31 patients met only three NH-CSS criteria, but were clinically suspected as having SRS. To detect UPD(16)mat, we performed methylation analysis for the ZNF597:TSS-differentially methylated region (DMR) on chromosome 16 and subsequently performed microsatellite, SNP array and exome analyses in the patients with hypomethylated ZNF597:TSS-DMR. RESULTS: We identified two patients (2.1%) with a mixture of maternal isodisomy and heterodisomy of chromosome 16 in 94 aetiology-unknown patients with SRS phenotype. Both patients exhibited preterm birth and prenatal and postnatal growth failure. The male patient had ventricular septal defect and hypospadias. Whole-exome sequencing detected no gene mutations related to their phenotypes. CONCLUSION: We suggest considering genetic testing for UPD(16)mat in SRS phenotypic patients without known aetiology.


Asunto(s)
Cromosomas Humanos Par 16 , Metilación de ADN , Fenotipo , Síndrome de Silver-Russell/diagnóstico , Síndrome de Silver-Russell/etiología , Disomía Uniparental , Adolescente , Adulto , Niño , Preescolar , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Lactante , Masculino , Factores de Transcripción/genética , Adulto Joven
9.
Endocr J ; 67(8): 853-857, 2020 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-32321882

RESUMEN

Cytochrome P450 oxidoreductase deficiency (PORD) is a disorder of steroidogenesis that causes various symptoms such as skeletal malformations, disorders of sex development, and adrenal insufficiency. The aim of this study was to elucidate the clinical characteristics, especially age at diagnosis and treatment, of PORD from the perinatal period to adulthood in Japan. The first questionnaire was sent to 183 council members of the Japanese Society for Pediatric Endocrinology on 1 September 2018. The response rate was 65%, and a total of 39 patients with PORD were examined at 20 hospitals. The second questionnaire was sent in November 2018 to the council members examining these 39 patients with PORD. The response rate was 77%, and we received clinical information on 30 of the 39 patients. The two novel clinical findings were the age at diagnosis and the treatment of Japanese patients with PORD. In many cases, PORD can be diagnosed at <3 months of age. Hydrocortisone as the primary treatment during infancy can be used daily or in stressful situations; however, because patients with PORD generally have mild to moderate adrenal insufficiency, some might be able to avoid hydrocortisone treatment. Patients with PORD should be carefully followed up, and treatment should be optimized as for patients with other types of adrenal insufficiency. Other characteristics in the present study were similar to those described in previous reports.


Asunto(s)
Fenotipo del Síndrome de Antley-Bixler/epidemiología , Fenotipo del Síndrome de Antley-Bixler/terapia , Adolescente , Adulto , Edad de Inicio , Fenotipo del Síndrome de Antley-Bixler/diagnóstico , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Japón/epidemiología , Masculino , Persona de Mediana Edad , Embarazo , Encuestas y Cuestionarios , Adulto Joven
10.
J Med Genet ; 55(8): 567-570, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29455159

RESUMEN

BACKGROUND: Paternal uniparental disomy for chromosome 7 (upd(7)pat) is extremely rare, and only four cases have been previously reported. As these cases were accompanied by autosomal-recessive disorders which are likely to be involved in growth restriction, the relevance of upd(7)pat to the overgrowth phenotype remains unclear. Here we describe one case of upd(7)pat with no additional genetic diseases, which may answer the question. METHODS: A 5-year-old Japanese boy presented with a tall stature of unknown causes. To detect the genetic cause of the tall stature, we performed Sanger sequencing, targeted resequencing, comparative genomic hybridisation and single-nucleotide polymorphism (SNP) array analyses, methylation analysis and microsatellite analysis. RESULTS: We could not detect pathogenic variants in causative genes for overgrowth syndrome or apparent copy number alterations. DNA methylation analysis revealed hypomethylation at the GRB10, PEG1 and PEG10 differentially methylated regions. SNP array and microsatellite analyses suggested paternal uniparental isodisomy for chromosome 7. Furthermore, we could not identify homozygous mutations of known causative genes for inherited disorders on chromosome 7. CONCLUSION: We report the first case of upd(7)pat with an overgrowth phenotype.


Asunto(s)
Cromosomas Humanos Par 7 , Trastornos del Crecimiento/diagnóstico , Trastornos del Crecimiento/genética , Herencia Paterna , Fenotipo , Disomía Uniparental , Alelos , Preescolar , Hibridación Genómica Comparativa , Metilación de ADN , Estudios de Asociación Genética , Marcadores Genéticos , Gráficos de Crecimiento , Humanos , Masculino , Polimorfismo de Nucleótido Simple
11.
Hum Mutat ; 39(6): 830-833, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29570232

RESUMEN

STX2 encodes a sulfoglycolipid transporter. Although Stx2 nullizygosity is known to cause spermatogenic failure in mice, STX2 mutations have not been identified in humans. Here, we performed STX2 mutation analysis for 131 Japanese men clinically diagnosed with nonobstructive azoospermia. As a result, we identified a homozygous frameshift mutation [c.8_12delACCGG, p.(Asp3Alafs*8)] in one patient. The mutation-positive patient exhibited loss-of-heterozygosity for 58.4 Mb genomic regions involving STX2, suggesting possible parental consanguinity. The patient showed azoospermia, relatively small testes, and a mildly elevated follicle stimulating hormone level, but no additional clinical features. Testicular histology of the patient showed universal maturation arrest and multinucleated spermatocytes, which have also been observed in mice lacking Stx2. PCR-based cDNA screening revealed wildtype STX2 expression in various tissues including the testis. Our results indicate that STX2 nullizygosity results in nonsyndromic maturation arrest with multinucleated spermatocytes, and accounts for a small fraction of cases with nonobstructive azoospermia.


Asunto(s)
Azoospermia/genética , Espermatogénesis/genética , Sintaxina 1/genética , Adulto , Animales , Azoospermia/patología , Humanos , Pérdida de Heterocigocidad/genética , Masculino , Ratones , Mutación , Testículo/crecimiento & desarrollo , Testículo/metabolismo
12.
J Hum Genet ; 63(3): 377-381, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29311684

RESUMEN

The predominant symptoms of trisomy 14 mosaicism are prenatal and postnatal growth failure, ear abnormalities, congenital heart disease, developmental delay, and genitourinary abnormalities. Maternal uniparental disomy of chromosome 14 (upd(14)mat) presents discernible clinical features such as prenatal and postnatal growth failure, hypotonia, precocious puberty, and obesity. Given the small number of previously reported patients with a combination of trisomy 14 mosaicism and upd(14)mat, the detailed clinical features of these patients remain to be elucidated. Here we report a severely short-statured girl with feeding difficulties and failure to thrive, ear abnormalities, deafness, small hands, and developmental delay. Karyotyping, FISH analysis, methylation analysis, and microsatellite marker analysis using her leukocytes and buccal cells showed that she had a combination of trisomy 14 mosaicism and upd(14)mat. Furthermore, a comparison of the clinical features of this patient with those of previously reported patients with genetic anomalies including the combination of trisomy 14 mosaicism and upd(14)mat or upd(14)mat suggested that the severe short stature observed in patients with a combination of trisomy 14 mosaicism and upd(14)mat stemmed from the synergic effect of these two events. In severely short-statured patients with trisomy 14 mosaicism, we should be aware of the possible coexistence of upd(14)mat.


Asunto(s)
Cariotipo Anormal , Enanismo/diagnóstico , Enanismo/genética , Estudios de Asociación Genética , Fenotipo , Trisomía/genética , Disomía Uniparental , Biomarcadores , Preescolar , Bandeo Cromosómico , Cromosomas Humanos Par 14/genética , Femenino , Impresión Genómica , Gráficos de Crecimiento , Humanos , Hibridación Fluorescente in Situ , Japón , Repeticiones de Microsatélite , Mosaicismo
13.
Pediatr Diabetes ; 19(2): 243-250, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28597946

RESUMEN

BACKGROUND: Mutations in causative genes for neonatal diabetes or maturity-onset diabetes of the young have been identified in multiple patients with autoantibody-negative type 1 diabetes (T1D). OBJECTIVES: We aimed to clarify the prevalence and phenotypic characteristics of monogenic abnormalities among 89 children with autoantibody-negative insulin-requiring T1D. METHODS: Mutations in 30 genes were screened using next-generation sequencing, and copy-number alterations of 4 major causative genes were examined using multiplex-ligation-dependent probe amplification. We compared the clinical characteristics between mutation carriers and non-carriers. RESULTS: We identified 11 probable pathogenic substitutions (6 in INS , 2 in HNF1A , 2 in HNF4A , and 1 in HNF1B ) in 11 cases, but no copy-number abnormalities. Only 2 mutation carriers had affected parents. De novo occurrence was confirmed for 3 mutations. The non-carrier group, but not the carrier group, was enriched with susceptible HLA alleles. Mutation carriers exhibited comparable phenotypes to those of non-carriers, except for a relatively normal body mass index (BMI) at diagnosis. CONCLUSIONS: This study demonstrated significant genetic overlap between autoantibody-negative T1D and monogenic diabetes. Mutations in INS and HNF genes, but not those in GCK and other monogenic diabetes genes, likely play critical roles in children with insulin-requiring T1D. This study also suggests the relatively high de novo rates of INS and HNF mutations, and the etiological link between autoimmune abnormalities and T1D in the non-carrier group. Carriers of monogenic mutations show non-specific phenotypes among all T1D cases, although they are more likely to have a normal BMI at diagnosis than non-carriers.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Predisposición Genética a la Enfermedad , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 1-beta del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/genética , Insulina/genética , Mutación , Niño , Preescolar , Estudios de Cohortes , Variaciones en el Número de Copia de ADN , Análisis Mutacional de ADN , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/fisiopatología , Femenino , Estudios de Asociación Genética , Pruebas Genéticas , Factor Nuclear 1-alfa del Hepatocito/química , Factor Nuclear 1-beta del Hepatocito/química , Factor Nuclear 4 del Hepatocito/química , Heterocigoto , Humanos , Hipoglucemiantes/uso terapéutico , Insulina/química , Insulina/uso terapéutico , Japón , Masculino
14.
Genet Med ; 19(4): 476-482, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27632690

RESUMEN

PURPOSE: Recent studies have identified multilocus imprinting disturbances (MLIDs) in a subset of patients with imprinting diseases (IDs) caused by epimutations. We examined MLIDs in patients with Temple syndrome (TS14) and Kagami-Ogata syndrome (KOS14). METHODS: We studied four TS14 patients (patients 1-4) and five KOS14 patients (patients 5-9) with epimutations. We performed HumanMethylation450 BeadChip (HM450k) analysis for 43 differentially methylated regions (DMRs) (753 CpG sites) and pyrosequencing for 12 DMRs (62 CpG sites) using leukocyte genomic DNA (Leu-gDNA) of patients 1-9, and performed HM450k analysis for 43 DMRs (a slightly different set of 753 CpG sites) using buccal cell gDNA (Buc-gDNA) of patients 1, 3, and 4. We also performed mutation analysis for six causative and candidate genes for MLIDs and quantitative expression analysis using immortalized lymphocytes in MLID-positive patients. RESULTS: Methylation analysis showed hypermethylated ZDBF2-DMR and ZNF597/NAA60-DMR, hypomethylated ZNF597-DMR in both Leu-gDNA and Buc-gDNA, and hypomethylated PPIEL-DMR in Buc-gDNA of patient 1, and hypermethylated GNAS-A/B-DMR in Leu-gDNA of patient 3. No mutations were detected in the six genes for MLIDs. Expression patterns of ZDBF2, ZNF597, and GNAS-A/B were consistent with the identified MLIDs. CONCLUSION: This study indicates the presence of MLIDs in TS14 patients but not in KOS14 patients.Genet Med 19 4, 476-482.


Asunto(s)
Cromosomas Humanos Par 14/genética , Metilación de ADN , Pruebas Genéticas/métodos , Disomía Uniparental/genética , Islas de CpG , Proteínas de Unión al ADN/genética , Impresión Genómica , Humanos , Mutación , Factores de Transcripción/genética
15.
Genet Med ; 19(12): 1356-1366, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28640239

RESUMEN

PurposeTemple syndrome (TS14) is a rare imprinting disorder caused by aberrations at the 14q32.2 imprinted region. Here, we report comprehensive molecular and clinical findings in 32 Japanese patients with TS14.MethodsWe performed molecular studies for TS14 in 356 patients with variable phenotypes, and clinical studies in all TS14 patients, including 13 previously reported.ResultsWe identified 19 new patients with TS14, and the total of 32 patients was made up of 23 patients with maternal uniparental disomy (UPD(14)mat), six patients with epimutations, and three patients with microdeletions. Clinical studies revealed both Prader-Willi syndrome (PWS)-like marked hypotonia and Silver-Russell syndrome (SRS)-like phenotype in 50% of patients, PWS-like hypotonia alone in 20% of patients, SRS-like phenotype alone in 20% of patients, and nonsyndromic growth failure in the remaining 10% of patients in infancy, and gonadotropin-dependent precocious puberty in 76% of patients who were pubescent or older.ConclusionThese results suggest that TS14 is not only a genetically diagnosed entity but also a clinically recognizable disorder. Genetic testing for TS14 should be considered in patients with growth failure plus both PWS-like hypotonia and SRS-like phenotypes in infancy, and/or precocious puberty, as well as a familial history of Kagami-Ogata syndrome due to maternal microdeletion at 14q32.2.


Asunto(s)
Aberraciones Cromosómicas , Trastornos de los Cromosomas/diagnóstico , Trastornos de los Cromosomas/genética , Cromosomas Humanos Par 14 , Impresión Genómica , Fenotipo , Adolescente , Adulto , Niño , Preescolar , Facies , Femenino , Estudios de Asociación Genética , Pruebas Genéticas , Gráficos de Crecimiento , Humanos , Lactante , Japón , Masculino , Persona de Mediana Edad , Embarazo , Adulto Joven
16.
Pediatr Diabetes ; 18(8): 934-941, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28271591

RESUMEN

BACKGROUND: Wolfram syndrome (WS) is a disorder characterized by the association of insulin-dependent diabetes mellitus (DM), diabetes insipidus, deafness, and optic nerve atrophy. WS is caused by WFS1 mutations encoding WFS1 protein expressed in endoplasmic reticulum (ER). During ER protein synthesis, misfolded and unfolded proteins accumulate, known as "ER stress". This is attenuated by the unfolded protein response (UPR), which recovers and maintains ER functions. Because WFS1 is a UPR component, mutant WFS1 might cause unresolvable ER stress conditions and cell apoptosis, the major causes underlying WS symptoms. We encountered an 11-month-old Japanese female WS patient with insulin-dependent DM, congenital cataract and severe bilateral hearing loss. OBJECTIVE: Analyze the WFS1 and functional consequence of the patient WFS1 in vitro. RESULTS: The patient WFS1 contained a heterozygous 4 amino acid in-frame deletion (p.N325_I328del). Her mutant WFS1 increased GRP78 and ATF6α promoter activities in the absence of thapsigargin, indicating constitutive ER stress and nuclear factor of activated T-cell reporter activity, reflecting elevated cytosolic Ca2+ signals. Mutant transfection into cells reduced mRNA expression levels of sarcoplasmic/endoplasmic reticulum Ca2+ transport ATPase 2b (SERCA2b) compared with wild type. Because SERCA2b is required for ER and cytoplasmic Ca2+ homeostasis, decreased SERCA2b expression might affect ER Ca2+ efflux, causing cell apoptosis. CONCLUSION: A novel heterozygous mutation of WFS1 induced constitutive ER stress through ATF6α activation and ER Ca2+ efflux, resulting in cell apoptosis. These results provide new insights into the roles of WFS1 in UPR and mechanism of monogenic DM.


Asunto(s)
Estrés del Retículo Endoplásmico , Proteínas de la Membrana/genética , Síndrome de Wolfram/genética , Calcio/metabolismo , Chaperón BiP del Retículo Endoplásmico , Femenino , Heterocigoto , Humanos , Lactante , Proteínas de la Membrana/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Factor de Transcripción CHOP/metabolismo , Síndrome de Wolfram/diagnóstico , Síndrome de Wolfram/metabolismo
17.
Endocr J ; 64(8): 813-817, 2017 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-28659543

RESUMEN

Hypogonadotropic hypogonadism (HH) is a genetically heterogeneous condition that occurs either as an isolated disorder or as a component of congenital malformation syndromes. SOX2 is a causative gene of syndromic HH characterized by anophthalmia, microphthalmia, or coloboma and other neurological defects such as epilepsy. To date, the causal relationship between SOX2 abnormalities and non-syndromic HH remains speculative. Here, we identified a nonsense mutation of SOX2 in a male patient clinically diagnosed with non-syndromic HH. The patient had epilepsy but no additional clinical features. Ophthalmological examination revealed no abnormalities except for decreased thickness of the retinal nerve fiber layer. Audiometry showed mild sensorineural hearing impairment of both ears. Hormonal evaluation suggested isolated gonadotropin deficiency. Next-generation sequencing-based mutation screening of 13 major causative genes for HH identified a p.Lys35∗ mutation in SOX2 and excluded pathogenic mutations in other tested genes. The p.Lys35∗ mutation appeared to encode a non-functioning SOX2 protein that lacks 283 of 317 amino acids. The SOX2 mutation was absent in the maternal DNA sample, while a paternal sample was unavailable for sequence analysis. These results expand the clinical consequences of SOX2 haploinsufficiency to include non-syndromic HH. Systematic mutation screening using a next-generation sequencer and detailed evaluation of nonspecific ocular/neurological features may help identify SOX2 mutation-positive individuals among HH patients.


Asunto(s)
Codón sin Sentido , Hipogonadismo/genética , Factores de Transcripción SOXB1/genética , Análisis Mutacional de ADN , Humanos , Masculino , Fenotipo , Adulto Joven
18.
Endocr J ; 64(10): 947-954, 2017 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-28768959

RESUMEN

Although mutations in ACAN, FGFR3, NPR2, and SHOX typically lead to skeletal dysplasia, and mutations in GHRHR, GH1, GHR, STAT5B, IGF1, IGFALS, and IGF1R usually underlie hormonal defects of the growth hormone (GH)-insulin-like growth factor 1 (IGF1) axis, such mutations have also been identified in patients with idiopathic short stature (ISS). Of these, SHOX abnormalities are known to account for a certain percentage of ISS cases, whereas the frequency of mutations in the other 10 genes in ISS cohorts remains unknown. Here, we performed next-generation sequencing-based mutation screening of the 10 genes in 86 unrelated Japanese ISS patients without SHOX abnormalities. We searched for rare protein-altering variants. The functional significance of the identified variants was assessed by in silico analyses. Consequently, we identified 18 heterozygous rare variants in 19 patients, including four probable damaging variants in ACAN, six pathogenicity-unknown variants in FGFR3, GHRHR, GHR, and IGFALS, and eight possible benign variants. Pathogenic variants in NPR2, GH1, and IGF1 were absent from our cohort. Unlike previously reported patients with ACAN mutations, our four patients with ACAN variants manifested non-specific short stature with age-appropriate or mildly delayed bone ages, and had parents of normal stature. These results indicate that ACAN mutations can underlie ISS without characteristic skeletal features, and that such mutations are possibly associated with de novo occurrence or low penetrance. In addition, our data imply that mutations in FGFR3, NPR2, and GH-IGF1 axis genes play only limited roles in the etiology of ISS.


Asunto(s)
Agrecanos/genética , Predisposición Genética a la Enfermedad , Trastornos del Crecimiento/genética , Mutación , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Receptores de Neuropéptido/genética , Receptores de Hormona Reguladora de Hormona Hipofisaria/genética , Agrecanos/química , Agrecanos/metabolismo , Sustitución de Aminoácidos , Proteínas Portadoras/química , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Niño , Preescolar , Estudios de Cohortes , Biología Computacional , Bases de Datos Genéticas , Sistemas Especialistas , Femenino , Estudios de Asociación Genética , Pruebas Genéticas , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/metabolismo , Trastornos del Crecimiento/sangre , Trastornos del Crecimiento/metabolismo , Trastornos del Crecimiento/fisiopatología , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Japón , Masculino , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/química , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor IGF Tipo 1 , Receptores de Neuropéptido/química , Receptores de Neuropéptido/metabolismo , Receptores de Hormona Reguladora de Hormona Hipofisaria/química , Receptores de Hormona Reguladora de Hormona Hipofisaria/metabolismo , Receptores de Somatomedina/química , Receptores de Somatomedina/genética , Receptores de Somatomedina/metabolismo , Factor de Transcripción STAT5/química , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo
19.
Pediatr Int ; 58(11): 1229-1231, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27882740

RESUMEN

Pseudohypoparathyroidism type 1b (PHP-1b) is usually diagnosed on various symptoms of hypocalcemia. Previous studies reported a few cases of autosomal dominant pattern PHP-1b identified on familial analysis with asymptomatic hypocalcemia. Herein we report the case of a 6-year-old male patient with sporadic PHP-1b incidentally detected on preoperative examination. He had neither characteristic findings of Albright hereditary osteodystrophy nor evidence of tetany. Sporadic PHP-1b was diagnosed on the basis of clinical observation and laboratory examination. In addition, genetic testing using methylation-specific multiplex ligation-dependent probe amplification indicated broad methylation abnormalities and confirmed the sporadic form of PHP-1b. Sporadic PHP-1b might often be overlooked when diagnosis is done simply on definitive clinical features. To avoid this, DNA sequencing and methylation analysis should be performed even in the absence of definitive clinical features.


Asunto(s)
Cromograninas/genética , ADN/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Hipocalcemia/etiología , Seudohipoparatiroidismo/diagnóstico , Niño , Cromograninas/metabolismo , Análisis Mutacional de ADN , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Pruebas Genéticas , Humanos , Hipocalcemia/sangre , Hipocalcemia/diagnóstico , Masculino , Seudohipoparatiroidismo/complicaciones , Seudohipoparatiroidismo/genética , Seudohipoparatiroidismo
20.
J Hum Genet ; 60(9): 553-6, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26040210

RESUMEN

Pseudoautosomal region 1 (PAR1) contains SHOX, in addition to seven highly conserved non-coding DNA elements (CNEs) with cis-regulatory activity. Microdeletions involving SHOX exons 1-6a and/or the CNEs result in idiopathic short stature (ISS) and Leri-Weill dyschondrosteosis (LWD). Here, we report six rare copy-number variations (CNVs) in PAR1 identified through copy-number analyzes of 245 ISS/LWD patients and 15 unaffected individuals. The six CNVs consisted of three microduplications encompassing SHOX and some of the CNEs, two microduplications in the SHOX 3'-region affecting one or four of the downstream CNEs, and a microdeletion involving SHOX exon 6b and its neighboring CNE. The amplified DNA fragments of two SHOX-containing duplications were detected at chromosomal regions adjacent to the original positions. The breakpoints of a SHOX-containing duplication resided within Alu repeats. A microduplication encompassing four downstream CNEs was identified in an unaffected father-daughter pair, whereas the other five CNVs were detected in ISS patients. These results suggest that microduplications involving SHOX cause ISS by disrupting the cis-regulatory machinery of this gene and that at least some of microduplications in PAR1 arise from Alu-mediated non-allelic homologous recombination. The pathogenicity of other rare PAR1-linked CNVs, such as CNE-containing microduplications and exon 6b-flanking microdeletions, merits further investigation.


Asunto(s)
Variaciones en el Número de Copia de ADN , Trastornos del Crecimiento/genética , Proteínas de Homeodominio/genética , Región de Flanqueo 3'/genética , Región de Flanqueo 5'/genética , Estudios de Casos y Controles , Niño , Preescolar , Enanismo/genética , Femenino , Duplicación de Gen , Frecuencia de los Genes , Humanos , Lactante , Masculino , Persona de Mediana Edad , Osteocondrodisplasias/genética , Eliminación de Secuencia , Proteína de la Caja Homeótica de Baja Estatura , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA