Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Diabetes Res ; 2018: 3061620, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30599002

RESUMO

Precision medicine, the concept that specific treatments can be targeted to groups of individuals with specific genetic, cellular, or molecular features, is a key aspect of modern healthcare, and its use is rapidly expanding. In diabetes, the application of precision medicine has been demonstrated in monogenic disease, where sulphonylureas are used to treat patients with neonatal diabetes due to mutations in ATP-dependent potassium (KATP) channel genes. However, diabetes is highly heterogeneous, both between and within polygenic and monogenic subtypes. Making the correct diagnosis and using the correct treatment from diagnosis can be challenging for clinicians, but it is crucial to prevent long-term morbidity and mortality. To facilitate precision medicine in diabetes, research is needed to develop a better understanding of disease heterogeneity and its impact on potential treatments for specific subtypes. Animal models have been used in diabetes research, but they are not translatable to humans in the majority of cases. Advances in molecular genetics and functional laboratory techniques and availability and sharing of large population data provide exciting opportunities for human studies. This review will map the key elements of future diabetes research in humans and its potential for clinical translation to promote precision medicine in all diabetes subtypes.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Diabetes Mellitus Tipo 2/terapia , Medicina de Precisão , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/genética , Humanos
3.
Pediatr Diabetes ; 18(4): 320-323, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28318089

RESUMO

Congenital hyperinsulinaemic hypoglycaemia (HH) can occur in isolation or it may present as part of a wider syndrome. For approximately 40%-50% of individuals with this condition, sequence analysis of the known HH genes identifies a causative mutation. Identifying the underlying genetic aetiology in the remaining cases is important as a genetic diagnosis will inform on recurrence risk, may guide medical management and will provide valuable insights into ß-cell physiology. We sequenced the exome of a child with persistent diazoxide-responsive HH, mild aortic insufficiency, severe hypotonia, and developmental delay as well as the unaffected parents. This analysis identified a de novo mutation, p.G403D, in the proband's CACNA1D gene. CACNA1D encodes the main L-type voltage-gated calcium channel in the pancreatic ß-cell, a key component of the insulin secretion pathway. The p.G403D mutation had been reported previously as an activating mutation in an individual with primary hyper-aldosteronism, neuromuscular abnormalities, and transient hypoglycaemia. Sequence analysis of the CACNA1D gene in 60 further cases with HH did not identify a pathogenic mutation. Identification of an activating CACNA1D mutation in a second patient with congenital HH confirms the aetiological role of CACNA1D mutations in this disorder. A genetic diagnosis is important as treatment with a calcium channel blocker may be an option for the medical management of this patient.


Assuntos
Canais de Cálcio Tipo L/genética , Cardiopatias Congênitas/genética , Hiperinsulinismo/genética , Hipoglicemia/genética , Mutação , Doenças Neuromusculares/genética , Substituição de Aminoácidos , Insuficiência da Valva Aórtica/etiologia , Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/metabolismo , Análise Mutacional de DNA , Deficiências do Desenvolvimento/etiologia , Feminino , Macrossomia Fetal/etiologia , Cardiopatias Congênitas/metabolismo , Cardiopatias Congênitas/fisiopatologia , Humanos , Hiperinsulinismo/metabolismo , Hiperinsulinismo/fisiopatologia , Hipoglicemia/metabolismo , Hipoglicemia/fisiopatologia , Recém-Nascido , Hipotonia Muscular/etiologia , Doenças Neuromusculares/metabolismo , Doenças Neuromusculares/fisiopatologia
4.
Diabet Med ; 34(7): 1000-1004, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28173619

RESUMO

BACKGROUND: Children with neonatal diabetes often present with diabetic ketoacidosis and hence are at risk of cerebral oedema and subsequent long-term neurological deficits. These complications are difficult to identify because neurological features can also occur as a result of the specific genetic aetiology causing neonatal diabetes. CASE REPORTS: We report two cases of neonatal diabetes where ketoacidosis-related cerebral oedema was the major cause of their permanent neurological disability. Case 1 (male, 18 years, compound heterozygous ABCC8 mutation) and case 2 (female, 29 years, heterozygous KCNJ11 mutation) presented with severe diabetic ketoacidosis at 6 and 16 weeks of age. Both had reduced consciousness, seizures and required intensive care for cerebral oedema. They subsequently developed spastic tetraplegia. Neurological examination in adulthood confirmed spastic tetraplegia and severe disability. Case 1 is wheelchair-bound and needs assistance for transfers, washing and dressing, whereas case 2 requires institutional care for all activities of daily living. Both cases have first-degree relatives with the same mutation with diabetes, who did not have ketoacidosis at diagnosis and do not have neurological disability. DISCUSSION: Ketoacidosis-related cerebral oedema at diagnosis in neonatal diabetes can cause long-term severe neurological disability. This will give additional neurological features to those directly caused by the genetic aetiology of the neonatal diabetes. Our cases highlight the need for increased awareness of neonatal diabetes and earlier and better initial treatment of the severe hyperglycaemia and ketoacidosis often seen at diagnosis of these children.


Assuntos
Edema Encefálico/etiologia , Deficiências do Desenvolvimento/etiologia , Diabetes Mellitus/fisiopatologia , Cetoacidose Diabética/etiologia , Neuropatias Diabéticas/etiologia , Hiperglicemia/etiologia , Quadriplegia/etiologia , Adolescente , Adulto , Edema Encefálico/fisiopatologia , Deficiências do Desenvolvimento/fisiopatologia , Diabetes Mellitus/sangue , Diabetes Mellitus/genética , Cetoacidose Diabética/fisiopatologia , Neuropatias Diabéticas/fisiopatologia , Pessoas com Deficiência , Saúde da Família , Feminino , Humanos , Hiperglicemia/fisiopatologia , Masculino , Mutação , Canais de Potássio Corretores do Fluxo de Internalização/genética , Quadriplegia/fisiopatologia , Qualidade de Vida , Índice de Gravidade de Doença , Receptores de Sulfonilureias/genética
5.
Diabetes ; 65(7): 2094-2099, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27207547

RESUMO

Distinguishing patients with monogenic diabetes from those with type 1 diabetes (T1D) is important for correct diagnosis, treatment, and selection of patients for gene discovery studies. We assessed whether a T1D genetic risk score (T1D-GRS) generated from T1D-associated common genetic variants provides a novel way to discriminate monogenic diabetes from T1D. The T1D-GRS was highly discriminative of proven maturity-onset diabetes of young (MODY) (n = 805) and T1D (n = 1,963) (receiver operating characteristic area under the curve 0.87). A T1D-GRS of >0.280 (>50th T1D centile) was indicative of T1D (94% specificity, 50% sensitivity). We then analyzed the T1D-GRS of 242 white European patients with neonatal diabetes (NDM) who had been tested for all known NDM genes. Monogenic NDM was confirmed in 90, 59, and 8% of patients with GRS <5th T1D centile, 50-75th T1D centile, and >75th T1D centile, respectively. Applying a GRS 50th T1D centile cutoff in 48 NDM patients with no known genetic cause identified those most likely to have a novel monogenic etiology by highlighting patients with probable early-onset T1D (GRS >50th T1D centile) who were diagnosed later and had less syndromic presentation but additional autoimmune features compared with those with proven monogenic NDM. The T1D-GRS is a novel tool to improve the use of biomarkers in the discrimination of monogenic diabetes from T1D.


Assuntos
Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 2/diagnóstico , Predisposição Genética para Doença , Mutação , Adolescente , Adulto , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/genética , Diagnóstico Diferencial , Feminino , Genótipo , Humanos , Masculino , Adulto Jovem
6.
Diabet Med ; 33(10): 1387-91, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27086753

RESUMO

AIMS: Mutations in the KCNJ11 gene, which encodes the Kir6.2 subunit of the pancreatic KATP channel, cause neonatal diabetes. KCNJ11 is also expressed in the brain, and ~ 20% of those affected have neurological features, which may include features suggestive of psychiatric disorder. No previous studies have systematically characterized the psychiatric morbidity in people with KCNJ11 neonatal diabetes. We aimed to characterize the types of psychiatric disorders present in children with KCNJ11 mutations, and explore their impact on families. METHODS: The parents and teachers of 10 children with neonatal diabetes due to KCNJ11 mutations completed the Strengths and Difficulties Questionnaire and the Development and Wellbeing Assessment. Strengths and Difficulties Questionnaire scores were compared with normative data. Diagnoses from the Development and Wellbeing Assessment were compared with known clinical diagnoses. RESULTS: Strengths and Difficulties Questionnaire scores indicated high levels of psychopathology and impact. Psychiatric disorder(s) were present in all six children with the V59M or R201C mutation, and the presence of more than one psychiatric disorder was common. Only two children had received a formal clinical diagnosis, with a further one awaiting assessment, and the coexistence of more than one psychiatric disorder had been missed. Neurodevelopmental (attention deficit hyperactivity disorder and autism) and anxiety disorders predominated. CONCLUSIONS: Systematic assessment using standardized validated questionnaires reveals a range of psychiatric morbidity in children with KCNJ11 neonatal diabetes. This is under-recognized clinically and has a significant impact on affected children and their families. An integrated collaborative approach to clinical care is needed to manage the complex needs of people with KCNJ11 neonatal diabetes.


Assuntos
Diabetes Mellitus/genética , Diabetes Mellitus/psicologia , Transtornos do Neurodesenvolvimento/genética , Canais de Potássio Corretores do Fluxo de Internalização/genética , Adolescente , Substituição de Aminoácidos , Criança , Transtornos do Comportamento Infantil/complicações , Transtornos do Comportamento Infantil/epidemiologia , Transtornos do Comportamento Infantil/genética , Comorbidade , Diabetes Mellitus/epidemiologia , Feminino , Humanos , Recém-Nascido , Doenças do Recém-Nascido/genética , Masculino , Mutação de Sentido Incorreto , Transtornos do Neurodesenvolvimento/complicações , Transtornos do Neurodesenvolvimento/epidemiologia , Manifestações Neurológicas
7.
Diabet Med ; 31(1): e1-5, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23796040

RESUMO

BACKGROUND: Hepatocyte nuclear factor 4 alpha (HNF4A) gene mutations have a well-recognized role in maturity-onset diabetes of the young and have recently been described in congenital hyperinsulinism. A biphasic phenotype has been postulated, with macrosomia and congenital hyperinsulinism in infancy, and diabetes in young adulthood. In this case series, we report three children with HNF4A mutations (two de novo) and diazoxide-responsive congenital hyperinsulinism, highlighting the potential for ongoing diazoxide requirement and the importance of screening for these mutations even in the absence of family history. CASE REPORTS: All patients presented with macrosomia (mean birthweight 4.26 kg) and hyperinsulinaemic hypoglycaemia soon after birth (median age 1 day). All three (age range 7 months to 11 years 10 months) remain on diazoxide therapy, with dose requirements increasing in one patient. There was no prior family history of diabetes, neonatal hypoglycaemia or macrosomia. Parents were screened for HNF4A mutations post-diagnosis and one father was subsequently found to have maturity-onset diabetes of the young. CONCLUSIONS: This case series follows the evolving course of three patients with confirmed HNF4A-mediated congenital hyperinsulinism, highlighting (1) the variable natural history of these mutations, (2) the potential for prolonged diazoxide requirement, even into adolescence, and (3) the need for screening, regardless of family history.


Assuntos
Anti-Hipertensivos/uso terapêutico , Hiperinsulinismo Congênito/diagnóstico , Diazóxido/uso terapêutico , Fator 4 Nuclear de Hepatócito/sangue , Hipoglicemia/diagnóstico , Idade de Início , Peso ao Nascer , Glicemia/metabolismo , Criança , Pré-Escolar , Hiperinsulinismo Congênito/tratamento farmacológico , Hiperinsulinismo Congênito/genética , Diagnóstico Diferencial , Feminino , Macrossomia Fetal/metabolismo , Fator 4 Nuclear de Hepatócito/genética , Humanos , Hipoglicemia/tratamento farmacológico , Hipoglicemia/genética , Lactente , Masculino , Linhagem , Fenótipo
8.
Diabet Med ; 31(3): e11-5, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24299156

RESUMO

BACKGROUND: Hepatocyte nuclear factor 4α (HNF4A) is a member of the nuclear receptor family of ligand-activated transcription factors. HNF4A mutations cause hyperinsulinaemic hypoglycaemia in early life and maturity-onset diabetes of the young. Regular screening of HNF4A mutation carriers using the oral glucose tolerance test has been recommended to diagnose diabetes mellitus at an early stage. Glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide are incretin hormones, responsible for up to 70% of the secreted insulin after a meal in healthy individuals. We describe, for the first time, gradual alteration of glucose homeostasis in a patient with HNF4A mutation after resolution of hyperinsulinaemic hypoglycaemia, on serial oral glucose tolerance testing. We also measured the incretin response to a mixed meal in our patient. CASE REPORT: Our patient was born with macrosomia and developed hyperinsulinaemic hypoglycaemia in the neonatal period. Molecular genetic analysis confirmed HNF4A mutation (p.M116I, c.317G>A) as an underlying cause of hyperinsulinaemic hypoglycaemia. Serial oral glucose tolerance testing, after the resolution of hyperinsulinaemic hypoglycaemia, confirmed the diagnosis of maturity-onset diabetes of the young at the age of 10 years. Interestingly, the intravenous glucose tolerance test revealed normal glucose disappearance rate and first-phase insulin secretion. Incretin hormones showed a suboptimal rise in response to the mixed meal, potentially explaining the discrepancy between the oral glucose tolerance test and the intravenous glucose tolerance test. CONCLUSIONS: Maturity-onset diabetes of the young can develop as early as the first decade of life in persons with an HNF4A mutation. Impaired incretin response might be contributory in the early stages of HNF4A maturity-onset diabetes of the young.


Assuntos
Hiperinsulinismo Congênito/genética , Diabetes Mellitus Tipo 2/genética , Macrossomia Fetal/genética , Fator 4 Nuclear de Hepatócito/genética , Incretinas/metabolismo , Mutação , Glicemia/metabolismo , Criança , Hiperinsulinismo Congênito/fisiopatologia , Diabetes Mellitus Tipo 2/fisiopatologia , Macrossomia Fetal/fisiopatologia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Teste de Tolerância a Glucose , Humanos , Insulina/metabolismo , Masculino
9.
Diabetologia ; 56(9): 1958-63, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23771172

RESUMO

AIMS/HYPOTHESIS: Current genetic tests for diagnosing monogenic diabetes rely on selection of the appropriate gene for analysis according to the patient's phenotype. Next-generation sequencing enables the simultaneous analysis of multiple genes in a single test. Our aim was to develop a targeted next-generation sequencing assay to detect mutations in all known MODY and neonatal diabetes genes. METHODS: We selected 29 genes in which mutations have been reported to cause neonatal diabetes, MODY, maternally inherited diabetes and deafness (MIDD) or familial partial lipodystrophy (FPLD). An exon-capture assay was designed to include coding regions and splice sites. A total of 114 patient samples were tested--32 with known mutations and 82 previously tested for MODY (n = 33) or neonatal diabetes (n = 49) but in whom a mutation had not been found. Sequence data were analysed for the presence of base substitutions, small insertions or deletions (indels) and exonic deletions or duplications. RESULTS: In the 32 positive controls we detected all previously identified variants (34 mutations and 36 polymorphisms), including 55 base substitutions, ten small insertions or deletions and five partial/whole gene deletions/duplications. Previously unidentified mutations were found in five patients with MODY (15%) and nine with neonatal diabetes (18%). Most of these patients (12/14) had mutations in genes that had not previously been tested. CONCLUSIONS/INTERPRETATION: Our novel targeted next-generation sequencing assay provides a highly sensitive method for simultaneous analysis of all monogenic diabetes genes. This single test can detect mutations previously identified by Sanger sequencing or multiplex ligation-dependent probe amplification dosage analysis. The increased number of genes tested led to a higher mutation detection rate.


Assuntos
Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/genética , Testes Genéticos/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Feminino , Humanos , Recém-Nascido , Masculino , Mutação
10.
Diabetes Metab ; 39(4): 370-4, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23639568

RESUMO

Permanent neonatal diabetes mellitus is a rare condition mostly due to heterozygous mutations in the KCNJ11, ABCC8 and INS genes. Neonatal diabetes due to pancreatic agenesis is extremely rare. Mutations in PDX1, PTF1A, HNF1B, EIF2AK3, RFX6 and GATA6 genes have been shown to result in pancreatic agenesis or hypoplasia. This report describes a 40-day-old male infant diagnosed with permanent neonatal diabetes associated with atrial septal defect, pulmonary stenosis, patent ductus arteriosus and a novel de novo heterozygous missense mutation (p.N466S) in the GATA6 gene with no evidence of exocrine pancreas insufficiency. In addition to permanent neonatal diabetes, the patient had transient idiopathic neonatal cholestasis and hypoglycaemic episodes unrelated to insulin treatment, features that are rarely described in children with permanent neonatal diabetes.


Assuntos
Diabetes Mellitus/genética , Fator de Transcrição GATA6/genética , Cardiopatias Congênitas/genética , Anormalidades Múltiplas/genética , Cardiopatias Congênitas/complicações , Humanos , Lactente , Masculino , Mutação de Sentido Incorreto , Estenose da Valva Pulmonar/complicações , Estenose da Valva Pulmonar/congênito , Estenose da Valva Pulmonar/genética
11.
Diabetologia ; 56(4): 758-62, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23385738

RESUMO

AIMS/HYPOTHESIS: 6q24 transient neonatal diabetes mellitus (TNDM) is a rare form of diabetes presenting in the neonatal period that remits during infancy but, in a proportion of cases, recurs in later life. We aim to describe the clinical presentation of 6q24 TNDM in the largest worldwide cohort of patients with defined molecular aetiology, in particular seeking differences in presentation or clinical history between aetiological groups. METHODS: One-hundred and sixty-three patients with positively diagnosed 6q24 TNDM were ascertained from Europe, the Americas, Asia and Australia. Clinical data from referrals were recorded and stratified by the molecular aetiology of patients. RESULTS: 6q24 TNDM patients presented at a modal age of one day, with growth retardation and hyperglycaemia, irrespective of molecular aetiology. There was a positive correlation between age of presentation and gestational age, and a negative correlation between adjusted birthweight SD and age of remission. Congenital anomalies were significantly more frequent in patients with paternal uniparental disomy of chromosome 6 or hypomethylation of multiple imprinted loci defects than in those with 6q24 duplication or isolated hypomethylation defects. Patients with hypomethylation had an excess representation of assisted conception at 15%. CONCLUSIONS/INTERPRETATION: This, the largest case series of 6q24 TNDM published, refines and extends the clinical phenotype of the disorder and confirms its clinical divergence from other monogenic TNDM in addition to identifying previously unreported clinical differences between 6q24 subgroups.


Assuntos
Cromossomos Humanos Par 6 , Diabetes Mellitus/genética , Anormalidades Múltiplas/genética , Idade de Início , Estudos de Coortes , Metilação de DNA , Diabetes Mellitus/diagnóstico , Feminino , Estudos de Associação Genética , Impressão Genômica , Genótipo , Idade Gestacional , Humanos , Recém-Nascido , Doenças do Recém-Nascido/genética , Masculino , Fenótipo , Indução de Remissão , Dissomia Uniparental/genética
12.
Diabet Med ; 30(5): e197-200, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23320570

RESUMO

AIMS: Recessive PDX1 (IPF1) mutations are a rare cause of pancreatic agenesis, with three cases reported worldwide. A recent report described two cousins with a homozygous hypomorphic PDX1 mutation causing permanent neonatal diabetes with subclinical exocrine insufficiency. The aim of our study was to investigate the possibility of hypomorphic PDX1 mutations in a large cohort of patients with permanent neonatal diabetes and no reported pancreatic hypoplasia or exocrine insufficiency. METHODS: PDX1 was sequenced in 103 probands with isolated permanent neonatal diabetes in whom ABCC8, KCNJ11 and INS mutations had been excluded. RESULTS: Sequencing analysis identified biallelic PDX1 mutations in three of the 103 probands with permanent neonatal diabetes (2.9%). One proband and his affected brother were compound heterozygotes for a frameshift and a novel missense mutation (p.A34fsX191; c.98dupC and p.P87L; c.260C>T). The other two probands were homozygous for novel PDX1 missense mutations (p.A152G; c.455C>G and p.R176Q; c.527G>A). Both mutations affect highly conserved residues located within the homeobox domain. None of the four cases showed any evidence of exocrine pancreatic insufficiency, either clinically, or, where data were available, biochemically. In addition a heterozygous nonsense mutation (p.C18X; c.54C>A) was identified in a fourth case. CONCLUSIONS: This study demonstrates that recessive PDX1 mutations are a rare but important cause of isolated permanent neonatal diabetes in patients without pancreatic hypoplasia/agenesis. Inclusion of the PDX1 gene in mutation screening for permanent neonatal diabetes is recommended as a genetic diagnosis reveals the mode of inheritance, allows accurate estimation of recurrence risks and confirms the requirement for insulin treatment.


Assuntos
Diabetes Mellitus Tipo 1/genética , Glândulas Exócrinas/fisiopatologia , Proteínas de Homeodomínio/genética , Doenças do Recém-Nascido/genética , Mutação de Sentido Incorreto , Transativadores/genética , Feminino , Humanos , Hipoglicemiantes/uso terapêutico , Lactente , Recém-Nascido , Doenças do Recém-Nascido/sangue , Doenças do Recém-Nascido/fisiopatologia , Insulina/uso terapêutico , Masculino , Pâncreas/anormalidades , Pancreatopatias/congênito , Pancreatopatias/genética
14.
Artigo em Inglês | MEDLINE | ID: mdl-24616771

RESUMO

BACKGROUND: Congenital hyperinsulinism (CHI) is a rare genetic disorder characterised by inappropriate insulin secretion in the face of severe hypoglycaemia. There are two histological subtypes of CHI namely diffuse and focal. Diffuse CHI is most common due to recessive mutations in ABCC8/KCNJ11 (which encode the SUR/KIR6.2 components of the pancreatic ß-cell KATP channel) whereas focal CHI is due to a paternally inherited ABCC8/KCNJ11 mutation and somatic loss of heterozygosity for the 11p allele inside the focal lesion. Fluorine-18-l-dihydroxyphenylalanine positron emission tomography/computed tomography ((18)F-DOPA-PET/CT) is used in the pre-operative localisation of focal lesions prior to surgery. Diffuse CHI if medically unresponsive will require a near total pancreatectomy whereas focal CHI will only require a limited lesionectomy, thus curing the patient from the hypoglycaemia. AIMS: To report the first case of genetically confirmed CHI in Singapore from a heterozygous paternally inherited ABCC8 mutation. METHODS/RESULTS: A term male infant presented with severe hyperinsulinaemic hypoglycaemia (HH) after birth and failed medical treatment with diazoxide and octreotide. Genetic testing (paternally inherited mutation in ABCC8/p.D1472N) suggested focal disease, but due to the unavailability of (18)F-DOPA-PET/CT to confirm focal disease, a partial pancreatectomy was performed. Interestingly, histology of the resected pancreatic tissue showed changes typical of diffuse disease. CONCLUSION: Heterozygous paternally inherited ABCC8/KCNJ11 mutations can lead to diffuse or focal CHI. LEARNING POINTS: HH is a cause of severe hypoglycaemia in the newborn period.Paternal mutations in ABCC8/KCNJ11 can lead to diffuse or focal disease.(18)F-DOPA-PET/CT scan is the current imaging of choice for localising focal lesions.Gallium-68 tetra-aza-cyclododecane-N N'N″N-‴-tetra-acetate octreotate PET scan is not a useful imaging tool for localising focal lesions.The molecular mechanism by which a heterozygous ABCC8 mutation leads to diffuse disease is currently unclear.Focal lesions are curable by lesionectomy and so genetic studies in patients with HH must be followed by imaging using (18)F-DOPA-PET/CT scan.

15.
J Perinatol ; 32(8): 645-7, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22842804

RESUMO

Neonatal/infancy-onset diabetes mellitus is a monogenic form of diabetes with onset within 6 months of age. Two distinct types of neonatal diabetes mellitus have been recognized: permanent and transient. Mutations within the K(+)ATP channel and insulin genes are found in most patients with permanent diabetes mellitus. There have been several reports of the successful transition from insulin to sulfonylurea agents in patients with permanent diabetes mellitus caused by mutations in the KCNJ11 gene. We report on a term female neonate with a novel missense mutation, p.P1199L, in the ABCC8 gene that encodes the sulfonylurea receptor 1 whose treatment was successfully converted from insulin to sulfonylurea.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Diabetes Mellitus/tratamento farmacológico , Hipoglicemiantes/uso terapêutico , Canais de Potássio Corretores do Fluxo de Internalização/genética , Receptores de Droga/genética , Compostos de Sulfonilureia/uso terapêutico , Diabetes Mellitus/genética , Feminino , Humanos , Recém-Nascido , Mutação de Sentido Incorreto , Receptores de Sulfonilureias
16.
Diabetologia ; 55(9): 2381-5, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22660720

RESUMO

AIMS: The gene SLC2A2 encodes GLUT2, which is found predominantly in pancreas, liver, kidney and intestine. In mice, GLUT2 is the major glucose transporter into pancreatic beta cells, and biallelic Slc2a2 inactivation causes lethal neonatal diabetes. The role of GLUT2 in human beta cells is controversial, and biallelic SLC2A2 mutations cause Fanconi-Bickel syndrome (FBS), with diabetes rarely reported. We investigated the potential role of GLUT2 in the neonatal period by testing whether SLC2A2 mutations can present with neonatal diabetes before the clinical features of FBS appear. METHODS: We studied SLC2A2 in patients with transient neonatal diabetes mellitus (TNDM; n = 25) or permanent neonatal diabetes mellitus (PNDM; n = 79) in whom we had excluded the common genetic causes of neonatal diabetes, using a combined approach of sequencing and homozygosity mapping. RESULTS: Of 104 patients, five (5%) were found to have homozygous SLC2A2 mutations, including four novel mutations (S203R, M376R, c.963+1G>A, F114LfsX16). Four out of five patients with SLC2A2 mutations presented with isolated diabetes and later developed features of FBS. Four out of five patients had TNDM (16% of our TNDM cohort of unknown aetiology). One patient with PNDM remains on insulin at 28 months. CONCLUSIONS: SLC2A2 mutations are an autosomal recessive cause of neonatal diabetes that should be considered in consanguineous families or those with TNDM, after excluding common causes, even in the absence of features of FBS. The finding that patients with homozygous SLC2A2 mutations can have neonatal diabetes supports a role for GLUT2 in the human beta cell.


Assuntos
Diabetes Mellitus/genética , Síndrome de Fanconi/genética , Transportador de Glucose Tipo 2/genética , Insulina/metabolismo , Mutação , Sequência de Bases , Erros Inatos do Metabolismo dos Carboidratos/genética , Primers do DNA , Diabetes Mellitus/diagnóstico , Síndrome de Fanconi/diagnóstico , Feminino , Humanos , Recém-Nascido , Insulina/genética , Secreção de Insulina , Masculino
17.
Diabetologia ; 55(1): 123-7, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21989597

RESUMO

AIMS/HYPOTHESIS: The ABCC8 gene encodes the sulfonylurea receptor 1 (SUR1) subunit of the pancreatic beta cell ATP-sensitive potassium (K(ATP)) channel. Inactivating mutations cause congenital hyperinsulinism (CHI) and activating mutations cause transient neonatal diabetes (TNDM) or permanent neonatal diabetes (PNDM) that can usually be treated with sulfonylureas. Sulfonylurea sensitivity is also a feature of HNF1A and HNF4A MODY, but patients referred for genetic testing with clinical features of these types of diabetes do not always have mutations in the HNF1A/4A genes. Our aim was to establish whether mutations in the ABCC8 gene cause MODY that is responsive to sulfonylurea therapy. METHODS: We sequenced the ABCC8 gene in 85 patients with a BMI <30 kg/m², no family history of neonatal diabetes and who were deemed sensitive to sulfonylureas by the referring clinician or were sulfonylurea-treated. All had tested negative for mutations in the HNF1A and HNF4A genes. RESULTS: ABCC8 mutations were found in seven of the 85 (8%) probands. Four patients were heterozygous for previously reported mutations and four novel mutations, E100K, G214R, Q485R and N1245D, were identified. Only four probands fulfilled MODY criteria, with two diagnosed after 25 years and one patient, who had no family history of diabetes, as a result of a proven de novo mutation. CONCLUSIONS/INTERPRETATION: ABCC8 mutations can cause MODY in patients whose clinical features are similar to those with HNF1A/4A MODY. Therefore, sequencing of ABCC8 in addition to the known MODY genes should be considered if such features are present, to facilitate optimal clinical management of these patients.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Diabetes Mellitus Tipo 2/genética , Heterozigoto , Mutação , Canais de Potássio Corretores do Fluxo de Internalização/genética , Receptores de Droga/genética , Transportadores de Cassetes de Ligação de ATP/química , Adulto , Substituição de Aminoácidos , Estudos de Coortes , Análise Mutacional de DNA , Diabetes Mellitus Tipo 2/tratamento farmacológico , Feminino , Estudos de Associação Genética , Humanos , Hipoglicemiantes/uso terapêutico , Leucócitos/metabolismo , Masculino , Repetições de Microssatélites , Pessoa de Meia-Idade , Linhagem , Canais de Potássio Corretores do Fluxo de Internalização/química , Subunidades Proteicas/química , Subunidades Proteicas/genética , Receptores de Droga/química , Compostos de Sulfonilureia/uso terapêutico , Receptores de Sulfonilureias , Reino Unido , Adulto Jovem
18.
Diabetologia ; 54(10): 2575-83, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21674179

RESUMO

AIMS/HYPOTHESIS: Dominantly acting loss-of-function mutations in the ABCC8/KCNJ11 genes can cause mild medically responsive hyperinsulinaemic hypoglycaemia (HH). As controversy exists over whether these mutations predispose to diabetes in adulthood we investigated the prevalence of diabetes in families with dominantly inherited ATP-sensitive potassium (K(ATP)) channel mutations causing HH in the proband. METHODS: We studied the phenotype of 30 mutation carriers (14 children and 16 adults) from nine families with dominant ABCC8/KCNJ11 mutations. Functional consequences of six novel missense mutations were examined by reconstituting the K(ATP) channel in human embryonic kidney 293 (HEK293) cells and evaluating the effect of drugs and metabolic poisoning on the channels using the (86)Rb flux assay. RESULTS: The mutant channels all showed a lack of (86)Rb efflux on exposure to the channel agonist diazoxide or metabolic inhibition. In the families, dominant ABCC8/KCNJ11 mutations were associated with increased birthweight (median + 1.56 SD score [SDS]). Fourteen children had HH and five adults were reported with HH or hypoglycaemic episodes (63%). Progression from hypoglycaemia to diabetes mellitus occurred in two individuals. Eight adults had a history of gestational diabetes in multiple pregnancies or were diabetic (diagnosed at a median age of 31 years). Within these families, none of the 19 adults who were not carriers of the ABCC8/KCNJ11 mutation was known to be diabetic. CONCLUSIONS/INTERPRETATION: The phenotype associated with dominant ABCC8/KCNJ11 mutations ranges from asymptomatic macrosomia to persistent HH in childhood. In adults, it may also be an important cause of dominantly inherited early-onset diabetes mellitus.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Diabetes Mellitus/etiologia , Diabetes Mellitus/genética , Hiperinsulinismo/etiologia , Hiperinsulinismo/genética , Hipoglicemia/etiologia , Hipoglicemia/genética , Canais de Potássio Corretores do Fluxo de Internalização/genética , Receptores de Droga/genética , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Receptores de Sulfonilureias , Adulto Jovem
19.
Eur J Endocrinol ; 164(5): 733-40, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21378087

RESUMO

OBJECTIVE: In children with congenital hyperinsulinism (CHI), K(ATP) channel genes (ABCC8 and KCNJ11) can be screened rapidly for potential pathogenic mutations. We aimed to assess the contribution of rapid genetic testing to the clinical management of CHI. DESIGN: Follow-up observational study at two CHI referral hospitals. METHODS: Clinical outcomes such as subtotal pancreatectomy, (18)F-Dopa positron emission tomography-computed tomography (PET-CT) scanning, stability on medical treatment and remission were assessed in a cohort of 101 children with CHI. RESULTS: In total, 32 (32%) children had pathogenic mutations in K(ATP) channel genes (27 in ABCC8 and five in KCNJ11), of which 11 (34%) were novel. In those negative at initial screening, other mutations (GLUD1, GCK, and HNF4A) were identified in three children. Those with homozygous/compound heterozygous ABCC8/KCNJ11 mutations were more likely to require a subtotal pancreatectomy CHI (7/10, 70%). Those with paternal heterozygous mutations were investigated with (18)F-Dopa PET-CT scanning and 7/13 (54%) had a focal lesionectomy, whereas four (31%) required subtotal pancreatectomy for diffuse CHI. Those with maternal heterozygous mutations were most likely to achieve remission (5/5, 100%). In 66 with no identified mutation, 43 (65%) achieved remission, 22 (33%) were stable on medical treatment and only one child required a subtotal pancreatectomy. CONCLUSIONS: Rapid genetic analysis is important in the management pathway of CHI; it provides aetiological confirmation of the diagnosis, indicates the likely need for a subtotal pancreatectomy and identifies those who require (18)F-Dopa PET-CT scanning. In the absence of a mutation, reassurance of a favourable outcome can be given early in the course of CHI.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Hiperinsulinismo Congênito/diagnóstico , Hiperinsulinismo Congênito/genética , Mutação/genética , Canais de Potássio Corretores do Fluxo de Internalização/genética , Receptores de Droga/genética , Adolescente , Criança , Pré-Escolar , Estudos de Coortes , Hiperinsulinismo Congênito/terapia , Gerenciamento Clínico , Feminino , Seguimentos , Humanos , Lactente , Masculino , Receptores de Sulfonilureias , Fatores de Tempo
20.
Clin Genet ; 79(6): 582-7, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20573158

RESUMO

Recessive inactivating mutations in the ABCC8 and KCNJ11 genes encoding the adenosine triphosphate-sensitive potassium (K(ATP)) channel subunit sulphonylurea receptor 1 (SUR1) and inwardly rectifying potassium channel subunit (Kir6.2) are the most common cause of hyperinsulinaemic hypoglycaemia (HH). Most of these patients do not respond to treatment with the (K(ATP)) channel agonist diazoxide. Dominant inactivating ABCC8 and KCNJ11 mutations are less frequent, but are usually associated with a milder form of hypoglycaemia that is responsive to diazoxide therapy. We studied five patients from four families with HH who were unresponsive to diazoxide and required a near total pancreatectomy. Mutations in KCNJ11 and ABCC8 were sought by sequencing and dosage analysis. Three novel heterozygous ABCC8 mis-sense mutations (G1485E, D1506E and M1514K) were identified in four probands. All the mutations affect residues located within the Nucleotide Binding Domain 2 of the SUR1 subunit. Testing of family members showed that the mutations had arisen de novo with dominant inheritance in one pedigree. This study extends the clinical phenotype associated with dominant (K(ATP)) channel mutations to include severe congenital HH requiring near total pancreatectomy in addition to a milder form of diazoxide responsive hypoglycaemia. The identification of dominant vs recessive mutations does not predict clinical course but it is important for estimating the risk of HH in future siblings and offspring.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Hiperinsulinismo Congênito/genética , Genes Dominantes , Mutação de Sentido Incorreto , Canais de Potássio Corretores do Fluxo de Internalização/genética , Receptores de Droga/genética , Hiperinsulinismo Congênito/diagnóstico , Hiperinsulinismo Congênito/terapia , Análise Mutacional de DNA , Resistência a Medicamentos , Feminino , Fármacos Gastrointestinais/uso terapêutico , Estudos de Associação Genética , Marcadores Genéticos , Haplótipos , Humanos , Recém-Nascido , Masculino , Octreotida/uso terapêutico , Pâncreas/patologia , Pâncreas/cirurgia , Pancreatectomia , Receptores de Sulfonilureias
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...