Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Clin Endocrinol (Oxf) ; 97(1): 72-80, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35419873

RESUMEN

OBJECTIVE: We recently reported cases of adipsic hypernatremia caused by autoantibodies against the subfornical organ in patients with hypothalamic-pituitary lesions. This study aimed to clarify the clinical features of newly identified patients with adipsic hypernatremia whose sera displayed immunoreactivity to the mouse subfornical organ. DESIGN: Observational cohort study of patients diagnosed with adipsic hypernatremia in Japan, United States, and Europe. METHODS: The study included 22 patients with adipsic hypernatremia but without overt structural changes in the hypothalamic-pituitary region and congenital disease. Antibody response to the mouse subfornical organ was determined using immunohistochemistry. The clinical characteristics were compared between the patients with positive and negative antibody responses. RESULTS: Antibody response to the mouse subfornical organ was detected in the sera of 16 patients (72.7%, female/male ratio, 1:1, 12 pediatric and 4 adult patients). The prolactin levels at the time of diagnosis were significantly higher in patients with positive subfornical organ (SFO) immunoreactivity than in those with negative SFO immunoreactivity (58.9 ± 33.5 vs. 22.9 ± 13.9 ng/ml, p < .05). Hypothalamic disorders were found in 37.5% of the patients with positive SFO immunoreactivity. Moreover, six patients were diagnosed with rapid-onset obesity with hypothalamic dysfunction, hypoventilation, and autonomic dysregulation/neural tumor syndrome after the diagnosis of adipsic hypernatremia. Plasma renin activity levels were significantly higher in patients with serum immunoreactivity to the Nax channel. CONCLUSIONS: The patients with serum immunoreactivity to the SFO had higher prolactin levels and hypothalamic disorders compared to those without the immunoreactivity. The clinical characteristics of patients with serum immunoreactivity to the subfornical organ included higher prolactin levels and hypothalamic disorders, which were frequently associated with central hypothyroidism and the presence of retroperitoneal tumors.


Asunto(s)
Hipernatremia , Enfermedades Hipotalámicas , Órgano Subfornical , Animales , Niño , Femenino , Humanos , Hipotálamo , Inmunidad , Masculino , Ratones , Prolactina , Órgano Subfornical/fisiología
2.
Kidney Int ; 98(5): 1308-1322, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32554042

RESUMEN

To understand the genetics of steroid-sensitive nephrotic syndrome (SSNS), we conducted a genome-wide association study in 987 childhood SSNS patients and 3,206 healthy controls with Japanese ancestry. Beyond known associations in the HLA-DR/DQ region, common variants in NPHS1-KIRREL2 (rs56117924, P=4.94E-20, odds ratio (OR) =1.90) and TNFSF15 (rs6478109, P=2.54E-8, OR=0.72) regions achieved genome-wide significance and were replicated in Korean, South Asian and African populations. Trans-ethnic meta-analyses including Japanese, Korean, South Asian, African, European, Hispanic and Maghrebian populations confirmed the significant associations of variants in NPHS1-KIRREL2 (Pmeta=6.71E-28, OR=1.88) and TNFSF15 (Pmeta=5.40E-11, OR=1.33) loci. Analysis of the NPHS1 risk alleles with glomerular NPHS1 mRNA expression from the same person revealed allele specific expression with significantly lower expression of the transcript derived from the risk haplotype (Wilcox test p=9.3E-4). Because rare pathogenic variants in NPHS1 cause congenital nephrotic syndrome of the Finnish type (CNSF), the present study provides further evidence that variation along the allele frequency spectrum in the same gene can cause or contribute to both a rare monogenic disease (CNSF) and a more complex, polygenic disease (SSNS).


Asunto(s)
Síndrome Nefrótico , Alelos , Niño , Estudio de Asociación del Genoma Completo , Haplotipos , Humanos , Proteínas de la Membrana , Mutación , Síndrome Nefrótico/tratamiento farmacológico , Síndrome Nefrótico/genética , Esteroides , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
3.
Clin Exp Nephrol ; 23(2): 158-168, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30128941

RESUMEN

Alport syndrome (AS) is a progressive hereditary renal disease that is characterized by sensorineural hearing loss and ocular abnormalities. It is divided into three modes of inheritance, namely, X-linked Alport syndrome (XLAS), autosomal recessive AS (ARAS), and autosomal dominant AS (ADAS). XLAS is caused by pathogenic variants in COL4A5, while ADAS and ARAS are caused by those in COL4A3/COL4A4. Diagnosis is conventionally made pathologically, but recent advances in comprehensive genetic analysis have enabled genetic testing to be performed for the diagnosis of AS as first-line diagnosis. Because of these advances, substantial information about the genetics of AS has been obtained and the genetic background of this disease has been revealed, including genotype-phenotype correlations and mechanisms of onset in some male XLAS cases that lead to milder phenotypes of late-onset end-stage renal disease (ESRD). There is currently no radical therapy for AS and treatment is only performed to delay progression to ESRD using nephron-protective drugs. Angiotensin-converting enzyme inhibitors can remarkably delay the development of ESRD. Recently, some new drugs for this disease have entered clinical trials or been developed in laboratories. In this article, we review the diagnostic strategy, genotype-phenotype correlation, mechanisms of onset of milder phenotypes, and treatment of AS, among others.


Asunto(s)
Autoantígenos/genética , Colágeno Tipo IV/genética , Mutación , Nefritis Hereditaria/genética , Adulto , Animales , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Herencia , Humanos , Riñón/química , Riñón/patología , Masculino , Nefritis Hereditaria/diagnóstico , Nefritis Hereditaria/terapia , Fenotipo , Pronóstico , Factores de Riesgo , Adulto Joven
4.
J Am Soc Nephrol ; 29(8): 2189-2199, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30012571

RESUMEN

Background Nephrotic syndrome is the most common cause of chronic glomerular disease in children. Most of these patients develop steroid-sensitive nephrotic syndrome (SSNS), but the loci conferring susceptibility to childhood SSNS are mainly unknown.Methods We conducted a genome-wide association study (GWAS) in the Japanese population; 224 patients with childhood SSNS and 419 adult healthy controls were genotyped using the Affymetrix Japonica Array in the discovery stage. Imputation for six HLA genes (HLA-A, -C, -B, -DRB1, -DQB1, and -DPB1) was conducted on the basis of Japanese-specific references. We performed genotyping for HLA-DRB1/-DQB1 using a sequence-specific oligonucleotide-probing method on a Luminex platform. Whole-genome imputation was conducted using a phased reference panel of 2049 healthy Japanese individuals. Replication was performed in an independent Japanese sample set including 216 patients and 719 healthy controls. We genotyped candidate single-nucleotide polymorphisms using the DigiTag2 assay.Results The most significant association was detected in the HLA-DR/DQ region and replicated (rs4642516 [minor allele G], combined Pallelic=7.84×10-23; odds ratio [OR], 0.33; 95% confidence interval [95% CI], 0.26 to 0.41; rs3134996 [minor allele A], combined Pallelic=1.72×10-25; OR, 0.29; 95% CI, 0.23 to 0.37). HLA-DRB1*08:02 (Pc=1.82×10-9; OR, 2.62; 95% CI, 1.94 to 3.54) and HLA-DQB1*06:04 (Pc=2.09×10-12; OR, 0.10; 95% CI, 0.05 to 0.21) were considered primary HLA alleles associated with childhood SSNS. HLA-DRB1*08:02-DQB1*03:02 (Pc=7.01×10-11; OR, 3.60; 95% CI, 2.46 to 5.29) was identified as the most significant genetic susceptibility factor.Conclusions The most significant association with childhood SSNS was detected in the HLA-DR/DQ region. Further HLA allele/haplotype analyses should enhance our understanding of molecular mechanisms underlying SSNS.


Asunto(s)
Predisposición Genética a la Enfermedad , Antígenos HLA-DQ/genética , Cadenas beta de HLA-DQ/genética , Cadenas HLA-DRB1/genética , Síndrome Nefrótico/genética , Adulto , Estudios de Casos y Controles , Niño , Femenino , Estudio de Asociación del Genoma Completo , Cadenas beta de HLA-DQ/inmunología , Haplotipos , Humanos , Japón , Masculino , Síndrome Nefrótico/tratamiento farmacológico , Síndrome Nefrótico/inmunología , Polimorfismo de Nucleótido Simple , Valores de Referencia , Esteroides/uso terapéutico
5.
J Am Soc Nephrol ; 29(8): 2244-2254, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29959198

RESUMEN

BACKGROUND: X-linked Alport syndrome (XLAS) is a progressive hereditary nephropathy caused by mutations in the COL4A5 gene. Genotype-phenotype correlation in male XLAS is relatively well established; relative to truncating mutations, nontruncating mutations exhibit milder phenotypes. However, transcript comparison between XLAS cases with splicing abnormalities that result in a premature stop codon and those with nontruncating splicing abnormalities has not been reported, mainly because transcript analysis is not routinely conducted in patients with XLAS. METHODS: We examined transcript expression for all patients with suspected splicing abnormalities who were treated at one hospital between January of 2006 and July of 2017. Additionally, we recruited 46 males from 29 families with splicing abnormalities to examine genotype-phenotype correlation in patients with truncating (n=21, from 14 families) and nontruncating (n=25, from 15 families) mutations at the transcript level. RESULTS: We detected 41 XLAS families with abnormal splicing patterns and described novel XLAS atypical splicing patterns (n=14) other than exon skipping caused by point mutations in the splice consensus sequence. The median age for developing ESRD was 20 years (95% confidence interval, 14 to 23 years) among patients with truncating mutations and 29 years (95% confidence interval, 25 to 40 years) among patients with nontruncating mutations (P=0.001). CONCLUSIONS: We report unpredictable atypical splicing in the COL4A5 gene in male patients with XLAS and reveal that renal prognosis differs significantly for patients with truncating versus nontruncating splicing abnormalities. Our results suggest that splicing modulation should be explored as a therapy for XLAS with truncating mutations.


Asunto(s)
Colágeno Tipo IV/genética , Estudios de Asociación Genética/métodos , Predisposición Genética a la Enfermedad/epidemiología , Nefritis Hereditaria/genética , Mutación Puntual/genética , Adulto , Estudios de Cohortes , Análisis Mutacional de ADN , Humanos , Japón , Masculino , Nefritis Hereditaria/diagnóstico , Nefritis Hereditaria/epidemiología , Linaje , Estudios Retrospectivos
6.
J Hum Genet ; 63(6): 755-759, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29556035

RESUMEN

Pathogenic variants in specific complement-related genes lead to atypical hemolytic uremic syndrome (aHUS). Some reports have indicated that patients with digenic variants in these genes might present severer phenotypes. Upon detecting novel intronic variants, transcriptional analysis is necessary to prove pathogenicity; however, when intronic variants are located in intron 1 and, as a result, no transcript is produced, no appropriate method had been established to reveal the pathogenicity. Recently, the minigene assay was used to assess the pathogenicity of intronic variants. Here, we report an infantile case of aHUS caused by digenic mutations in two different complement-related genes, C3 and MCP. Targeted sequencing detected a known variant in C3 and a novel variant in the intron 1 splicing donor site of MCP. To assess the pathogenicity of this intronic variant, we conducted functional splicing assay using a minigene construct and quantitative PCR analysis of the MCP transcript, revealing the pathogenicity of the intronic variant. In conclusion, the minigene assay revealed the pathogenicity of the intron 1 splicing donor site variant for the first time. This case showed a severe phenotype of infantile-onset aHUS associated with digenic variants in two complement-related genes.


Asunto(s)
Complemento C3/genética , Síndrome Hemolítico-Urémico/genética , Proteína Cofactora de Membrana/genética , Mutación , Empalme del ARN , Edad de Inicio , Humanos , Lactante , Intrones , Masculino , Fenotipo , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética
7.
Clin Exp Nephrol ; 22(4): 924-930, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29352455

RESUMEN

BACKGROUND: Thrombotic microangiopathy (TMA) includes hemolytic uremic syndrome (HUS) and thrombotic thrombocytopenic purpura (TTP). This study examined the epidemiological characteristics of pediatric patients with TMA classified according to etiology. METHODS: The survey evaluated 258 Japanese pediatric patients diagnosed with TMA between 2012 and 2015. RESULTS: The primary diseases responsible for TMA were categorized as TTP (15 cases), Shiga toxin-producing Escherichia coli-associated HUS (STEC-HUS) (166 cases), atypical HUS (aHUS) (40 cases), and secondary TMA (27 cases). The remaining 10 cases were unable to be classified to one of the four categories of the primary disease. Renal replacement therapy was required in the acute phase in 103 patients with TMA, including 65 with STEC-HUS, 22 with aHUS, two with TTP, 10 with secondary TMA, and four unclassified cases. The last observational findings were normal renal function in 95 patients and chronic kidney disease (CKD) stage 1 in 62. For 31 patients, chronic renal insufficiency (CKD stage 2-5) persisted, including four patients with end-stage kidney disease (CKD stage 5). Seventeen patients suffered recurrence of TMA, and eight patients died. CONCLUSION: This study clarified differences in the relative proportions of primary diseases between patients from Japan and North America and Europe. The difference may be attributable to the lower estimated incidence of STEC-HUS in Japan.


Asunto(s)
Microangiopatías Trombóticas/diagnóstico , Niño , Preescolar , Estudios Transversales , Europa (Continente) , Femenino , Humanos , Japón , Masculino , América del Norte , Microangiopatías Trombóticas/complicaciones , Microangiopatías Trombóticas/patología
8.
BMC Nephrol ; 18(1): 353, 2017 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-29202719

RESUMEN

BACKGROUND: Autosomal recessive distal renal tubular acidosis (dRTA) is a rare hereditary disease caused by pathogenic variants in the ATP6V0A4 gene or ATP6V1B1 gene, and characterized by hyperchloremic metabolic acidosis with normal anion gap, hypokalemia, hypercalciuria, hypocitraturia and nephrocalcinosis. Although several intronic nucleotide variants in these genes have been detected, all of them fell in the apparent splice consensus sequence. In general, transcriptional analysis is necessary to determine the effect on function of the novel intronic variants located out of splicing consensus sequences. In recent years, functional splicing analysis using minigene construction was used to assess the pathogenicity of novel intoronic variant in various field. METHODS: We investigated a sporadic case of dRTA with a compound heterozygous mutation in the ATP6V0A4 gene, revealed by next generation sequencing. One variant was already reported as pathogenic; however, the other was a novel variant in intron 11 (c.1029 + 5G > A) falling outside of the apparent splicing consensus sequence. Expression of ATP6V0A4 was not detected in peripheral leukocytes by RT-PCR analysis. Therefore, an in vitro functional splicing study using minigene construction was conducted to analyze the splicing pattern of the novel variant. RESULTS: A minigene assay revealed that the novel intronic variant leads to a 104 bp insertion immediately following exon 11. In addition, this result was confirmed using RNA extracted from the patient's cultured leukocytes. CONCLUSION: These results proved the pathogenicity of a novel intronic variant in our patient. We concluded that the minigene assay is a useful, non-invasive method for functional splicing analysis of inherited kidney disease, even if standard transcriptional analysis could not detect abnormal mRNA.


Asunto(s)
Acidosis Tubular Renal/diagnóstico , Acidosis Tubular Renal/genética , Variación Genética/genética , Intrones/genética , Empalme de Proteína/genética , ATPasas de Translocación de Protón Vacuolares/genética , Secuencia de Bases , Humanos , Lactante , Masculino
9.
Clin Exp Nephrol ; 20(4): 536-543, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27422619

RESUMEN

Atypical hemolytic uremic syndrome (aHUS) is a rare disease characterized by the triad of microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury. In 2013, we developed diagnostic criteria to enable early diagnosis and timely initiation of appropriate treatment for aHUS. Recent clinical and molecular findings have resulted in several proposed classifications and definitions of thrombotic microangiopathy and aHUS. Based on recent advances in this field and the emerging international consensus to exclude secondary TMAs from the definition of aHUS, we have redefined aHUS and proposed diagnostic algorithms, differential diagnosis, and therapeutic strategies for aHUS.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/diagnóstico , Síndrome Hemolítico Urémico Atípico/etiología , Síndrome Hemolítico Urémico Atípico/terapia , Humanos , Japón
10.
Pediatr Int ; 58(7): 549-55, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27460397

RESUMEN

Atypical hemolytic uremic syndrome (aHUS) is a rare disease characterized by the triad of microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury. In 2013, we developed diagnostic criteria to enable early diagnosis and timely initiation of appropriate treatment for aHUS. Recent clinical and molecular findings have resulted in several proposed classifications and definitions of thrombotic microangiopathy and aHUS. Based on recent advances in this field and the emerging international consensus to exclude secondary TMAs from the definition of aHUS, we have redefined aHUS and proposed diagnostic algorithms, differential diagnosis, and therapeutic strategies for aHUS.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/diagnóstico , Diagnóstico Precoz , Guías de Práctica Clínica como Asunto , Síndrome Hemolítico Urémico Atípico/epidemiología , Humanos , Incidencia , Japón/epidemiología
12.
Pediatr Diabetes ; 15(8): 606-10, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24552466

RESUMEN

The most common form of transient neonatal diabetes mellitus (TNDM) is 6q24-related TNDM. Patients are treated with insulin during the neonatal period until spontaneous remission. However, diabetes often recurs in adolescence, and there is no standard therapy for patients with a relapse. A paternal duplication at the 6q24 critical region spanning the pleiomorphic adenoma gene-like 1 PLAGL1 gene was found in a Japanese patient with TNDM relapse. The patient was treated with a dipeptidyl peptidase-4 (DPP4) inhibitor, alogliptin, at a dose of 25 mg per day. Immediately after treatment initiation, his hemoglobin A1c (HbA1c) levels dropped from 7.0-7.5% (52-58 mmol/mol) to 6.0-6.5% (41-47 mmol/mol) and remained stable for over a year. We reported the successful treatment of relapsed 6q24-related TNDM with a DPP4 inhibitor. Although insulin has been the conventional treatment for such patients, treatments targeting the GLP1 pathway can be a useful alternative because these patients retain the ß cell mass and responsiveness through G protein-coupled pathways.


Asunto(s)
Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Hipoglucemiantes/uso terapéutico , Diabetes Mellitus/tratamiento farmacológico , Humanos , Masculino , Piperidinas/uso terapéutico , Recurrencia , Resultado del Tratamiento , Uracilo/análogos & derivados , Uracilo/uso terapéutico , Adulto Joven
13.
Clin Exp Nephrol ; 18(1): 4-9, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24343712

RESUMEN

Atypical hemolytic uremic syndrome (aHUS) is rare and comprises the triad of microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury. Recently, abnormalities in the mechanisms underlying complement regulation have been focused upon as causes of aHUS. The prognosis for patients who present with aHUS is very poor, with the first aHUS attack being associated with a mortality rate of ~25 %, and with ~50 % of cases resulting in end-stage renal disease requiring dialysis. If treatment is delayed, there is a high risk of this syndrome progressing to renal failure. Therefore, we have developed diagnostic criteria for aHUS to enable its early diagnosis and to facilitate the timely initiation of appropriate treatment. We hope these diagnostic criteria will be disseminated to as many clinicians as possible and that they will be used widely.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/diagnóstico , Nefrología/normas , Pediatría/normas , Síndrome Hemolítico Urémico Atípico/sangre , Síndrome Hemolítico Urémico Atípico/etiología , Síndrome Hemolítico Urémico Atípico/inmunología , Síndrome Hemolítico Urémico Atípico/terapia , Biomarcadores/sangre , Activación de Complemento , Proteínas del Sistema Complemento , Consenso , Diagnóstico Precoz , Humanos , Recuento de Plaquetas/normas , Valor Predictivo de las Pruebas , Pronóstico , Factores de Riesgo
14.
Pediatr Int ; 56(1): 1-5, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24548192

RESUMEN

Atypical hemolytic uremic syndrome (aHUS) is rare and comprises the triad of microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury. Recently, abnormalities in the mechanisms underlying complement regulation have been focused upon as causes of aHUS. The prognosis for patients who present with aHUS is very poor, with the first aHUS attack being associated with a mortality rate of approximately 25%, and with approximately 50% of cases resulting in end-stage renal disease requiring dialysis. If treatment is delayed, there is a high risk of this syndrome progressing to renal failure. Therefore, we have developed diagnostic criteria for aHUS to enable its early diagnosis and to facilitate the timely initiation of appropriate treatment. We hope these diagnostic criteria will be disseminated to as many clinicians as possible and that they will be used widely.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/diagnóstico , Técnicas de Diagnóstico Urológico/normas , Guías como Asunto , Nefrología , Sociedades Médicas , Niño , Humanos , Japón
15.
Clin Endocrinol (Oxf) ; 78(6): 891-7, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23067144

RESUMEN

OBJECTIVE: To evaluate the efficacy of long-term, continuous, subcutaneous octreotide infusion for congenital hyperinsulinism caused by mutations in the KATP-channel genes, KCNJ11 and ABCC8. PATIENTS: Fifteen Japanese patients with diazoxide-unresponsive, KATP-channel hyperinsulinism. METHODS: Molecular diagnoses were made by sequencing and multiple ligation-dependent probe amplification analysis. In patients with paternally inherited, monoallelic mutations, 18F-DOPA PET scans were performed to determine the location of the lesion. The patients were treated with continuous, subcutaneous octreotide infusion at a dosage of up to 25 µg/kg/day, using an insulin pump to maintain blood glucose levels higher than 3.33 mmol/l. Additional treatments (IV glucose, glucagon or enteral feeding) were administered as needed. The efficacy of the treatment was assessed in patients who received octreotide for 4 months to 5.9 years. RESULTS: Three patients had biallelic mutations, and 12 had monoallelic, paternally inherited mutations. Four patients with monoallelic mutations showed diffuse 18F-DOPA uptake, whereas seven patients showed focal uptake. Octreotide was effective in all the patients. The patients with biallelic mutations required a higher dosage (17-25 µg/kg/day), and two patients required additional treatments. By contrast, the patients with monoallelic mutations required a lower dosage (0.5-21 µg/kg/day) irrespective of the PET results and mostly without additional treatments. Treatment was discontinued in three patients at 2.5, 3.3 and 5.9 years of age, without psychomotor delay. Except for growth deceleration at a higher dosage, no significant adverse effects were noted. CONCLUSIONS: Long-term, continuous, subcutaneous octreotide infusion is a feasible alternative to surgery especially for patients with monoallelic KATP-channel mutations.


Asunto(s)
Hiperinsulinismo Congénito/tratamiento farmacológico , Canales KATP/genética , Octreótido/administración & dosificación , Transportadoras de Casetes de Unión a ATP/genética , Desarrollo Infantil/efectos de los fármacos , Preescolar , Hiperinsulinismo Congénito/diagnóstico , Dihidroxifenilalanina/análogos & derivados , Femenino , Radioisótopos de Flúor , Humanos , Lactante , Recién Nacido , Infusiones Subcutáneas , Masculino , Octreótido/efectos adversos , Tomografía de Emisión de Positrones , Canales de Potasio de Rectificación Interna/genética , Receptores de Droga/genética , Receptores de Sulfonilureas
16.
J Med Genet ; 49(10): 642-3, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22962692

RESUMEN

GATA6 haploinsufficiency has recently been reported as the most frequent cause of neonatal diabetes with pancreatic agenesis. Although all previously reported cases represented a de novo mutation with complete agenesis or pronounced hypoplasia of the pancreas, in this study we identified a family with a dominantly inherited mutation. Unlike previously reported cases, the degree of pancreatic hypoplasia and the severity of diabetes varied among members of the family, ranging from neonatally lethal diabetes with only a remnant of pancreatic tissue to adult-onset diabetes associated with dorsal agenesis of the pancreas. These observations further broaden the clinical spectrum of diabetes associated with GATA6 haploinsufficiency.


Asunto(s)
Diabetes Mellitus/genética , Factor de Transcripción GATA6/genética , Genes Dominantes , Haploinsuficiencia , Adolescente , Adulto , Secuencia de Bases , Niño , Preescolar , Diabetes Mellitus/diagnóstico , Exones , Resultado Fatal , Femenino , Genotipo , Humanos , Masculino , Mutación , Linaje , Fenotipo , Adulto Joven
17.
Pediatr Int ; 55(4): e100-2, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23910809

RESUMEN

Pheochromocytoma and central nervous system primitive neuroectodermal tumor are both neural crest-derived tumors. The former is usually benign and develops mainly in adulthood and the latter brain tumor mainly occurs in childhood and has a poor prognosis. We report a case of a 15-year-old boy who developed pheochromocytoma after more than 10 years of complete remission of central primitive neuroectodermal tumor. Thus far, there have been no reports of childhood cancer survivors who developed pheochromocytoma. This quite rare occurrence of two tumors in a single patient may imply some unidentified linkage or common genetic background.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/diagnóstico , Neoplasias Encefálicas/diagnóstico , Neoplasias Primarias Múltiples , Feocromocitoma/diagnóstico , Neoplasias de las Glándulas Suprarrenales/terapia , Neoplasias Encefálicas/terapia , Terapia Combinada , Diagnóstico Diferencial , Humanos , Lactante , Imagen por Resonancia Magnética , Masculino , Tumores Neuroectodérmicos Primitivos , Feocromocitoma/terapia , Tomografía Computarizada por Rayos X
18.
Pediatr Diabetes ; 13(1): 26-32, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22060211

RESUMEN

BACKGROUND: In Asians, mutations in the known maturity-onset diabetes of the young (MODY) genes have been identified in only <15% of patients. These results were obtained mostly through studies on adult patients. OBJECTIVE: To investigate the molecular basis of Japanese patients with pediatric-onset MODY-type diabetes. SUBJECTS: Eighty Japanese patients with pediatric-onset MODY-type diabetes. METHODS: Mitochondrial 3243A>G mutation was first tested by the polymerase chain reaction restriction fragment length polymorphism analysis for maternally inherited families. Then, all coding exons and exon-intron boundaries of the HNF1A, HNF1B, GCK, and HNF4A genes were amplified from genomic DNA and directly sequenced. Multiplex ligation-dependent probe amplification analysis was also performed to detect whole-exon deletions. RESULTS: After excluding one patient with a mitochondrial 3243A>G, mutations were identified in 38 (48.1%) patients; 18 had GCK mutations, 11 had HNF1A mutations, 3 had HNF4A mutations, and 6 had HNF1B mutations. In patients aged <8 yr, mutations were detected mostly in GCK at a higher frequency (63.6%). In patients >9 yr of age, mutations were identified less frequently (45.1%), with HNF1A mutations being the most frequent. A large fraction of mutation-negative patients showed elevated homeostasis model assessment (HOMA) insulin-resistance and normal HOMA-ß indices. Most of the HNF1B mutations were large deletions, and, interestingly, renal cysts were undetectable in two patients with whole-gene deletion of HNF1B. CONCLUSION: In Japanese patients with pediatric-onset MODY-type diabetes, mutations in known genes were identified at a much higher frequency than previously reported for adult Asians. A fraction of mutation-negative patients presented with insulin-resistance and normal insulin-secretory capacities resembling early-onset type 2 diabetes.


Asunto(s)
Pueblo Asiatico/genética , Análisis Mutacional de ADN , Diabetes Mellitus Tipo 2/genética , Adolescente , Edad de Inicio , Pueblo Asiatico/estadística & datos numéricos , Niño , Preescolar , Análisis Mutacional de ADN/métodos , ADN Mitocondrial/análisis , ADN Mitocondrial/genética , Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/etnología , Femenino , Frecuencia de los Genes , Glucoquinasa/genética , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 1-beta del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/genética , Humanos , Lactante , Masculino
19.
Neurology ; 98(19): 808-813, 2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35351784

RESUMEN

Onasemnogene abeparvovec is an adeno-associated virus vector-based gene therapy for spinal muscular atrophy (SMA). Although several cases of drug-induced thrombotic microangiopathy due to onasemnogene abeparvovec have been reported, none has been confirmed pathologically. Here, we present renal histopathologic findings of TMA due to onasemnogene abeparvovec. On day 5 after receiving onasemnogene abeparvovec, a 23-month-old girl with SMA type 1 developed thrombocytopenia, microangiopathic hemolytic anemia, liver dysfunction, acute kidney injury, and hypertension. She was diagnosed with TMA and received an increased dose of prednisolone, antihypertensives, diuretics, packed red blood cell and platelet transfusion, a single dose of eculizumab, 4 cycles of plasmapheresis, and intermittent and continuous hemodialysis. Her TMA resolved by day 30. On day 49, renal biopsy was performed. Light microscopy revealed proliferation of glomerular mesangial cells and matrix, with mesangiolysis, endothelial cell swelling, and partial double contours of the glomerular basement membrane. Electron microscopy showed endothelial injury, with edematous changes of the subendothelial spaces and neoformation of the basement membrane, without electron-dense depositions. These findings are compatible with the recovery phase of TMA. One year after drug administration, her motor function is improved. She can hold her posture against gravity and has neither dysphagia nor respiratory disturbance, but mild hypertension persists. Physicians should be vigilant regarding TMA as a severe side effect of onasemnogene abeparvovec treatment, especially when thrombocytopenia, hemolytic anemia, increased lactate dehydrogenase, or acute kidney injury is present.


Asunto(s)
Lesión Renal Aguda , Anemia , Hipertensión , Atrofia Muscular Espinal , Neurología , Microangiopatías Trombóticas , Femenino , Humanos , Lactante , Atrofia Muscular Espinal/genética , Microangiopatías Trombóticas/inducido químicamente , Microangiopatías Trombóticas/terapia
20.
Sci Rep ; 10(1): 14026, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32820208

RESUMEN

Galactose-deficient IgA1 (Gd-IgA1) is important in the pathogenesis of IgA nephropathy (IgAN). A Gd-IgA1-specific monoclonal antibody (KM55) has revealed glomerular Gd-IgA1 deposition solely in patients with IgAN and IgA vasculitis with nephritis (IgAV-N). However, this specificity is controversial and has not been demonstrated in pediatric patients. Here, we conducted double-immunofluorescence staining of IgA and Gd-IgA1 in 60 pediatric patients with various glomerular diseases. We divided patients into four groups: (1) patients with IgAN and IgAV-N (n = 23); (2) patients with immunocomplex-mediated glomerulonephritis accompanied by IgA deposition, including lupus nephritis, membranoproliferative glomerulonephritis, and membranous nephropathy (n = 14); (3) patients with other glomerular diseases involving IgA deposition, including idiopathic nephrotic syndrome (INS), oligomeganephronia, Alport syndrome, dense deposit disease, and crescentic glomerulonephritis (n = 11); and (4) patients with IgA-negative diseases including INS, membranoproliferative glomerulonephritis, membranous nephropathy, oligomeganephronia, Alport syndrome, C3 glomerulonephritis, poststreptococcal acute glomerulonephritis, and hemolytic uremic syndrome (n = 12). KM55 staining revealed Gd-IgA1-positive findings in 23/23 patients in Group 1 and 13/14 patients in Group 2, but not in patients in Groups 3 or 4. Therefore, KM55 may detect incidental IgA deposition in pediatric patients. Gd-IgA1 may be involved in the pathogenesis of these immune-related diseases; alternatively, KM55 may recognize IgA-related immunocomplexes in a non-specific manner.


Asunto(s)
Galactosa/metabolismo , Glomerulonefritis por IGA/metabolismo , Glomerulonefritis/metabolismo , Inmunoglobulina A/metabolismo , Niño , Femenino , Técnica del Anticuerpo Fluorescente , Glomerulonefritis/patología , Glomerulonefritis por IGA/patología , Humanos , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA