Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Wound Repair Regen ; 31(3): 384-392, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36866489

RESUMEN

Standard non-invasive methods for diagnosing and selecting the best treatment for osteomyelitis in patients with multiple chronic conditions remain to be established. We aimed to evaluate the ability of quantitative 67 Ga-citrate single-photon emission computed tomography (67 Ga-SPECT/CT) to determine the indication for either non-surgical treatment or osteotomy in patients with lower-limb osteomyelitis (LLOM) associated with diabetes mellitus and lower-extremity ischemia, based on monitoring of inflammatory activity in bone tissue. This single-centre prospective study conducted from January 2012 to July 2017 included 90 consecutive patients with suspected LLOM. Regions of interest were drawn on SPECT images during quantification of Ga accumulation. Subsequently, the inflammation-to-background ratio (IBR) was calculated by dividing the maximal accumulated lesion number by the mean number for the distal femur bone marrow of the unaffected side. Osteotomy was performed in 28 of 90 patients (31%). The osteotomy rate was higher for patients with IBR >8.4 (71.4%) than for those with IBR ≤8.4 (5.5%) (p < 0.001, sensitivity: 0.89, specificity: 0.84). In the multivariate Cox regression analysis, IBR >8.4 was an independent risk factor for osteotomy (hazard ratio [HR]: 19.0, 95% confident interval [CI]: 5.6-63.9, p < 0.001). Transcutaneous oxygen tension (TcPO2 ) was identified as an independent risk factor for lower-limb amputation (HR: 0.96, 95% CI: 0.92-0.99, p = 0.01). The current results indicate that quantitative 67 Ga-SPECT/CT is useful for distinguishing patients with LLOM likely to require osteotomy.


Asunto(s)
Osteomielitis , Radiofármacos , Humanos , Estudios Prospectivos , Radiofármacos/farmacología , Radiofármacos/uso terapéutico , Cicatrización de Heridas , Tomografía Computarizada de Emisión de Fotón Único/efectos adversos , Osteomielitis/diagnóstico por imagen , Osteomielitis/cirugía , Inflamación , Radioisótopos de Galio/uso terapéutico , Osteotomía/efectos adversos , Tomografía Computarizada por Rayos X/efectos adversos
2.
Int Heart J ; 63(2): 235-240, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35354745

RESUMEN

Sustained ventricular tachycardia (sVT), leading to sudden cardiac death, is one of the common manifestations in cardiac sarcoidosis (CS). Although late gadolinium enhancement (LGE) on cardiac magnetic resonance (CMR) has been reported to be associated with sVT, the relationships of its localization to sVT have not been fully evaluated.To evaluate the localization of LGE and its relationships to sVT in patients with CS, we reviewed medical record of consecutive 31 patients with CS who underwent CMR. The localization of LGE was divided into four categories: Left ventricular (LV) septum, LV free wall, right ventricular (RV) septum, and RV free wall. We investigated the association of sVT with localization of LGE and other parameters including serum biomarkers LV ejection fraction on echocardiography and Fluorine-18-fluorodeoxyglucose (FDG) accumulation on positron emission tomography (PET) -CT.Of the studied population, 8 patients (25.8%) were known to present with sVT among 31 CS patients. LGE was observed in the RV free wall in 6 patients with sVT, whereas it was in 5 patients without sVT (75.0% versus 21.7%, P = 0.022). Univariate analysis showed that only LGE in the RV free wall was associated with sVT (odds ratio [OR]: 10.80; 95% confidence interval [CI]: 1.64-70.93, P = 0.013).LGE in the RV free wall was associated with sVT in patients with CS.


Asunto(s)
Cardiomiopatías , Sarcoidosis , Taquicardia Ventricular , Tabique Interventricular , Cardiomiopatías/diagnóstico , Cardiomiopatías/diagnóstico por imagen , Medios de Contraste , Gadolinio , Humanos , Sarcoidosis/diagnóstico , Sarcoidosis/diagnóstico por imagen , Taquicardia Ventricular/diagnóstico por imagen , Taquicardia Ventricular/etiología , Tabique Interventricular/patología
3.
Am J Med Genet A ; 185(7): 2175-2179, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33884742

RESUMEN

Transient receptor potential channel C6 encoded by TRPC6 is involved in slit diaphragm formation in podocytes, and abnormalities of the TRPC6 protein cause various glomerular diseases. The first identified pathogenic variant of TRPC6 was found to cause steroid-resistant nephrotic syndrome that typically developed in adulthood and then slowly led to end-stage renal disease, along with a renal pathology of focal segmental glomerulosclerosis. Here, we report a patient with rapidly progressing infantile nephrotic syndrome and a heterozygous missense TRPC6 variant. The patient, a 2-year-old Japanese boy, developed steroid-resistant nephrotic syndrome at age 11 months. His renal function deteriorated rapidly, and peritoneal dialysis was introduced at age 1 year and 6 months. His renal pathology, obtained at age 1 year and 1 month, was consistent with diffuse mesangial sclerosis (DMS). Clinical exome analysis and custom panel analysis for hereditary renal diseases revealed a reported heterozygous missense variant in TRPC6 (NM_004621.5:c.523C > T:p.Arg175Trp). This is the first report of a patient with a TRPC6-related renal disorder associated with DMS.


Asunto(s)
Enfermedades Renales/genética , Síndrome Nefrótico/genética , Esclerosis/genética , Canal Catiónico TRPC6/genética , Preescolar , Exoma/genética , Predisposición Genética a la Enfermedad , Glomeruloesclerosis Focal y Segmentaria/complicaciones , Glomeruloesclerosis Focal y Segmentaria/diagnóstico por imagen , Glomeruloesclerosis Focal y Segmentaria/genética , Glomeruloesclerosis Focal y Segmentaria/patología , Heterocigoto , Humanos , Lactante , Riñón/diagnóstico por imagen , Riñón/patología , Enfermedades Renales/complicaciones , Enfermedades Renales/diagnóstico por imagen , Enfermedades Renales/patología , Masculino , Mutación Missense/genética , Síndrome Nefrótico/complicaciones , Síndrome Nefrótico/diagnóstico por imagen , Síndrome Nefrótico/patología , Podocitos/metabolismo , Podocitos/patología , Esclerosis/complicaciones , Esclerosis/diagnóstico por imagen , Esclerosis/patología
4.
Am J Med Genet A ; 179(6): 948-957, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30941898

RESUMEN

PIEZO2 encodes a mechanically activated cation channel, which is abundantly expressed in dorsal root ganglion neuron and sensory endings of proprioceptors required for light touch sensation and proprioception in mice. Biallelic loss-of-function mutations in PIEZO2 (i.e., PIEZO2 deficiency) were recently found to cause an arthrogryposis syndrome. Sixteen patients from eight families have been reported to date. Herein we report a new case, including detailed clinical characteristics and courses as well as comprehensive neurological features. The patient was a 12-year-old girl presenting with congenital multiple contractures, progressive severe scoliosis, prenatal-onset growth impairment, motor developmental delay with hypotonia and myopathy-like muscle pathology, mild facial features, and normal intelligence. Her neurological features included areflexia, impaired proprioception, and decreased senses. Neurophysiological examination revealed decreased amplitude of sensory nerve action potentials, absent H reflex, and prolongation of central conduction times. Clinical exome sequencing revealed a novel homozygous frameshift mutation in PIEZO2 (NM_022068: c.4171_4174delGTCA: p.Val1391Lysfs*39) with no detectable mRNA expression of the gene. PIEZO2 deficiency represents a clinical entity involving characteristic neuromuscular abnormalities and physical features. Next generation sequencing-based comprehensive molecular screening and extensive neurophysiological examination could be valuable for diagnosis of the disorder.


Asunto(s)
Artrogriposis/diagnóstico , Artrogriposis/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Canales Iónicos/deficiencia , Fenotipo , Niño , Electromiografía , Facies , Femenino , Expresión Génica , Estudios de Asociación Genética/métodos , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mutación , Degradación de ARNm Mediada por Codón sin Sentido , Análisis de Secuencia de ADN , Síndrome
5.
Gan To Kagaku Ryoho ; 44(2): 107-110, 2017 Feb.
Artículo en Japonés | MEDLINE | ID: mdl-28223668

RESUMEN

Genetic testing, which reveals germline mutations, is extremely important for HBOC patients and their families. The evolution of this field has created a need for accurate cancer genetic counseling and risk assessment. Prevention and early therapy are possible in HBOC. If a patient has BRCA1 or BRCA2 mutations, the at-risk relatives should receive the information through genetic counseling. Genetic counseling provides not only information but also psychological and social support so that the patient or subject can autonomously decide. Clinical practice for the treatment of HBOC needs a multidisciplinary network that includes clinical geneticists, breast cancer surgeons, and ovarian cancer surgeons. Thus, the Japan Society of Human Genetics, Japan Breast Cancer Society, and Japan Society of Obstetrics and Gynecology established a new corporation named Japanese Organization of HBOC(JOHBOC)for a HBOC comprehensive medical care system.


Asunto(s)
Asesoramiento Genético , Síndrome de Cáncer de Mama y Ovario Hereditario/genética , Femenino , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Humanos , Factores de Riesgo
6.
Am J Med Genet A ; 170A(2): 322-328, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26481852

RESUMEN

Beta-propeller protein-associated neurodegeneration (BPAN), also known as static encephalopathy of childhood with neurodegeneration in adulthood (SENDA), is a subtype of neurodegeneration with brain iron accumulation (NBIA). BPAN is caused by mutations in an X-linked gene WDR45 that is involved in autophagy. BPAN is characterized by developmental delay or intellectual disability until adolescence or early adulthood, followed by severe dystonia, parkinsonism, and progressive dementia. Brain magnetic resonance imaging (MRI) shows iron deposition in the bilateral globus pallidus (GP) and substantia nigra (SN). Clinical manifestations and laboratory findings in early childhood are limited. We report a 3-year-old girl with BPAN who presented with severe developmental delay and characteristic facial features. In addition to chronic elevation of serum aspartate transaminase, lactate dehydrogenase, creatine kinase, and soluble interleukin-2 receptor, she had persistent elevation of neuron specific enolase (NSE) in serum and cerebrospinal fluid. MRI using susceptibility-weighted imaging (SWI) demonstrated iron accumulation in the GP and SN bilaterally. Targeted next-generation sequencing identified a de novo splice-site mutation, c.831-1G>C in WDR45, which resulted in aberrant splicing evidenced by reverse transcriptase-PCR. Persistent elevation of NSE and iron deposition on SWI may provide clues for diagnosis of BPAN in early childhood.


Asunto(s)
Trastornos del Metabolismo del Hierro/sangre , Trastornos del Metabolismo del Hierro/diagnóstico , Hierro/metabolismo , Imagen por Resonancia Magnética/métodos , Distrofias Neuroaxonales/sangre , Distrofias Neuroaxonales/diagnóstico , Fosfopiruvato Hidratasa/sangre , Proteínas Portadoras/genética , Preescolar , Femenino , Genes Ligados a X/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Trastornos del Metabolismo del Hierro/genética , Mutación/genética , Distrofias Neuroaxonales/genética , Pronóstico
7.
Am J Med Genet A ; 167A(2): 407-11, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25423878

RESUMEN

Noonan syndrome with multiple lentigines (NSML), formerly referred to as LEOPARD syndrome, is a rare autosomal-dominant condition, characterized by multiple lentigines, electrocardiographic conduction abnormalities, ocular hypertelorism, pulmonary stenosis, abnormal genitalia, growth retardation, and sensorineural deafness. To date, PTPN11, RAF1, and BRAF have been reported to be causal for NSML. We report on a 13-year-old Japanese boy, who was diagnosed with NSML. He was found to have a novel heterozygous missense variant (c.305A > G; p.E102G) in MAP2K1, a gene mostly causal for cardio-facio-cutaneous syndrome (CFCS). He manifested fetal macrosomia, and showed hypotonia and poor sucking in the neonatal period. He had mild developmental delay, and multiple lentigines appearing at approximately age 3 years, as well as flexion deformity of knees bilaterally, subtle facial characteristics including ocular hypertelorism, sensorineural hearing loss, and precocious puberty. He lacked congenital heart defects or hypertrophic cardiomyopathy, frequently observed in patients with NSML, mostly caused by PTPN11 mutations. He also lacked congenital heart defects, characteristic facial features, or intellectual disability, frequently observed in those with CFCS caused by MAP2K1 or MAP2K2 mutations. This may be the first patient clinically diagnosed with NSML, caused by a mutation in MAP2K1.


Asunto(s)
Heterocigoto , Síndrome LEOPARD/diagnóstico , Síndrome LEOPARD/genética , MAP Quinasa Quinasa 1/genética , Mutación , Fenotipo , Adolescente , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Análisis Mutacional de ADN , Facies , Humanos , MAP Quinasa Quinasa 2/genética , Masculino , Datos de Secuencia Molecular
8.
Am J Med Genet A ; 167A(3): 592-601, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25691411

RESUMEN

6p duplication syndrome is a rare chromosomal disorder that frequently manifests renal complications, including proteinuria, hypoplastic kidney, and hydronephrosis. We report a girl with the syndrome, manifesting left hydronephrosis, proteinuria/hematuria, and focal segmental glomerular sclerosis (FSGS) resulting in chronic end-stage renal failure, successfully treated with renal transplantation. Microarray comparative genomic hybridization showed the derivative chromosome 6 to have a 6.4-Mb duplication at 6p25.3-p25.1 with 32 protein-coding genes and a 220-Kb deletion at 6p25.3 with two genes of no possible relation to the renal pathology. Review of the literature shows that variation of renal complications in the syndrome is compatible with congenital anomalies of the kidney and urinary tract (CAKUT). FSGS, observed in another patient with 6p duplication syndrome, could be a non-coincidental complication. FOXC1, located within the 6.4-Mb duplicated region at 6p25.3-p25.2, could be a candidate gene for CAKUT, but its single gene duplication effect would not be sufficient. FSGS would be a primary defect associated with duplicated gene(s) albeit no candidate could be proposed, or might occur in association with CAKUT.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/genética , Hidronefrosis/genética , Proteinuria/genética , Trisomía , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Biopsia , Niño , Bandeo Cromosómico , Cromosomas Humanos Par 6 , Hibridación Genómica Comparativa , Facies , Femenino , Estudio de Asociación del Genoma Completo , Glomeruloesclerosis Focal y Segmentaria/diagnóstico , Humanos , Hidronefrosis/diagnóstico , Riñón/anomalías , Riñón/patología , Proteinuria/diagnóstico , Síndrome , Ultrasonografía , Sistema Urinario/anomalías
9.
Am J Med Genet A ; 164A(2): 324-30, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24311518

RESUMEN

Trisomy 18 is a common chromosomal aberration syndrome involving growth impairment, various malformations, poor prognosis, and severe developmental delay in survivors. Although esophageal atresia (EA) with tracheoesophageal fistula (TEF) is a potentially fatal complication that can only be rescued through surgical correction, no reports have addressed the efficacy of surgical intervention for EA in patients with trisomy 18. We reviewed detailed clinical information of 24 patients with trisomy 18 and EA who were admitted to two neonatal intensive care units in Japan and underwent intensive treatment including surgical interventions from 1982 to 2009. Nine patients underwent only palliative surgery, including six who underwent only gastrostomy or both gastrostomy and jejunostomy (Group 1) and three who underwent gastrostomy and TEF division (Group 2). The other 15 patients underwent radical surgery, including 10 who underwent single-stage esophago-esophagostomy with TEF division (Group 3) and five who underwent two-stage operation (gastrostomy followed by esophago-esophagostomy with TEF division) (Group 4). No intraoperative death or anesthetic complications were noted. Enteral feeding was accomplished in 17 patients, three of whom were fed orally. Three patients could be discharged home. The 1-year survival rate was 17%: 27% in those receiving radical surgery (Groups 3 and 4); 0% in those receiving palliative surgery (Groups 1 and 2). Most causes of death were related to cardiac complications. EA is not an absolute poor prognostic factor in patients with trisomy 18 undergoing radical surgery for EA and intensive cardiac management.


Asunto(s)
Atresia Esofágica/etiología , Atresia Esofágica/cirugía , Trisomía , Causas de Muerte , Preescolar , Cromosomas Humanos Par 18 , Atresia Esofágica/mortalidad , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Mortalidad , Complicaciones Posoperatorias , Diagnóstico Prenatal , Pronóstico , Resultado del Tratamiento , Trisomía/diagnóstico , Síndrome de la Trisomía 18
10.
Am J Med Genet A ; 164A(5): 1272-6, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24664492

RESUMEN

Congenital cataracts are the most important cause of severe visual impairment in infants. Genetic factors contribute to the disease development and 29 genes are known to cause congenital cataracts. Identifying the genetic cause of congenital cataracts can be difficult because of genetic heterogeneity. V-maf avian musculoaponeurotic fibrosarcoma oncogene homolog (MAF) encodes a basic region/leucine zipper transcription factor that plays a key role as a regulator of embryonic lens fiber cell development. MAF mutations have been reported to cause juvenile-onset pulverulent cataract, microcornea, iris coloboma, and other anterior segment dysgenesis. We report on six patients in a family who have congenital cataracts were identified MAF mutation by whole exome sequencing (WES). The heterozygous MAF mutation Q303L detected in the present family occurs in a well conserved glutamine residue at the basic region of the DNA-binding domain. All affected members showed congenital cataracts. Three of the six members showed microcornea and one showed iris coloboma. Congenital cataracts with MAF mutation exhibited phenotypically variable cataracts within the family. Review of the patients with MAF mutations supports the notion that congenital cataracts caused by MAF mutations could be accompanied by microcornea and/or iris coloboma. WES is a useful tool for detecting disease-causing mutations in patients with genetically heterogeneous conditions.


Asunto(s)
Pueblo Asiatico/genética , Catarata/congénito , Catarata/genética , Mutación Missense , Proteínas Proto-Oncogénicas c-maf/genética , Alelos , Secuencia de Aminoácidos , Catarata/diagnóstico , Preescolar , Análisis Mutacional de ADN , Exoma , Femenino , Estudios de Asociación Genética , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Japón , Masculino , Datos de Secuencia Molecular , Linaje , Fenotipo , Proteínas Proto-Oncogénicas c-maf/química , Alineación de Secuencia
11.
Am J Med Genet A ; 164A(3): 597-609, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24357569

RESUMEN

Wolf-Hirschhorn syndrome (WHS) is a contiguous gene deletion syndrome of the distal 4p chromosome, characterized by craniofacial features, growth impairment, intellectual disability, and seizures. Although genotype-phenotype correlation studies have previously been published, several important issues remain to be elucidated including seizure severity. We present detailed clinical and molecular-cytogenetic findings from a microarray and fluorescence in situ hybridization (FISH)-based genotype-phenotype analysis of 22 Japanese WHS patients, the first large non-Western series. 4p deletions were terminal in 20 patients and interstitial in two, with deletion sizes ranging from 2.06 to 29.42 Mb. The new Wolf-Hirschhorn syndrome critical region (WHSCR2) was deleted in all cases, and duplication of other chromosomal regions occurred in four. Complex mosaicism was identified in two cases: two different 4p terminal deletions; a simple 4p terminal deletion and an unbalanced translocation with the same 4p breakpoint. Seizures began in infancy in 33% (2/6) of cases with small (<6 Mb) deletions and in 86% (12/14) of cases with larger deletions (>6 Mb). Status epilepticus occurred in 17% (1/6) with small deletions and in 87% (13/15) with larger deletions. Renal hypoplasia or dysplasia and structural ocular anomalies were more prevalent in those with larger deletions. A new susceptible region for seizure occurrence is suggested between 0.76 and 1.3 Mb from 4 pter, encompassing CTBP1 and CPLX1, and distal to the previously-supposed candidate gene LETM1. The usefulness of bromide therapy for seizures and additional clinical features including hypercholesterolemia are also described.


Asunto(s)
Estudios de Asociación Genética , Hibridación Fluorescente in Situ , Análisis de Secuencia por Matrices de Oligonucleótidos , Síndrome de Wolf-Hirschhorn/diagnóstico , Síndrome de Wolf-Hirschhorn/genética , Adolescente , Pueblo Asiatico/genética , Niño , Preescolar , Aberraciones Cromosómicas , Bandeo Cromosómico , Facies , Femenino , Humanos , Hibridación Fluorescente in Situ/métodos , Lactante , Japón , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Fenotipo
12.
J Hum Genet ; 58(8): 560-3, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23635952

RESUMEN

A follow-up nationwide survey on predictive genetic testing for late-onset neurological diseases in Japan was conducted. A questionnaire was sent to 89 institutional members of the Japan's National Liaison Council for Clinical Sections of Medical Genetics, and was returned by 60 (67.4%). A total of 301 clients with an interest in predictive testing were accumulated from April 2006 to March 2011. The greatest interest was shown for spinocerebellar degeneration (SCD, n=110), followed by myotonic dystrophy type 1 (DM1, n=69), Huntington's disease (HD, n=52) and familial amyloid polyneuropathy (FAP, n=35). The ratios of clients who actually underwent predictive testing were: SCD, 21.8%; DM1, 39.1%; HD, 26.9%; and FAP, 74.3%, indicating that predictive testing was conducted very cautiously for untreatable neurological diseases in Japan. Clinical geneticists were predominantly involved in genetic counseling, whereas the participation of non-medical doctor (non-MD) staff, including nurses, clinical psychologists and genetic counselors, was not common. Lack of non-MD counseling staff was one of the most serious issues in conducting predictive testing, which has not been improved since the previous survey performed in 2006. Institutional arrangements, such as revision of medical insurance system regarding genetic testing and counseling, might be necessary to resolve this issue.


Asunto(s)
Enfermedades Genéticas Congénitas/diagnóstico , Enfermedades Genéticas Congénitas/epidemiología , Pruebas Genéticas/estadística & datos numéricos , Encuestas Epidemiológicas/estadística & datos numéricos , Enfermedades del Sistema Nervioso/epidemiología , Enfermedades del Sistema Nervioso/genética , Edad de Inicio , Humanos , Japón/epidemiología , Enfermedades del Sistema Nervioso/diagnóstico , Encuestas y Cuestionarios
13.
Am J Med Genet A ; 161A(3): 518-26, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23401378

RESUMEN

It is debatable whether Hajdu-Cheney syndrome (HCS) and serpentine fibula-polycystic kidney syndrome (SFPKS) represent a single clinical entity with a variable degree of expression or two different entities, because both disorders share common clinical and radiological manifestations, including similar craniofacial characteristics, and defective bone mineralization. Since it was shown that heterozygous truncating mutations in NOTCH2 are responsible for both HCS and SFPKS, 37 patients with HCS and four patients with SFPKS are reported. To elucidate the clinical consequences of NOTCH2 mutations, we present detailed clinical information for seven patients with truncating mutations in exon 34 of NOTCH2, six with HCS and one with SFPKS. In addition, we review all the reported patients whose clinical manifestations are available. We found 13 manifestations including craniofacial features, acroosteolysis, Wormian bones, and osteoporosis in >75% of NOTCH2-positive patients. Acroosteolysis was observed in two patients with SFPKS and bowing fibulae were found in two patients with HCS. These clinical and molecular data would support the notion that HCS and SFPKS are a single disorder.


Asunto(s)
Síndrome de Hajdu-Cheney/diagnóstico por imagen , Receptor Notch2/genética , Adolescente , Adulto , Preescolar , Codón sin Sentido , Análisis Mutacional de ADN , Exones , Estudios de Asociación Genética , Síndrome de Hajdu-Cheney/genética , Humanos , Persona de Mediana Edad , Radiografía , Adulto Joven
14.
Am J Med Genet A ; 161A(6): 1221-37, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23637025

RESUMEN

Mutations in the components of the SWItch/sucrose nonfermentable (SWI/SNF)-like chromatin remodeling complex have recently been reported to cause Coffin-Siris syndrome (CSS), Nicolaides-Baraitser syndrome (NCBRS), and ARID1B-related intellectual disability (ID) syndrome. We detail here the genotype-phenotype correlations for 85 previously published and one additional patient with mutations in the SWI/SNF complex: four with SMARCB1 mutations, seven with SMARCA4 mutations, 37 with SMARCA2 mutations, one with an SMARCE1 mutation, three with ARID1A mutations, and 33 with ARID1B mutations. The mutations were associated with syndromic ID and speech impairment (severe/profound in SMARCB1, SMARCE1, and ARID1A mutations; variable in SMARCA4, SMARCA2, and ARID1B mutations), which was frequently accompanied by agenesis or hypoplasia of the corpus callosum. SMARCB1 mutations caused "classical" CSS with typical facial "coarseness" and significant digital/nail hypoplasia. SMARCA4 mutations caused CSS without typical facial coarseness and with significant digital/nail hypoplasia. SMARCA2 mutations caused NCBRS, typically with short stature, sparse hair, a thin vermillion of the upper lip, an everted lower lip and prominent finger joints. A SMARCE1 mutation caused CSS without typical facial coarseness and with significant digital/nail hypoplasia. ARID1A mutations caused the most severe CSS with severe physical complications. ARID1B mutations caused CSS without typical facial coarseness and with mild digital/nail hypoplasia, or caused syndromic ID. Because of the common underlying mechanism and overlapping clinical features, we propose that these conditions be referred to collectively as "SWI/SNF-related ID syndromes".


Asunto(s)
Anomalías Múltiples/genética , Ensamble y Desensamble de Cromatina/genética , Cara/anomalías , Deformidades Congénitas del Pie/genética , Deformidades Congénitas de la Mano/genética , Hipotricosis/genética , Discapacidad Intelectual/genética , Micrognatismo/genética , Cuello/anomalías , Factores de Transcripción/genética , Proteínas Cromosómicas no Histona/genética , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Facies , Femenino , Estudios de Asociación Genética , Humanos , Masculino , Mutación , Proteínas Nucleares/genética , Proteína SMARCB1 , Síndrome
15.
Chromosome Res ; 20(6): 659-72, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22801776

RESUMEN

The three-dimensional (3D) structure of the genome is organized non-randomly and plays a role in genomic function via epigenetic mechanisms in the eukaryotic nucleus. Here, we analyzed the spatial positioning of three target regions; the SNRPN, UBE3A, and GABRB3 genes on human chromosome 15q11.2-q12, a representative cluster of imprinted regions, in the interphase nuclei of B lymphoblastoid cell lines, peripheral blood cells, and skin fibroblasts derived from normal individuals to look for evidence of genomic organization and function. The positions of these genes were simultaneously visualized, and all inter-gene distances were calculated for each homologous chromosome in each nucleus after three-color 3D fluorescence in situ hybridization. None of the target genes were arranged linearly in most cells analyzed, and GABRB3 was positioned closer to SNRPN than UBE3A in a high proportion of cells in all cell types. This was in contrast to the genomic map in which GABRB3 was positioned closer to UBE3A than SNRPN. We compared the distances from SNRPN to UBE3A (SU) and from UBE3A to GABRB3 (UG) between alleles in each nucleus, 50 cells per subject. The results revealed that the gene-to-gene distance of one allele was longer than that of the other and that the SU ratio (longer/shorter SU distance between alleles) was larger than the UG ratio (longer/shorter UG distance between alleles). The UG distance was relatively stable between alleles; in contrast, the SU distance of one allele was obviously longer than the distance indicated by the genome size. The results therefore indicate that SNRPN, UBE3A, and GABRB3 have non-linear and non-random curved spatial positioning in the normal nucleus, with differences in the SU distance between alleles possibly representing epigenetic evidence of nuclear organization and gene expression.


Asunto(s)
Núcleo Celular/ultraestructura , Cromosomas Humanos Par 15/ultraestructura , Imagenología Tridimensional/métodos , Hibridación Fluorescente in Situ/métodos , Receptores de GABA-A/ultraestructura , Ubiquitina-Proteína Ligasas/ultraestructura , Proteínas Nucleares snRNP/ultraestructura , Linfocitos B , Mapeo Cromosómico , Cromosomas Humanos Par 15/genética , Femenino , Fibroblastos , Humanos , Masculino , Microscopía Confocal , Receptores de GABA-A/genética , Ubiquitina-Proteína Ligasas/genética , Proteínas Nucleares snRNP/genética
16.
Nat Genet ; 30(4): 365-6, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11896389

RESUMEN

We isolated NSD1 from the 5q35 breakpoint in an individual with Sotos syndrome harboring a chromosomal translocation. We identified 1 nonsense, 3 frameshift and 20 submicroscopic deletion mutations of NSD1 among 42 individuals with sporadic cases of Sotos syndrome. The results indicate that haploinsufficiency of NSD1 is the major cause of Sotos syndrome.


Asunto(s)
Acromegalia/genética , Proteínas Portadoras/genética , Péptidos y Proteínas de Señalización Intracelular , Proteínas Nucleares/genética , Secuencia de Bases , Mapeo Cromosómico , Cromosomas Humanos Par 5 , Clonación Molecular , Codón sin Sentido , Cósmidos , ADN Complementario/metabolismo , Exones , Huesos Faciales/anomalías , Mutación del Sistema de Lectura , Eliminación de Gen , Gigantismo/genética , Trastornos del Crecimiento/genética , Heterocigoto , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina , Humanos , Hibridación Fluorescente in Situ , Modelos Genéticos , Datos de Secuencia Molecular , Homología de Secuencia de Ácido Nucleico , Síndrome , Translocación Genética
17.
Circ Rep ; 5(6): 252-259, 2023 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-37305793

RESUMEN

Background: Definitions of cardiac sarcoidosis (CS) differ among guidelines. Any systemic histological finding of CS is essential for the diagnosis of CS in the 2014 Heart Rhythm Society statement, but not necessary in the Japanese Circulation Society 2016 guidelines. This study aimed to reveal the differences in outcomes by comparing 2 groups, namely CS patients with or without systemic histologically proven granuloma. Methods and Results: This study retrospectively included 231 consecutive patients with CS. CS with granulomas in ≥1 organs was diagnosed in 131 patients (Group G), whereas CS without any granulomas was diagnosed in the remaining 100 patients (Group NG). Left ventricular ejection fraction (LVEF) was significantly reduced in Group NG compared with Group G (44±13% vs. 50±16%, respectively; P=0.001). However, Kaplan-Meier curves showed that major adverse cardiovascular events (MACE)-free survival outcomes were comparable between the 2 groups (log-rank P=0.167). Univariable analyses showed that significant predictors of MACE were Groups G/NG, histological CS, LVEF, and high B-type natriuretic peptide (BNP) or N-terminal pro BNP concentrations, but none of these was significant in multivariable analyses. Conclusions: Overall risks of MACE were similar between the 2 groups despite different manifestations in cardiac dysfunction. The data not only validate the prognostic value of non-invasive diagnosis of CS, but also show the need for careful observation and therapeutic strategy in patients with CS without any granuloma.

18.
Hum Mol Genet ; 19(14): 2898-906, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20423928

RESUMEN

Kawasaki disease (KD; OMIM 611775) is an acute vasculitis syndrome which predominantly affects small- and medium-sized arteries of infants and children. Epidemiological data suggest that host genetics underlie the disease pathogenesis. Here we report that multiple variants in the caspase-3 gene (CASP3) that are in linkage disequilibrium confer susceptibility to KD in both Japanese and US subjects of European ancestry. We found that a G to A substitution of one commonly associated SNP located in the 5' untranslated region of CASP3 (rs72689236; P = 4.2 x 10(-8) in the Japanese and P = 3.7 x 10(-3) in the European Americans) abolished binding of nuclear factor of activated T cells to the DNA sequence surrounding the SNP. Our findings suggest that altered CASP3 expression in immune effecter cells influences susceptibility to KD.


Asunto(s)
Caspasa 3/genética , Predisposición Genética a la Enfermedad , Síndrome Mucocutáneo Linfonodular/genética , Polimorfismo de Nucleótido Simple , Adulto , Pueblo Asiatico/genética , Sitios de Unión/genética , Estudios de Casos y Controles , Caspasa 3/metabolismo , Caspasa 3/fisiología , Niño , Preescolar , Femenino , Frecuencia de los Genes , Pruebas Genéticas , Humanos , Lactante , Desequilibrio de Ligamiento , Masculino , Factores de Transcripción NFATC/metabolismo , Polimorfismo de Nucleótido Simple/fisiología , Unión Proteica , Población Blanca/genética
19.
Am J Med Genet A ; 158A(4): 861-8, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22407754

RESUMEN

2q23.1 microdeletion syndrome is a recently characterized chromosomal aberration disorder uncovered through array comparative genomic hybridization (array CGH). Although the cardinal feature is intellectual disability (ID), neurodevelopmental features of the syndrome have not been systematically reviewed. We present a 5-year-old boy with severe psychomotor developmental delay/ID, progressive microcephaly with brain atrophy, growth retardation, and several external anomalies. He manifested intractable epilepsy, effectively treated with combined antiepileptic drug therapy including topiramate. Array CGH demonstrated a de novo interstitial deletion of approximately 1 Mb at 2q23.1-q23.2, involving four genes including MBD5. Nineteen patients have been reported to have the syndrome, including present patient. All patients whose data were available had ID, 17 patients (89%) had seizures, and microcephaly was evident in 9 of 18 patients (50%). Deletion sizes ranged from 200 kb to 5.5 Mb, comprising 1-15 genes. MBD5, the only gene deleted in all patients, is considered to be responsible for ID and epilepsy. Furthermore, the deletion junction was sequenced for the first time in a patient with the syndrome; and homology of three nucleotides, identified at the distal and proximal breakpoints, suggested that the deletion might have been mediated by recently-delineated genomic rearrangement mechanism Fork Stalling and Template Switching (FoSTeS)/microhomology-mediated break-induced replication (MMBIR).


Asunto(s)
Cromosomas Humanos Par 2/genética , Proteínas de Unión al ADN/genética , Convulsiones/genética , Eliminación de Secuencia , Anomalías Múltiples/genética , Preescolar , Hibridación Genómica Comparativa , Discapacidades del Desarrollo/genética , Fructosa/análogos & derivados , Fructosa/uso terapéutico , Humanos , Discapacidad Intelectual/genética , Masculino , Microcefalia/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Convulsiones/tratamiento farmacológico , Topiramato
20.
Am J Med Genet A ; 158A(2): 412-6, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22140075

RESUMEN

15q24 deletion syndrome is a recently-described chromosomal disorder, characterized by developmental delay, growth deficiency, distinct facial features, digital abnormalities, loose connective tissue, and genital malformations in males. To date, 19 patients have been reported. We report on a 13-year-old boy with this syndrome manifesting childhood myelodysplastic syndrome (MDS). He had characteristic facial features, hypospadias, and mild developmental delay. He showed neutropenia and thrombocytopenia for several years. At age 13 years, bone marrow examination was performed, which showed a sign suggestive of childhood MDS: mild dysplasia in the myeloid, erythroid, and megakaryocytic cell lineages. Array comparative genomic hybridization (array CGH) revealed a de novo 3.4 Mb 15q24.1q24.3 deletion. Although MDS has not been described in patients with the syndrome, a boy was reported to have acute lymphoblastic leukemia (ALL). The development of MDS and hematological malignancy in the syndrome might be caused by the haploinsufficiency of deleted 15q24 segment either alone or in combination with other genetic abnormalities in hematopoietic cells. Further hematological investigation is recommended to be beneficial if physical and hematological examination results are suggestive of hematopoietic disturbance in patients with the syndrome.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 15 , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Adolescente , Hibridación Genómica Comparativa , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Humanos , Hipospadias/diagnóstico , Hipospadias/patología , Hibridación Fluorescente in Situ , Linfocitos/citología , Masculino , Síndromes Mielodisplásicos/diagnóstico , Neutropenia/diagnóstico , Neutropenia/patología , Trombocitopenia/diagnóstico , Trombocitopenia/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA