Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 794
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Diabetologia ; 67(1): 113-123, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37897565

RESUMEN

AIMS/HYPOTHESIS: Monogenic diabetes is estimated to account for 1-6% of paediatric diabetes cases in primarily non-consanguineous populations, while the incidence and genetic spectrum in consanguineous regions are insufficiently defined. In this single-centre study we aimed to evaluate diabetes subtypes, obtain the consanguinity rate and study the genetic background of individuals with syndromic and neonatal diabetes in a population with a high rate of consanguinity. METHODS: Data collection was carried out cross-sectionally in November 2021 at the paediatric diabetic clinic, Dr Jamal Ahmad Rashed Hospital, in Sulaimani, Kurdistan, Iraq. At the time of data collection, 754 individuals with diabetes (381 boys) aged up to 16 years were registered. Relevant participant data was obtained from patient files. Consanguinity status was known in 735 (97.5%) participants. Furthermore, 12 families of children with neonatal diabetes and seven families of children with syndromic diabetes consented to genetic testing by next-generation sequencing. Prioritised variants were evaluated using the American College of Medical Genetics and Genomics guidelines and confirmed by Sanger sequencing. RESULTS: A total of 269 of 735 participants (36.5%) with known consanguinity status were offspring of consanguineous families. An overwhelming majority of participants (714/754, 94.7%) had clinically defined type 1 diabetes (35% of them were born to consanguineous parents), whereas only eight (1.1%) had type 2 diabetes (38% consanguineous). Fourteen (1.9%) had neonatal diabetes (50% consanguineous), seven (0.9%) had syndromic diabetes (100% consanguineous) and 11 (1.5%) had clinically defined MODY (18% consanguineous). We found that consanguinity was significantly associated with syndromic diabetes (p=0.0023) but not with any other diabetes subtype. The genetic cause was elucidated in ten of 12 participants with neonatal diabetes who consented to genetic testing (homozygous variants in GLIS3 [sibling pair], PTF1A and ZNF808 and heterozygous variants in ABCC8 and INS) and four of seven participants with syndromic diabetes (homozygous variants in INSR, SLC29A3 and WFS1 [sibling pair]). In addition, a participant referred as syndromic diabetes was diagnosed with mucolipidosis gamma and probably has type 2 diabetes. CONCLUSIONS/INTERPRETATION: This unique single-centre study confirms that, even in a highly consanguineous population, clinically defined type 1 diabetes is the prevailing paediatric diabetes subtype. Furthermore, a pathogenic cause of monogenic diabetes was identified in 83% of tested participants with neonatal diabetes and 57% of participants with syndromic diabetes, with most variants being homozygous. Causative genes in our consanguineous participants were markedly different from genes reported from non-consanguineous populations and also from those reported in other consanguineous populations. To correctly diagnose syndromic diabetes in consanguineous populations, it may be necessary to re-evaluate diagnostic criteria and include additional phenotypic features such as short stature and hepatosplenomegaly.


Asunto(s)
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Enfermedades del Recién Nacido , Masculino , Recién Nacido , Humanos , Niño , Anciano , Diabetes Mellitus Tipo 1/epidemiología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/diagnóstico , Consanguinidad , Estudios de Cohortes , Irak/epidemiología , Enfermedades del Recién Nacido/genética , Mutación/genética , Proteínas de Transporte de Nucleósidos/genética
2.
Mol Biol Rep ; 51(1): 753, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38874636

RESUMEN

BACKGROUND: The diagnosis of neonatal diabetes can be problematic in preterm infants with fetal growth restriction (FGR). Growth restricted fetuses may have impaired insulin production and secretion; low birthweight infants may have a reduced response to insulin. We report a novel missense ABCC8 variant associated with a clinical phenotype compatible with transient neonatal diabetes mellitus (TNDM) in a fetal growth restricted preterm infant. METHODS AND RESULTS: A preterm growth restricted infant experienced hyperglycemia from the first day of life, requiring insulin therapy on the 13th and 15th day of life and leading to the diagnosis of TNDM. Glycemic values normalized from the 35th day of life onwards. Genetic screening was performed by next generation sequencing, using a Clinical Exon panel of 4800 genes, filtered for those associated with the clinical presentation and by means of methylation-specific multiplex ligation-dependent probe amplification analysis to identify chromosomal aberrations at 6q24. Genetic tests excluded defects at 6q24 and were negative for KCNJ11, SLC2A2 (GLUT-2) and HNF1B, but revealed the presence of the heterozygous missense variant c.2959T > C (p.Ser987Pro) in ABCC8 gene. The presence of the variant was excluded in parents' DNA and the proband variant was then considered de novo. CONCLUSIONS: In our infant, the persistence of hyperglycemia beyond 3 weeks of life led us to the diagnosis of TNDM and to hypothesize a possible genetic cause. The genetic variant we found could be, most likely, the main cause of both FGR and TNDM.


Asunto(s)
Diabetes Mellitus , Retardo del Crecimiento Fetal , Mutación Missense , Receptores de Sulfonilureas , Humanos , Retardo del Crecimiento Fetal/genética , Mutación Missense/genética , Receptores de Sulfonilureas/genética , Recién Nacido , Diabetes Mellitus/genética , Femenino , Masculino , Recien Nacido Prematuro , Insulina/metabolismo , Enfermedades del Recién Nacido/genética , Enfermedades del Recién Nacido/diagnóstico
3.
Clin Genet ; 103(4): 434-447, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36510364

RESUMEN

Neonatal diabetes mellitus (NDM) is defined as the occurrence of severe hyperglycemia in infants under 6 months old and may be permanent (PNDM) or transient (TNDM). When diabetes is diagnosed at 6-12 months of age (early onset diabetes [EOD]), the etiology may be monogenic; however, most cases consist of type 1 diabetes mellitus (T1DM). Molecular diagnosis was determined in a cohort of 35 unrelated Brazilian patients with NDM or EOD based on targeted next-generation sequencing panel and/or chromosome 6q24 abnormalities. The impact of genetic testing on treatment and follow-up was evaluated. Overall, 24 patients had NDM: with 18 (75.0%) having PNDM, 5 TNDM (20.8%) and 1 case in which this information was unknown. Eleven patients had EOD. Genetic testing was positive in 20/24 patients with NDM (83.3%) and in 18.2% of cases of EOD. The commonest causes were ATP-sensitive potassium (KATP) channel genes, and GCK and IPEX mutations (37.1%, 11.4% and 5.7%, respectively). Patients with PNDM due to KCNJ11 and ABCC8 mutations transitioned successfully to sulfonylureas in almost 60% of cases, reinforcing the benefit of performing genetic testing in NDM as early as possible. This report refers to the largest series of cases of NDM (TNDM and PNDM) and EOD in Brazil in which patients were submitted to molecular investigation and in which the clinical impact of genetic diagnosis was also evaluated.


Asunto(s)
Diabetes Mellitus Tipo 1 , Diabetes Mellitus , Enfermedades del Recién Nacido , Canales de Potasio de Rectificación Interna , Lactante , Recién Nacido , Humanos , Brasil , Canales de Potasio de Rectificación Interna/genética , Diabetes Mellitus Tipo 1/genética , Mutación , Pruebas Genéticas , Enfermedades del Recién Nacido/genética , Diabetes Mellitus/genética
4.
Diabet Med ; 40(5): e15013, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36398453

RESUMEN

AIMS: The aim of this study is to elucidate the aetiology and clinical features of neonatal and early-onset diabetes in a large database for pediatric diabetes patients in Ukraine. METHODS: We established a Pediatric Diabetes Register to identify patients diagnosed with diabetes before 9 months of age. Genetic testing was undertaken for 66 patients from 65 unrelated families with diabetes diagnosed within the first 6 months of life (neonatal diabetes, n = 36) or between 6 and 9 months (early-onset diabetes, n = 30). RESULTS: We determined the genetic aetiology in 86.1% of patients (31/36) diagnosed before 6 months and in 20% (6/30) diagnosed between 6 and 9 months. Fourteen individuals (37.8% of those with a genetic cause identified) had activating heterozygous variants in ABCC8 or KCNJ11. An additional 10 individuals had pathogenic variants in the INS or GCK genes, while 4 had 6q24 transient neonatal diabetes. Rare genetic subtypes (including pathogenic variants in EIF2AK3, GLIS3, INSR, PDX1, LRBA, RFX6 and FOXP3) were identified in nine probands (24.3% of solved cases), 6 of whom died. In total, eight individuals died between infancy and childhood, all of them were diagnosed before 6 months and had received a genetic diagnosis. CONCLUSIONS: In the last decade, the increased availability of comprehensive genetic testing has resulted in increased recognition of the contribution of rare genetic subtypes within pediatric diabetes cohorts. In our study, we identified a high mortality rate among these patients.


Asunto(s)
Diabetes Mellitus , Enfermedades del Recién Nacido , Recién Nacido , Humanos , Niño , Ucrania , Diabetes Mellitus/diagnóstico , Pruebas Genéticas , Enfermedades del Recién Nacido/genética , Proteínas Adaptadoras Transductoras de Señales/genética
5.
Am J Hum Genet ; 104(2): 213-228, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30639323

RESUMEN

Primary defects in lung branching morphogenesis, resulting in neonatal lethal pulmonary hypoplasias, are incompletely understood. To elucidate the pathogenetics of human lung development, we studied a unique collection of samples obtained from deceased individuals with clinically and histopathologically diagnosed interstitial neonatal lung disorders: acinar dysplasia (n = 14), congenital alveolar dysplasia (n = 2), and other lethal lung hypoplasias (n = 10). We identified rare heterozygous copy-number variant deletions or single-nucleotide variants (SNVs) involving TBX4 (n = 8 and n = 2, respectively) or FGF10 (n = 2 and n = 2, respectively) in 16/26 (61%) individuals. In addition to TBX4, the overlapping ∼2 Mb recurrent and nonrecurrent deletions at 17q23.1q23.2 identified in seven individuals with lung hypoplasia also remove a lung-specific enhancer region. Individuals with coding variants involving either TBX4 or FGF10 also harbored at least one non-coding SNV in the predicted lung-specific enhancer region, which was absent in 13 control individuals with the overlapping deletions but without any structural lung anomalies. The occurrence of rare coding variants involving TBX4 or FGF10 with the putative hypomorphic non-coding SNVs implies a complex compound inheritance of these pulmonary hypoplasias. Moreover, they support the importance of TBX4-FGF10-FGFR2 epithelial-mesenchymal signaling in human lung organogenesis and help to explain the histopathological continuum observed in these rare lethal developmental disorders of the lung.


Asunto(s)
Factor 10 de Crecimiento de Fibroblastos/genética , Enfermedades del Recién Nacido/genética , Enfermedades del Recién Nacido/mortalidad , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/mortalidad , Transducción de Señal/genética , Proteínas de Dominio T Box/genética , Variaciones en el Número de Copia de ADN/genética , Femenino , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica , Edad Gestacional , Humanos , Recién Nacido , Enfermedades del Recién Nacido/metabolismo , Enfermedades del Recién Nacido/patología , Pulmón/embriología , Pulmón/crecimiento & desarrollo , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/patología , Masculino , Herencia Materna , Organogénesis , Herencia Paterna , Linaje , Polimorfismo de Nucleótido Simple/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Proteínas de Dominio T Box/metabolismo
6.
Pediatr Res ; 91(4): 896-902, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-33846556

RESUMEN

BACKGROUND: The contribution of pathogenic gene variants with development of epilepsy after acute symptomatic neonatal seizures is not known. METHODS: Case-control study of 20 trios in children with a history of acute symptomatic neonatal seizures: 10 with and 10 without post-neonatal epilepsy. We performed whole-exome sequencing (WES) and identified pathogenic de novo, transmitted, and non-transmitted variants from established and candidate epilepsy association genes and correlated prevalence of these variants with epilepsy outcomes. We performed a sensitivity analysis with genes associated with coronary artery disease (CAD). We analyzed variants throughout the exome to evaluate for differential enrichment of functional properties using exploratory KEGG searches. RESULTS: Querying 200 established and candidate epilepsy genes, pathogenic variants were identified in 5 children with post-neonatal epilepsy yet in only 1 child without subsequent epilepsy. There was no difference in the number of trios with non-transmitted pathogenic variants in epilepsy or CAD genes. An exploratory KEGG analysis demonstrated a relative enrichment in cell death pathways in children without subsequent epilepsy. CONCLUSIONS: In this pilot study, children with epilepsy after acute symptomatic neonatal seizures had a higher prevalence of coding variants with a targeted epilepsy gene sequencing analysis compared to those patients without subsequent epilepsy. IMPACT: We performed whole-exome sequencing (WES) in 20 trios, including 10 children with epilepsy and 10 without epilepsy, both after acute symptomatic neonatal seizures. Children with post-neonatal epilepsy had a higher burden of pathogenic variants in epilepsy-associated genes compared to those without post-neonatal epilepsy. Future studies evaluating this association may lead to a better understanding of the risk of epilepsy after acute symptomatic neonatal seizures and elucidate molecular pathways that are dysregulated after brain injury and implicated in epileptogenesis.


Asunto(s)
Epilepsia , Enfermedades del Recién Nacido , Estudios de Casos y Controles , Niño , Epilepsia/genética , Humanos , Recién Nacido , Enfermedades del Recién Nacido/genética , Mutación , Proyectos Piloto , Convulsiones/epidemiología , Convulsiones/genética , Secuenciación del Exoma
7.
Pediatr Diabetes ; 23(6): 668-674, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35394098

RESUMEN

Neonatal diabetes mellitus with congenital hypothyroidism (NDH) syndrome (MIM# 610199) is a rare disease caused by autosomal recessive mutations in the GLIS3 gene. GLIS3 is an important transcription factor that might acts as both a repressor and activator of transcription. To date, 22 cases of NDH syndrome from 16 families and 11 countries have been described. Herein, we report a child who developed diabetes during the first week of age. Additionally, she suffered from congenital hypothyroidism, cardiac abnormalities, and polycystic kidney disease. Genetic analysis revealed that patient is a carrier of two novel heterozygous mutations, p.Pro444fsdelG (c.1330delC) and p.His647Arg (c.1940A > G) in the GLIS3 gene. Each was inherited from clinically healthy father and mother, respectively. Bioinformatic tools (SIFT, PolyPhen2, PROVEAN and SWISS-MODEL) declared that the p.His647Arg (c.1940A > G) variant has strong detrimental effect and disturbs Kruppel-like zinc finger domain. All but one so far described cases of NDH syndrome have been caused by homozygous of GLIS3, making the described case the second case of pathogenic, compound heterozygosity of GLIS3 worldwide posing substantial clinical novelty and detailing an interesting interplay between the observed variants and GLIS3 expression, which seems to be autoregulated. Hence, the damaging missense mutation may further reduce the expression of any remaining functional alleles. This case report expands our understanding of the clinical phenotype, treatment approaches, and outcome of infants with GLIS3 mutations and indicates the need for further research to deepen our understanding of the role of GLIS3.


Asunto(s)
Hipotiroidismo Congénito , Diabetes Mellitus , Enfermedades del Recién Nacido , Hipotiroidismo Congénito/complicaciones , Hipotiroidismo Congénito/genética , Proteínas de Unión al ADN/genética , Diabetes Mellitus/congénito , Diabetes Mellitus/genética , Femenino , Humanos , Recién Nacido , Enfermedades del Recién Nacido/genética , Mutación , Proteínas Represoras/genética , Transactivadores/genética , Transactivadores/metabolismo
8.
Pediatr Diabetes ; 23(6): 675-692, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35657808

RESUMEN

OBJECTIVE: In monogenic diabetes due to KCNJ11 and ABCC8 mutations that impair KATP- channel function, sulfonylureas improve long-term glycemic control. Although KATP channels are extensively expressed in the brain, the effect of sulfonylureas on neurological function has varied widely. We evaluated published evidence about potential effects of sulfonylureas on neurological features, especially epilepsy, cognition, motor function and muscular tone, visuo-motor integration, and attention deficits in children and adults with KCNJ11 and ABCC8-related neonatal-onset diabetes mellitus. RESEARCH DESIGN AND METHODS: We conducted a systematic review and meta-analyses of the literature (PROSPERO, CRD42021254782), including individual-patient data, according to PRISMA, using RevMan software. We also graded the level of evidence. RESULTS: We selected 34 of 776 publications. The evaluation of global neurological function before and after sulfonylurea (glibenclamide) treatment in 114 patients yielded a risk difference (RD) of 58% (95%CI, 43%-74%; I2  = 54%) overall and 73% (95%CI, 32%-113%; I2  = 0%) in the subgroup younger than 4 years; the level of evidence was moderate and high, respectively. EEG studies of epilepsy showed a RD of 56% (95%CI, 23%-89%; I2  = 34%) in patients with KCNJ11 mutations, with a high quality of evidence. For hypotonia and motor function, the RDs were 90% (95%CI, 69%-111%; I2  = 0%) and 73% (95%CI, 35%-111%; I2  = 0%), respectively, with a high level of evidence. CONCLUSIONS: Glibenclamide significantly improved neurological abnormalities in patients with neonatal-onset diabetes due to KCNJ11 or ABCC8 mutations. Hypotonia was the symptom that responded best. Earlier treatment initiation was associated with greater benefits.


Asunto(s)
Diabetes Mellitus , Epilepsia , Enfermedades del Recién Nacido , Canales de Potasio de Rectificación Interna , Adulto , Niño , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/genética , Epilepsia/genética , Gliburida , Humanos , Recién Nacido , Enfermedades del Recién Nacido/tratamiento farmacológico , Enfermedades del Recién Nacido/genética , Canales KATP/genética , Hipotonía Muscular , Mutación , Canales de Potasio de Rectificación Interna/genética , Compuestos de Sulfonilurea/uso terapéutico , Receptores de Sulfonilureas/genética
9.
Pediatr Diabetes ; 23(7): 999-1008, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35822653

RESUMEN

OBJECTIVE: To describe clinical presentation/longterm outcomes of patients with ABCC8/KCNJ11 variants in a large cohort of patients with diabetes. RESEARCH DESIGN AND METHODS: We analyzed patients in the Diabetes Prospective Follow-up (DPV) registry with diabetes and pathogenic variants in the ABCC8/KCNJ11 genes. For patients with available data at three specific time-points-classification as K+ -channel variant, 2-year follow-up and most recent visit-the longitudinal course was evaluated in addition to the cross-sectional examination. RESULTS: We identified 93 cases with ABCC8 (n = 54)/KCNJ11 (n = 39) variants, 63 of them with neonatal diabetes. For 22 patients, follow-up data were available. Of these, 19 were treated with insulin at diagnosis, and the majority of patients was switched to sulfonylurea thereafter. However, insulin was still administered in six patients at the most recent visit. Patients were in good metabolic control with a median (IQR) A1c level of 6.0% (5.5-6.7), that is, 42.1 (36.6-49.7) mmol/mol after 2 years and 6.7% (6.0-8.0), that is, 49.7 (42.1-63.9) mmol/mol at the most recent visit. Five patients were temporarily without medication for a median (IQR) time of 4.0 (3.5-4.4) years, while two other patients continue to be off medication at the last follow-up. CONCLUSIONS: ABCC8/KCNJ11 variants should be suspected in children diagnosed with diabetes below the age of 6 months, as a high percentage can be switched from insulin to oral antidiabetic drugs. Thirty patients with diabetes due to pathogenic variants of ABCC8 or KCNJ11 were diagnosed beyond the neonatal period. Patients maintain good metabolic control even after a diabetes duration of up to 11 years.


Asunto(s)
Diabetes Mellitus Tipo 2 , Diabetes Mellitus , Enfermedades del Recién Nacido , Canales de Potasio de Rectificación Interna , Niño , Humanos , Lactante , Recién Nacido , Austria/epidemiología , Estudios Transversales , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/epidemiología , Diabetes Mellitus/genética , Diabetes Mellitus Tipo 2/genética , Hemoglobina Glucada , Hipoglucemiantes/uso terapéutico , Enfermedades del Recién Nacido/diagnóstico , Enfermedades del Recién Nacido/epidemiología , Enfermedades del Recién Nacido/genética , Insulina/uso terapéutico , Mutación , Canales de Potasio de Rectificación Interna/genética , Estudios Prospectivos , Sistema de Registros , Receptores de Sulfonilureas/genética
10.
J Pediatr Hematol Oncol ; 44(1): e250-e252, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33661169

RESUMEN

We report a female newborn with acute myelogenous leukemia (AML) associated with a MYB-GATA1 fusion gene. Morphologic findings of myeloid lineage were obtained using light microscopy. Cytogenetic analysis of peripheral blood showed a complex karyotype: 46,X,-X,add(3)(q21),der(6)add(6)(q21)del(6)(q?), +mar1[5]/46,XX[15]. Targeted RNA sequencing revealed a MYB-GATA1 fusion gene. Reduced-dose AML-type chemotherapy resulted in remission and survival for >3 years without relapse. The present case demonstrated the feasibility of carrying out targeted RNA sequencing for identifying MYB-GATA1 and supports the notion that neonatal AML with MYB-GATA1 with reduced chemotherapy may show better prognosis than other highly toxic therapies.


Asunto(s)
Aberraciones Cromosómicas , Factor de Transcripción GATA1/genética , Enfermedades del Recién Nacido , Leucemia Mieloide Aguda , Proteínas de Fusión Oncogénica/genética , Proteínas Proto-Oncogénicas c-myb/genética , Femenino , Humanos , Recién Nacido , Enfermedades del Recién Nacido/tratamiento farmacológico , Enfermedades del Recién Nacido/genética , Leucemia Mieloide Aguda/congénito , Leucemia Mieloide Aguda/tratamiento farmacológico
11.
BMC Pregnancy Childbirth ; 22(1): 88, 2022 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-35100977

RESUMEN

BACKGROUND: MTHFD1 gene may affect the embryonic development by elevated homocysteine levels, DNA synthesis and DNA methylation, but limited number of genetic variants of MTHFD1 gene was focused on the association with congenital heart disease (CHD). This study examined the role of MTHFD1 gene and maternal smoking on infant CHD risk, and investigated their interaction effects in Chinese populations. METHODS: A case-control study of 464 mothers of CHD infants and 504 mothers of health controls was performed. The exposures of interest were maternal tobacco exposure, single nucleotide polymorphisms (SNPs) of maternal MTHFD1 gene. The logistic regression model was used for accessing the strength of association. RESULTS: Mothers exposed to secondhand smoke during 3 months before pregnancy (adjusted odds ratio [aOR] = 1.56; 95% confidence interval [CI]: 1.13-2.15) and in the first trimester of pregnancy (aOR = 2.24; 95%CI: 1.57-3.20) were observed an increased risk of CHD. Our study also found that polymorphisms of maternal MTHFD1 gene at rs1950902 (AA vs. GG: aOR = 1.73, 95% CI: 1.01-2.97), rs2236222 (GG vs. AA: aOR = 2.38, 95% CI: 1.38-4.12), rs1256142 (GA vs.GG: aOR = 1.57, 95% CI: 1.01-2.45) and rs11849530 (GG vs. AA: aOR = 1.68, 95% CI: 1.02-2.77) were significantly associated with higher risk of CHD. However, we did not observe a significant association between maternal MTHFD1 rs2236225 and offspring CHD risk. Furthermore, we found the different degrees of interaction effects between polymorphisms of the MTHFD1 gene including rs1950902, rs2236222, rs1256142, rs11849530 and rs2236225, and maternal tobacco exposure. CONCLUSIONS: Maternal polymorphisms of MTHFD1 gene, maternal tobacco exposure and their interactions are significantly associated with the risk of CHD in offspring in Han Chinese populations. However, more studies in different ethnic populations with a larger sample and prospective designs are required to confirm our findings. TRIAL REGISTRATION: Registration number: ChiCTR1800016635 .


Asunto(s)
Cardiopatías Congénitas/genética , Enfermedades del Recién Nacido/genética , Metilenotetrahidrofolato Deshidrogenasa (NADP)/genética , Antígenos de Histocompatibilidad Menor/genética , Polimorfismo de Nucleótido Simple , Adulto , Pueblo Asiatico , Estudios de Casos y Controles , China/epidemiología , Femenino , Cardiopatías Congénitas/inducido químicamente , Humanos , Recién Nacido , Enfermedades del Recién Nacido/inducido químicamente , Modelos Logísticos , Exposición Materna/efectos adversos , Embarazo , Contaminación por Humo de Tabaco/efectos adversos , Fumar Tabaco/efectos adversos
12.
J Med Genet ; 58(4): 247-253, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32487539

RESUMEN

INTRODUCTION: TMEM16A is a calcium-activated chloride channel expressed in various secretory epithelia. Two siblings presented in early infancy with reduced intestinal peristalsis and recurrent episodes of haemorrhagic diarrhoea. In one of them, the episodes were characterised by hepatic pneumatosis with gas bubbles in the portal vein similar to necrotising enterocolitis of the newborn. METHODS: Exome sequencing identified a homozygous truncating pathogenic variant in ANO1. Expression analysis was performed using reverse transcription PCR, western blot and immunohistochemistry. Electrophysiological and cell biological studies were employed to characterise the effects on ion transport both in patient respiratory epithelial cells and in transfected HEK293 cells. RESULTS: The identified variant led to TMEM16A dysfunction, which resulted in abolished calcium-activated Cl- currents. Secondarily, CFTR function is affected due to the close interplay between both channels without inducing cystic fibrosis (CF). CONCLUSION: TMEM16A deficiency is a potentially fatal disorder caused by abolished calcium-activated Cl- currents in secretory epithelia. Secondary impairment of CFTR function did not cause a CF phenotyp, which may have implications for CF treatment.


Asunto(s)
Anoctamina-1/genética , Canales de Cloruro/genética , Predisposición Genética a la Enfermedad , Enfermedades del Recién Nacido/genética , Proteínas de Neoplasias/genética , Anoctamina-1/deficiencia , Transporte Biológico/genética , Calcio/metabolismo , Canales de Cloruro/metabolismo , Cloruros/metabolismo , Fibrosis Quística/genética , Fibrosis Quística/patología , Femenino , Células HEK293 , Humanos , Lactante , Recién Nacido , Enfermedades del Recién Nacido/epidemiología , Enfermedades del Recién Nacido/patología , Masculino , Proteínas de Neoplasias/deficiencia
13.
Curr Issues Mol Biol ; 43(2): 1036-1042, 2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34563042

RESUMEN

Liang-Wang syndrome (LIWAS) is a polymalformative syndrome first described in 2019 caused by heterozygous mutation of the KCNMA1 gene encoding the Ca2+ and voltage-activated K+ channel (BKC). The KCNMA1 variant p.(Gly356Arg) abolishes the function of BKC and blocks the generation of K+ current. The phenotype of this variant includes developmental delay, and visceral and connective tissue malformations. So far, only three cases of LWAS have been described, one of which also had neonatal diabetes (ND). We present the case of a newborn affected by LIWAS carrying the p.(Gly375Arg) variant who manifested diabetes in the first week of life. The description of our case strongly increases the frequency of ND in LIWAS patients and suggests a role of BK inactivation in human insulin secretion. The knowledge on the role of BKC in insulin secretion is very poor. Analyzing the possible mechanisms that could explain the association of LIWAS with ND, we speculate that BK inactivation might impair insulin secretion through the alteration of ion-dependent membrane activities and mitochondrial functions in ß-cells, as well as the impaired intra-islet vessel reactivity.


Asunto(s)
Anomalías Congénitas/diagnóstico , Diabetes Mellitus/diagnóstico , Enfermedades del Recién Nacido/diagnóstico , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Sustitución de Aminoácidos , Canalopatías , Anomalías Congénitas/genética , Anomalías Congénitas/patología , Discapacidades del Desarrollo , Diabetes Mellitus/genética , Diabetes Mellitus/patología , Femenino , Humanos , Recién Nacido , Enfermedades del Recién Nacido/genética , Enfermedades del Recién Nacido/patología , Secreción de Insulina , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Mutación , Fenotipo , Embarazo
14.
Br J Haematol ; 192(4): 778-784, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33529380

RESUMEN

Neonatal alloimmune neutropenia (NAIN) is caused by maternal alloimmunisation to fetal human neutrophil antigens (HNAs). This study investigated maternal HNA/HLA alloantibodies involved with NAIN and identified the frequency of NAIN in Brazilian neonates. Neonatal neutropenia (neutrophil count < 1.5 × 109 /L) was investigated in samples from 10,000 unselected neonates, resulting in 88 neutropenic newborns (NBs) and their 83 mothers. Genotyping was performed by PCR-SSP (HNA-1/-4) and PCR-RFLP (HNA-3/-5). Serologic studies were performed by GAT (granulocyte agglutination test), Flow-WIFT (white blood cells immunofluorescence test) and LABScreen-Multi-HNA-Kit (OneLambda®) (LSM). Neonatal neutropenia was identified in 88/10,000 (0·9%) NBs. Genotyping revealed 60·2% maternal-fetal HNA incompatibilities (31·8% for HNA-1; 14·8% for HNA-3; 15·9% for HNA-4; 21·6% for HNA-5). Serologic studies revealed 37·3% of mothers with positive results with at least one technique. The detected anti-HNA specificities were confirmed in eight positive cases related to HNA-1/-3 systems. In cases with maternal-fetal HNA-4/-5 incompatibility, no specific neutrophil alloantibodies were found but anti-HLA I/II were present. Anti-HNA-2 was not identified. This is a large Brazilian study which involved the investigation of antibodies against all five HNA systems in neutropenia cases and showed a frequency of NAIN in 8/10,000 neonates. Among the HNA antibodies identified, we highlight the anti-HNA-1d and anti-HNA-3b, antibodies unusual in alloimmunised women, and rarely related to NAIN cases.


Asunto(s)
Enfermedades del Recién Nacido/diagnóstico , Neutropenia/diagnóstico , Brasil/epidemiología , Femenino , Genotipo , Humanos , Recién Nacido , Enfermedades del Recién Nacido/sangre , Enfermedades del Recién Nacido/epidemiología , Enfermedades del Recién Nacido/genética , Isoanticuerpos/sangre , Isoanticuerpos/genética , Isoanticuerpos/inmunología , Recuento de Leucocitos , Masculino , Neutropenia/sangre , Neutropenia/epidemiología , Neutropenia/genética , Neutrófilos/inmunología
15.
Hepatology ; 71(6): 2067-2079, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31595528

RESUMEN

BACKGROUND AND AIMS: The clinical consequences of defective primary cilium (ciliopathies) are characterized by marked phenotypic and genetic heterogeneity. Although fibrocystic liver disease is an established ciliopathy phenotype, severe neonatal cholestasis is rarely recognized as such. APPROACH AND RESULTS: We describe seven individuals from seven families with syndromic ciliopathy clinical features, including severe neonatal cholestasis (lethal in one and necessitating liver transplant in two). Positional mapping revealed a single critical locus on chromosome 7. Whole-exome sequencing revealed three different homozygous variants in Tetratricopeptide Repeat Domain 26 (TTC26) that fully segregated with the phenotype. TTC26 (intraflagellar transport [IFT] 56/DYF13) is an atypical component of IFT-B complex, and deficiency of its highly conserved orthologs has been consistently shown to cause defective ciliary function in several model organisms. We show that cilia in TTC26-mutated patient cells display variable length and impaired function, as indicated by dysregulated sonic hedgehog signaling, abnormal staining for IFT-B components, and transcriptomic clustering with cells derived from individuals with closely related ciliopathies. We also demonstrate a strong expression of Ttc26 in the embryonic mouse liver in a pattern consistent with its proposed role in the normal development of the intrahepatic biliary system. CONCLUSIONS: In addition to establishing a TTC26-related ciliopathy phenotype in humans, our results highlight the importance of considering ciliopathies in the differential diagnosis of severe neonatal cholestasis even in the absence of more typical features.


Asunto(s)
Colestasis Intrahepática/genética , Enfermedades del Recién Nacido/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Repeticiones de Tetratricopéptidos/genética , Animales , Ciliopatías , Diagnóstico Diferencial , Proteínas Hedgehog , Humanos , Recién Nacido , Ratones , Proteínas Asociadas a Microtúbulos/genética , Mutación , Transporte de Proteínas/genética , Índice de Severidad de la Enfermedad , Secuenciación del Exoma/métodos
16.
Clin Genet ; 99(3): 376-383, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33191500

RESUMEN

Failure to thrive (FTT) causes significant morbidity, often without clear etiologies. Six individuals of a large consanguineous family presented in the neonatal period with recurrent vomiting and diarrhea, leading to severe FTT. Standard diagnostic work up did not ascertain an etiology. Autozygosity mapping and whole exome sequencing identified homozygosity for a novel genetic variant of the long chain fatty acyl-CoA synthetase 5 (ACSL5) shared among the affected individuals (NM_203379.1:c.1358C>A:p.(Thr453Lys)). Autosomal recessive genotype-phenotype segregation was confirmed by Sanger sequencing. Functional in vitro analysis of the ACSL5 variant by immunofluorescence, western blotting and enzyme assay suggested that Thr453Lys is a loss-of-function mutation without any remaining activity. ACSL5 belongs to an essential enzyme family required for lipid metabolism and is known to contribute the major activity in the mouse intestine. Based on the function of ACSL5 in intestinal long chain fatty acid metabolism and the gastroenterological symptoms, affected individuals were treated with total parenteral nutrition or medium-chain triglyceride-based formula restricted in long-chain triglycerides. The patients responded well and follow up suggests that treatment is only required during early life.


Asunto(s)
Coenzima A Ligasas/genética , Insuficiencia de Crecimiento/genética , Enfermedades del Recién Nacido/genética , Metabolismo de los Lípidos , Animales , Células COS , Chlorocebus aethiops , Coenzima A Ligasas/metabolismo , Insuficiencia de Crecimiento/metabolismo , Femenino , Estudios de Asociación Genética , Variación Genética , Humanos , Recién Nacido , Enfermedades del Recién Nacido/metabolismo , Masculino , Mutación
17.
Invest New Drugs ; 39(3): 775-784, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33411210

RESUMEN

Infantile hemangioma is the most common vascular tumor of childhood. It is characterized by clinical expansion of endothelial cells and promoted by angiogenic factors. Luteolin is a flavonoid compound that carries anti-cancer and anti-angiogenesis properties. The study aimed to investigate the effect of luteolin in treating infantile hemangioma. We first tested the effect of luteolin on cell proliferative potential and VEGFA expression in hemangioma-derived stem cells (HemSCs). We then examined the efficacy of systemic application of luteolin in a murine hemangioma model. We then identified the downstream factor regulated by luteolin in HemSCs and validated its causative relationship with knock-down method in both in vitro and in vivo models. We also investigated the protein expression change of this targeting factor in proliferating hemangiomas. Luteolin inhibited HemSC growth and suppressed VEGF-A expression in a dose-dependent manner. Luteolin inhibited microvessel formation and de novo vasculogenesis in the murine model. FZD6 was induced by luteolin and exerted the anti-angiogenesis effect in our tumor models. Lastly, FZD6 level was repressed in the clinical tissues of human proliferating hemangiomas. Luteolin is a promising new agent to treat infantile hemangioma. Targeting the Wnt pathway may represent a potential therapeutic strategic to inhibit angiogenesis in proliferating hemangiomas.


Asunto(s)
Antineoplásicos/uso terapéutico , Receptores Frizzled/antagonistas & inhibidores , Hemangioma/tratamiento farmacológico , Enfermedades del Recién Nacido/tratamiento farmacológico , Luteolina/uso terapéutico , Neovascularización Patológica/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Fibroblastos/efectos de los fármacos , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Hemangioma/genética , Hemangioma/metabolismo , Hemangioma/patología , Humanos , Recién Nacido , Enfermedades del Recién Nacido/genética , Enfermedades del Recién Nacido/metabolismo , Enfermedades del Recién Nacido/patología , Luteolina/farmacología , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones Desnudos , Microvasos/efectos de los fármacos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Vía de Señalización Wnt/efectos de los fármacos
18.
Diabet Med ; 38(12): e14728, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34665882

RESUMEN

Neonatal diabetes is diagnosed before the age of 6 months and is usually caused by single-gene mutations. More than 30 genetic causes of neonatal diabetes have been described to date, resulting in severely reduced ß-cell number or function. Seven of these genes are known to cause neonatal diabetes through disrupted development of the whole pancreas, resulting in diabetes and exocrine pancreatic insufficiency. Pathogenic variants in five transcription factors essential for ß-cell development cause neonatal diabetes without other pancreatic phenotypes. However, additional extra-pancreatic features are common. This review will focus on the genes causing neonatal diabetes through disrupted ß-cell development, discussing what is currently known about the genetic and phenotypic features of these genetic conditions, and what discoveries may come in the future.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Enfermedades del Recién Nacido/genética , Mutación , Páncreas/metabolismo , Factores de Transcripción/genética , Diabetes Mellitus Tipo 1/metabolismo , Humanos , Recién Nacido , Enfermedades del Recién Nacido/metabolismo , Fenotipo
19.
Am J Med Genet A ; 185(2): 453-460, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33247552

RESUMEN

In Central-Africa, neonatal infections, asphyxia and prematurity are main reasons for admission to the neonatal intensive care unit and major determinants of newborn survival. Also, the outcome of newborns with congenital anomalies is expected to be poor, due to a lack of state-of-the art care. We conducted a study of 102 newborns recruited in the Neonatal Intensive Care Unit (NICU) at the University Hospitals of Kinshasa, DR Congo, to assess the impact of congenital anomalies. The presence of a major anomaly was associated with a hazard ratio of death of 13.2 (95%CI: 3.7-46.7, p < .001). In addition, the presence of three or more minor anomalies was associated with a 4.5-fold increased risk of death (95%CI: 1.1-18.6, p = .04). We conclude that like major anomalies, the presence of three or more minor anomalies should also be given particular attention and that the evaluation of dysmorphism should be promoted in NICU.


Asunto(s)
Anomalías Múltiples/epidemiología , Enfermedades del Recién Nacido/epidemiología , Unidades de Cuidado Intensivo Neonatal , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , República Democrática del Congo/epidemiología , Femenino , Hospitalización , Humanos , Recién Nacido , Enfermedades del Recién Nacido/diagnóstico , Enfermedades del Recién Nacido/genética , Masculino
20.
Mol Biol Rep ; 48(4): 3303-3311, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33864587

RESUMEN

Pathogenic variants (PVs) in CYP24A1 gene are associated with Idiopathic Infantile Hypercalcemia disease (IIH). The identification of CYP24A1 PVs can be a useful tool for the improvement of target therapeutic strategies. Aim of this study is to set up a rapid and inexpensive High Resolution Melting Analysis (HRMA)-based method for the simultaneous genotyping of two hot spot PVs in CYP24A1 gene, involved in IIH. A duplex-HRMA (dHRMA) was designed in order to detect simultaneously CYP24A1 c.428_430delAAG, p.(Glu143del) (rs777676129) and c.1186C > T, p.(Arg396Trp) (rs114368325), in peculiar cases addressed to our Laboratory. dHRMA was able to identify clearly and simultaneously both hot spot CYP24A1 PVs evaluating melting curve shape and melting temperature (Tm). This is the first dHRMA approach to rapidly screen the two most frequent CYP24A1 PVs in peculiar case, providing useful information for diagnosis and patient management in IIH disease.


Asunto(s)
Análisis Mutacional de ADN/métodos , Hipercalcemia/genética , Enfermedades del Recién Nacido/genética , Errores Innatos del Metabolismo/genética , Mutación , Vitamina D3 24-Hidroxilasa/genética , Niño , Humanos , Hipercalcemia/diagnóstico , Enfermedades del Recién Nacido/diagnóstico , Masculino , Errores Innatos del Metabolismo/diagnóstico , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA