Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Clin Endocrinol Metab ; 100(10): E1362-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26259131

RESUMO

CONTEXT: GLIS3 (GLI-similar 3) is a member of the GLI-similar zinc finger protein family encoding for a nuclear protein with 5 C2H2-type zinc finger domains. The protein is expressed early in embryogenesis and plays a critical role as both a repressor and activator of transcription. Human GLIS3 mutations are extremely rare. OBJECTIVE: The purpose of this article was determine the phenotypic presentation of 12 patients with a variety of GLIS3 mutations. METHODS: GLIS3 gene mutations were sought by PCR amplification and sequence analysis of exons 1 to 11. Clinical information was provided by the referring clinicians and subsequently using a questionnaire circulated to gain further information. RESULTS: We report the first case of a patient with a compound heterozygous mutation in GLIS3 who did not present with congenital hypothyroidism. All patients presented with neonatal diabetes with a range of insulin sensitivities. Thyroid disease varied among patients. Hepatic and renal disease was common with liver dysfunction ranging from hepatitis to cirrhosis; cystic dysplasia was the most common renal manifestation. We describe new presenting features in patients with GLIS3 mutations, including craniosynostosis, hiatus hernia, atrial septal defect, splenic cyst, and choanal atresia and confirm further cases with sensorineural deafness and exocrine pancreatic insufficiency. CONCLUSION: We report new findings within the GLIS3 phenotype, further extending the spectrum of abnormalities associated with GLIS3 mutations and providing novel insights into the role of GLIS3 in human physiological development. All but 2 of the patients within our cohort are still alive, and we describe the first patient to live to adulthood with a GLIS3 mutation, suggesting that even patients with a severe GLIS3 phenotype may have a longer life expectancy than originally described.


Assuntos
Doenças Ósseas/genética , Hipotireoidismo Congênito/genética , Deficiências do Desenvolvimento/genética , Diabetes Mellitus/genética , Resistência à Insulina/genética , Hepatopatias/genética , Fenótipo , Fatores de Transcrição/genética , Doenças Ósseas/congênito , Proteínas de Ligação a DNA , Diabetes Mellitus/congênito , Feminino , Humanos , Lactente , Recém-Nascido , Hepatopatias/congênito , Masculino , Proteínas Repressoras , Transativadores
2.
Diabet Med ; 30(1): 114-7, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22587559

RESUMO

AIMS: Hepatocyte nuclear factor 1ß (HNF1B) mutations cause a syndrome of renal cysts and diabetes, with whole gene deletions accounting for approximately 50% of cases. The severity of the renal phenotype is variable, from enlarged cystic kidneys incompatible with life to normal renal development and function. We investigated the prevalence of HNF1B deletions in patients with diabetes but no known renal disease. METHODS: We tested 461 patients with familial diabetes diagnosed before 45 years, including 258 probands who met clinical criteria for maturity-onset diabetes of the young (two generations affected and at least one family member diagnosed under 25 years). A fluorescent polymerase chain reaction assay was used to analyse two intragenic polymorphic HNF1B markers and identify heterozygous patients who therefore did not have whole gene deletions. Those patients homozygous for both markers were then tested for an HNF1B deletion using multiplex ligation-dependent probe amplification. RESULTS: Heterozygous HNF1B intragenic polymorphisms were identified in 337/461 subjects. Multiplex ligation-dependent probe amplification analysis showed an HNF1B gene deletion in three of the remaining 124 probands, all of whom met the criteria for maturity-onset diabetes of the young. Testing of their relatives identified three additional deletion carriers and ultrasound scanning showed renal developmental abnormalities in three of these six patients. CONCLUSIONS: We estimate that HNF1B mutations account for < 1% of cases of maturity-onset diabetes of the young. Although HNF1B mutations are a rare cause of diabetes in the absence of known renal disease, a genetic diagnosis of renal cysts and diabetes syndrome is important as it raises the possibility of subclinical renal disease and the 50% risk of renal cysts and diabetes syndrome in the patient's offspring.


Assuntos
Diabetes Mellitus Tipo 2/genética , Nefropatias Diabéticas/genética , Deleção de Genes , Fator 1-beta Nuclear de Hepatócito/genética , Polimorfismo Genético/genética , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Testes Genéticos , Homozigoto , Humanos , Lactente , Doenças Renais Císticas/genética , Masculino , Pessoa de Meia-Idade , Adulto Jovem
3.
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
4.
J Perinatol ; 31(8): 567-70, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21796147

RESUMO

Permanent neonatal diabetes was previously assumed to require insulin injection or infusion for life. Recently, permanent neonatal diabetes resulting from mutations in the two protein subunits of the adenosine triphosphate-sensitive potassium channel (Kir6.2 and SUR1) has proven to be successfully treatable with high doses of sulfonylureas rather than insulin. Many patients with these mutations first develop hyperglycemia in the nursery or intensive care unit. The awareness of the neonatolgist of this entity can have dramatic effects on the long-term care and quality of life of these patients and their families. In this study, we present the experience of our center, highlighting aspects relevant to neonatal diagnosis and treatment.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/genética , Hipoglicemiantes/uso terapêutico , Canais de Potássio Corretores do Fluxo de Internalização/genética , Compostos de Sulfonilureia/uso terapêutico , Transportadores de Cassetes de Ligação de ATP/genética , Adulto , Diabetes Mellitus Tipo 1/congênito , Feminino , Humanos , Hipoglicemiantes/administração & dosagem , Lactente , Recém-Nascido , Insulina/administração & dosagem , Insulina/uso terapêutico , Mutação de Sentido Incorreto , Qualidade de Vida , Receptores de Droga/genética , Receptores de Sulfonilureias
5.
Diabet Med ; 28(9): 1028-33, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21395678

RESUMO

AIM: Maturity-onset diabetes of the young is a monogenic form of familial, young-onset diabetes. It is rare (∼1% diabetes) and may be misdiagnosed as Type 1 diabetes and inappropriately treated with insulin. Type 1 diabetes is characterized by the presence of islet autoantibodies, including glutamate decarboxylase (GAD) and islet antigen-2 (IA-2) antibodies. The prevalence of islet autoantibodies is unknown in maturity-onset diabetes of the young and may have the potential to differentiate this form of diabetes from Type 1 diabetes. The aim of this study was to determine the prevalence of GAD and IA-2 antibodies in patients with maturity-onset diabetes of the young and Type 1 diabetes. METHODS: We measured plasma GAD and IA-2 antibodies in 508 patients with the most common forms of maturity-onset diabetes of the young (GCK: n = 227; HNF1A: n = 229; HNF4A: n = 52) and 98 patients with newly diagnosed Type 1 diabetes (diagnosed < 6 months). Autoantibodies were considered positive if ≥ 99th centile of 500 adult control subjects. RESULTS: GAD and/or IA-2 antibodies were present in 80/98 (82%) patients with Type 1 diabetes and 5/508 (< 1%) patients with maturity-onset diabetes of the young. In the cohort with Type 1 diabetes, both GAD and IA-2 antibodies were detected in 37.8% of patients, GAD only in 24.5% and IA-2 only in 19.4%. All five patients with maturity-onset diabetes of the young with detectable antibodies had GAD antibodies and none had detectable IA-2 antibodies. CONCLUSION: The prevalence of GAD and IA-2 antibodies in maturity-onset diabetes of the young is the same as in control subjects (< 1%). The finding of islet autoantibodies, especially IA-2 antibodies, makes the diagnosis of maturity-onset diabetes of the young very unlikely and genetic testing should only be performed if other clinical characteristics strongly suggest this form of diabetes rather than Type 1 diabetes. This supports routine islet autoantibody testing before proceeding to more expensive molecular genetic testing.


Assuntos
Autoanticorpos/sangue , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 2/sangue , Glutamato Descarboxilase/sangue , Fatores Imunológicos/sangue , Adulto , Autoanticorpos/imunologia , Estudos de Coortes , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/imunologia , Diagnóstico Diferencial , Feminino , Glutamato Descarboxilase/imunologia , Humanos , Fatores Imunológicos/genética , Fatores Imunológicos/imunologia , Masculino , Programas de Rastreamento , Pessoa de Meia-Idade , Prevalência
6.
Eur J Endocrinol ; 164(3): 437-43, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21139041

RESUMO

INTRODUCTION: Mutations in the GLI-similar 3 (GLIS3) gene encoding the transcription factor GLIS3 are a rare cause of neonatal diabetes and congenital hypothyroidism with six affected cases from three families reported to date. Additional features, described previously, include congenital glaucoma, hepatic fibrosis, polycystic kidneys, developmental delay and facial dysmorphism. SUBJECTS: We report two new cases from unrelated families with distinct novel homozygous partial GLIS3 deletions. Both patients presented with neonatal diabetes mellitus, severe resistant hypothyroidism in the presence of elevated thyroglobulin and normal thyroid anatomy, degenerative liver disease, cystic renal dysplasia, recurrent infections and facial dysmorphism. These novel mutations have also resulted in osteopenia, bilateral sensorineural deafness and pancreatic exocrine insufficiency, features that have not previously been associated with GLIS3 mutations. Gene dosage analysis showed that the parents were carriers of a deletion encompassing exons 1-2 (case 1) or exons 1-4 (case 2) of the 11 exon gene. Genome-wide SNP analysis did not reveal a common ancestral GLIS3 haplotype in patient 2. CONCLUSIONS: Our results confirm partial gene deletions as the most common type of GLIS3 mutations, accounting for four of five families identified to date. We propose that mutations in GLIS3 lead to a wider clinical phenotype than previously recognised. We also report the first case of a recessive GLIS3 mutation causing neonatal diabetes and congenital hypothyroidism in a child from a non-consanguineous pedigree, highlighting the importance of molecular genetic testing in any patient with this phenotype.


Assuntos
Mutação , Fenótipo , Fatores de Transcrição/genética , Hipotireoidismo Congênito/genética , Proteínas de Ligação a DNA , Diabetes Mellitus/genética , Éxons/genética , Feminino , Dosagem de Genes/genética , Haplótipos/genética , Humanos , Recém-Nascido , Masculino , Proteínas Repressoras , Transativadores
7.
Clin Pharmacol Ther ; 89(2): 210-6, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21178985

RESUMO

SLCO1B1 gene variants are associated with severe statin-induced myopathy. We examined whether these variants are also associated with general statin intolerance in a large population of patients with type 2 diabetes receiving statins as part of routine clinical care. A total of 4,196 individuals were genotyped for rs4149056 (Val174Ala) and rs2306283 (Asp130Asn). Intolerance was defined by serum biochemistry and also by discontinuation, switching, or reduction in dose of the prescribed statin drug. Ala174 was associated with higher intolerance (odds ratio = 2.05, P = 0.043), whereas Asp130 was associated with lower intolerance (odds ratio = 0.71, P = 0.026). Ala174 was associated with a lower low-density lipoprotein cholesterol (LDLc) response to statins (P = 0.01) whereas 130D was associated with a greater LDLc response to statins (P = 0.048), as previously reported; however, this association was no longer present when data for statin-intolerant individuals were removed from the analysis. This study suggests that common genetic variants selected for an extreme phenotype of statin-induced myopathy also predispose to more common milder statin intolerance and may, for this reason, impact lipid-lowering efficacy.


Assuntos
Diabetes Mellitus Tipo 2/genética , Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Transportadores de Ânions Orgânicos/genética , Idoso , LDL-Colesterol/sangue , Feminino , Genótipo , Humanos , Transportador 1 de Ânion Orgânico Específico do Fígado , Modelos Logísticos , Masculino , Pessoa de Meia-Idade
8.
Diabet Med ; 27(2): 157-61, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20546258

RESUMO

AIMS: To investigate all-cause and cardiovascular mortality in subjects with diabetes caused by a mutation in the hepatocyte nuclear factor 1alpha gene (HNF1A). METHODS: We identified 39 British families with HNF1A mutations. Consenting individuals were asked details of age and cause of death of parents and siblings. Copies of death certificates were requested from the family or were obtained via the Offices for National Statistics. RESULTS: Data were collated on 241 control subjects and 153 mutation carriers. Of those who died, 66% of mutation carriers died from a cardiovascular-related illness compared with 43% of control subjects (P = 0.02). Family members with HNF1A mutations died at a younger age than familial control subjects [all-cause hazard ratio, adjusting for sex and smoking status: 1.9 (95% confidence interval 1.2, 2.9, P = 0.006; cardiovascular hazard ratio: 2.3, confidence interval 1.3, 4.2, P = 0.006)]. CONCLUSIONS: We have shown that individuals known to have diabetes caused by a mutation in the HNF1A gene have an increased risk of cardiovascular mortality compared with their unaffected family members. As with other forms of diabetes, consideration should be given to early statin therapy despite a seemingly protective lipid profile.


Assuntos
Doenças Cardiovasculares/mortalidade , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/genética , Fator 1-alfa Nuclear de Hepatócito/genética , Mortalidade , Mutação/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Doenças Cardiovasculares/etiologia , Análise Mutacional de DNA , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 1/mortalidade , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/mortalidade , Família , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Risco , Análise de Sequência de DNA , Análise de Sobrevida
9.
Diabetologia ; 53(12): 2504-8, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20499044

RESUMO

AIMS/HYPOTHESIS: Maturity-onset diabetes of the young is frequently misdiagnosed as type 1 or type 2 diabetes. A correct diagnosis of MODY is important for determining treatment, but can only be confirmed by molecular genetic testing. We aimed to compare the regional distribution of confirmed MODY cases in the UK and to estimate the minimum prevalence. METHODS: UK referrals for genetic testing in 2,072 probands and 1,280 relatives between 1996 and 2009 were examined by region, country and test result. Referral rate and prevalence were calculated using UK Census 2001 figures. RESULTS: MODY was confirmed in 1,177 (35%) patients, with HNF1A (52%) and GCK mutations (32%) being most frequent in probands confirmed with MODY. There was considerable regional variation in proband referral rates (from <20 per million in Wales and Northern Ireland to >50 per million for South West England and Scotland) and patients diagnosed with MODY (5.3 per million in Northern Ireland, 48.9 per million in South West England). Referral rates and confirmed cases were highly correlated (r = 0.96, p < 0.0001). The minimum prevalence of MODY was estimated to be 108 cases per million. CONCLUSIONS/INTERPRETATION: Assuming this minimal prevalence throughout the UK then >80% of MODY is not diagnosed by molecular testing. The marked regional variation in the prevalence of confirmed MODY directly results from differences in referral rates. This could reflect variation in awareness of MODY or unequal access to genetic testing. Increased referral for diagnostic testing is required if the majority of MODY patients are to have the genetic diagnosis necessary for optimal treatment.


Assuntos
Diabetes Mellitus Tipo 2/diagnóstico , Programas de Rastreamento/normas , Adolescente , Adulto , Idade de Início , Criança , Pré-Escolar , Diabetes Mellitus Tipo 2/epidemiologia , Diabetes Mellitus Tipo 2/genética , Inglaterra/epidemiologia , Família , Testes Genéticos/estatística & dados numéricos , Acessibilidade aos Serviços de Saúde/normas , Acessibilidade aos Serviços de Saúde/estatística & dados numéricos , Humanos , Lactente , Programas de Rastreamento/estatística & dados numéricos , Irlanda do Norte/epidemiologia , Prevalência , Encaminhamento e Consulta/estatística & dados numéricos , Escócia/epidemiologia , País de Gales/epidemiologia , Adulto Jovem
10.
Diabetologia ; 51(5): 802-10, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18335204

RESUMO

AIMS/HYPOTHESIS: Heterozygous activating mutations in the pancreatic ATP-sensitive K+ channel cause permanent neonatal diabetes mellitus (PNDM). This results from a decrease in the ability of ATP to close the channel, which thereby suppresses insulin secretion. PNDM mutations that cause a severe reduction in ATP inhibition may produce additional symptoms such as developmental delay and epilepsy. We identified a heterozygous mutation (L164P) in the pore-forming (Kir6.2) subunit of the channel in three unrelated patients and examined its functional effects. METHODS: The patients (currently aged 2, 8 and 20 years) developed diabetes shortly after birth. The two younger patients attempted transfer to sulfonylurea therapy but were unsuccessful (up to 1.1 mg kg(-1) day(-1)). They remain insulin dependent. None of the patients displayed neurological symptoms. Functional properties of wild-type and mutant channels were examined by electrophysiology in Xenopus oocytes. RESULTS: Heterozygous (het) and homozygous L164P K(ATP) channels showed a marked reduction in channel inhibition by ATP. Consistent with its predicted location within the pore, L164P enhanced the channel open state, which explains the reduction in ATP sensitivity. HetL164P currents exhibited greatly increased whole-cell currents that were unaffected by sulfonylureas. This explains the inability of sulfonylureas to ameliorate the diabetes of affected patients. CONCLUSIONS/INTERPRETATION: Our results provide the first demonstration that mutations such as L164P, which produce a severe reduction in ATP sensitivity, do not inevitably cause developmental delay or neurological problems. However, the neonatal diabetes of these patients is unresponsive to sulfonylurea therapy. Functional analysis of PNDM mutations can predict the sulfonylurea response.


Assuntos
Diabetes Mellitus/genética , Doenças do Recém-Nascido/genética , Mutação , Canais de Potássio Corretores do Fluxo de Internalização/genética , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Adulto , Substituição de Aminoácidos , Animais , Criança , Pré-Escolar , DNA/sangue , DNA/genética , DNA/isolamento & purificação , Diabetes Mellitus/tratamento farmacológico , Feminino , Hemoglobinas Glicadas/análise , Humanos , Hipoglicemiantes/uso terapêutico , Recém-Nascido , Modelos Moleculares , Oócitos/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/química , Canais de Potássio Corretores do Fluxo de Internalização/efeitos dos fármacos , Conformação Proteica , Compostos de Sulfonilureia/uso terapêutico , Xenopus laevis
11.
Diabetologia ; 50(11): 2313-7, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17828387

RESUMO

AIMS/HYPOTHESIS: Heterozygous mutations of glucokinase (GCK) and hepatocyte nuclear factor-1 alpha (HNF1A; also known as hepatic transcription factor 1 [TCF1]) genes are the most common cause of MODY. Genomic deletions of the HNF1B (also known as TCF2) gene have recently been shown to account for one third of mutations causing renal cysts and diabetes syndrome. We investigated the prevalence of partial and whole gene deletions in UK patients meeting clinical criteria for GCK or HNF-1alpha/-4alpha MODY and in whom no mutation had been identified by sequence analysis. METHODS: A multiplex ligation-dependent probe amplification (MLPA) assay was developed using synthetic oligonucleotide probes for 30 exons of the GCK, HNF1A and HNF4A genes. RESULTS: Partial or whole gene deletions were identified in 1/29 (3.5%) probands using the GCK MLPA assay and 4/60 (6.7%) of probands using the HNF1A/-4A MLPA assay. Four different deletions were detected: GCK exon 2, HNF1A exon 1, HNF1A exons 2 to 10 and HNF1A exons 1 to 10. An additional Danish pedigree with evidence of linkage to HNF1A had a deletion of exons 2 to 10. Testing other family members confirmed co-segregation of the deletion mutations with diabetes in the pedigrees. CONCLUSIONS/INTERPRETATION: Large deletions encompassing whole exons can cause GCK or HNF-1alpha MODY and will not be detected by sequencing. Gene dosage assays, such as MLPA, are a useful adjunct to sequence analysis when a diagnosis of MODY is strongly suspected.


Assuntos
Diabetes Mellitus Tipo 2/genética , Deleção de Genes , Glucoquinase/genética , Fator 1-alfa Nuclear de Hepatócito/genética , Adolescente , Adulto , Idade de Início , Pré-Escolar , Feminino , Glucoquinase/deficiência , Fator 1-alfa Nuclear de Hepatócito/deficiência , Humanos , Masculino , Linhagem , Fenótipo
12.
Diabet Med ; 23(3): 331-4, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16492220

RESUMO

AIMS: Familial clustering of diabetic nephropathy in patients with Type 2 diabetes suggests that inherited factors predispose to diabetic nephropathy, but the nature of these factors is uncertain. The aim of the study was to compare the prevalence of known risk factors for nephropathy in non-diabetic offspring of Type 2 diabetic patients with and without nephropathy and in control subjects. METHODS: Three groups of patients were recruited with 40 or 41 subjects in each group. These were subjects having one Type 2 diabetic parent with nephropathy (DN); subjects having one parent with Type 2 diabetes without nephropathy (DnoN), and non-diabetic unrelated control subjects with no personal or parental history of diabetes (Control subjects). RESULTS: The median (interquartile range) albumin/creatinine ratio (ACR) was 1.40 (0.96-2.90) mg/mmol in DN; 0.94 (0.50-1.46) mg/mmol in DnoN and 1.22 (0.66-1.83) mg/mmol in Controls (ANOVA: P = 0.03). ACR was higher in group DN than in DnoN (P < 0.006) and in Control subjects (P < 0.03), but there was no difference between DnoN and Control subjects. Twenty-four-hour ambulatory blood pressure monitoring showed mean daytime systolic blood pressure to be significantly higher in group DN than in DnoN (P < 0.02) or Control subjects (P < 0.01) (ANOVA: P = 0.004). Fasting insulin, HOMA-IR, interleukin-6 (IL-6) and C-reactive protein (CRP) were similar in the three groups. CONCLUSION: Our data provide further evidence that genetic factors are important in determining urinary albumin excretion and renal disease associated with Type 2 diabetes and suggest that genes that affect systemic arterial blood pressure but not those relating to insulin resistance or inflammation are likely to be implicated.


Assuntos
Diabetes Mellitus Tipo 2/genética , Nefropatias Diabéticas/genética , Saúde da Família , Adulto , Albuminúria/genética , Glicemia/análise , Pressão Sanguínea/fisiologia , Proteína C-Reativa/análise , Creatinina/sangue , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/urina , Nefropatias Diabéticas/sangue , Nefropatias Diabéticas/urina , Feminino , Humanos , Insulina/metabolismo , Interleucina-6/sangue , Masculino , Pais , Fatores de Risco
13.
J Med Genet ; 43(1): 84-90, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15930087

RESUMO

BACKGROUND: Hepatocyte nuclear factor-1 beta (HNF-1beta) is a widely distributed transcription factor which plays a critical role in embryonic development of the kidney, pancreas, liver, and Mullerian duct. Thirty HNF-1beta mutations have been reported in patients with renal cysts and other renal developmental disorders, young-onset diabetes, pancreatic atrophy, abnormal liver function tests, and genital tract abnormalities. METHODS: We sequenced the HNF-1beta gene in 160 unrelated subjects with renal disease, 40% of whom had a personal/family history of diabetes. RESULTS: Twenty three different heterozygous HNF-1beta mutations were identified in 23/160 subjects (14%), including 10 novel mutations (V61G, V110G, S148L, K156E, Q176X, R276Q, S281fsinsC, R295P, H324fsdelCA, Q470X). Seven (30%) cases were proven to be due to de novo mutations. Renal cysts were found in 19/23 (83%) patients (four with glomerulocystic kidney disease, GCKD) and diabetes in 11/23 (48%, while three other families had a family history of diabetes. Only 26% of families met diagnostic criteria for maturity-onset diabetes of the young (MODY) but 39% had renal cysts and diabetes (RCAD). We found no clear genotype/phenotype relationships. CONCLUSION: We report the largest series to date of HNF-1beta mutations and confirm HNF-1beta mutations as an important cause of renal disease. Despite the original description of HNF-1beta as a MODY gene, a personal/family history of diabetes is often absent and the most common clinical manifestation is renal cysts. Molecular genetic testing for HNF-1beta mutations should be considered in patients with unexplained renal cysts (including GCKD), especially when associated with diabetes, early-onset gout, or uterine abnormalities.


Assuntos
Fator 1-beta Nuclear de Hepatócito/genética , Mutação/genética , Fenótipo , Adolescente , Adulto , Criança , Pré-Escolar , Éxons/genética , Fator 1-beta Nuclear de Hepatócito/química , Humanos , Lactente , Nefropatias/genética , Linhagem
14.
Diabetologia ; 48(10): 2013-7, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16132956

RESUMO

AIMS/HYPOTHESIS: The proinflammatory cytokine TNF-alpha has been implicated in the pathogenesis of insulin resistance and type 2 diabetes, and variation in the gene encoding TNF-alpha (TNF) has shown inconsistent associations with susceptibility to both conditions. Additionally, the coding non-synonymous variant T60N in the neighbouring LTA gene has been reported to be associated with type 2 diabetes. The present study aimed to obtain a robust assessment of the role of variation in the tightly linked TNF/LTA region in diabetes susceptibility by genotyping TNF and LTA variants in large case-control resources. MATERIALS AND METHODS: The G-308A and G-238A TNF promoter variants and the LTA T60N polymorphism were genotyped in two UK case samples that were ascertained for positive family history and/or early onset of type 2 diabetes (combined n=858) and in 1,257 ethnically matched controls. RESULTS: There were no significant associations between the T60N, G-308A or G-238A genotype and type 2 diabetes in the combined analysis (exact Cochran-Mantel-Haenszel statistic for ordered genotypes for T60N, p=0.69; for G-308A, p=0.51; for G-238A, p=0.16). CONCLUSIONS/INTERPRETATION: The present study, one of the largest association analyses yet reported at this locus, provides no evidence that the specific TNF or LTA variants examined influence susceptibility to type 2 diabetes. More comprehensive studies of the TNF/LTA locus in substantially larger sample sets are required to establish whether genome sequence variation at this locus truly influences susceptibility to type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/epidemiologia , Diabetes Mellitus Tipo 2/genética , Linfotoxina-alfa/genética , Fator de Necrose Tumoral alfa/genética , Adulto , Idade de Início , Idoso , Feminino , Frequência do Gene , Variação Genética , Genótipo , Humanos , Desequilíbrio de Ligação , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único/genética , Reino Unido/epidemiologia
15.
Hum Genet ; 118(2): 214-24, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16133182

RESUMO

The nonsense-mediated decay (NMD) pathway is an mRNA surveillance mechanism that detects and degrades transcripts containing premature termination codons. The position of a truncating mutation can govern the resulting phenotype as mutations in the last exon evade NMD. In this study we investigated the susceptibility to NMD of six truncating HNF-1beta mutations by allele-specific quantitative real-time PCR using transformed lymphoblastoid cell lines. Four of six mutations (R181X, Q243fsdelC, P328L329fsdelCCTCT and A373fsdel29) showed evidence of NMD with levels of mutant transcript at 71% (p=0.009), 24% (p=0.008), 22% (p=0.008) and 3% (p=0.016) of the wild-type allele respectively. Comparable results were derived from lymphoblastoid cells and renal tubule cells isolated from a patient's overnight urine confirming that cell lines provide a good model for mRNA analysis. Two mutations (H69fsdelAC and P159fsdelT) produced transcripts unexpectedly immune to NMD. We conclude that truncating mutant transcripts of the HNF-1beta gene do not conform to the known rules governing NMD susceptibility, but instead demonstrate a previously unreported 5' to 3' polarity. We hypothesise that this may be due to reinitiation of translation downstream of the premature termination codon. Our study suggests that reinitiation of translation may be an important mechanism in the evasion of NMD, but that other factors such as the distance from the native initiation codon may also play a part.


Assuntos
Sequência de Bases , Códon sem Sentido , Fator 1-beta Nuclear de Hepatócito/genética , Biossíntese de Proteínas/genética , Estabilidade de RNA/genética , Deleção de Sequência , Adolescente , Linhagem Celular Tumoral , Códon/genética , Feminino , Doenças Genéticas Inatas/genética , Predisposição Genética para Doença , Humanos , Túbulos Renais/patologia , Masculino
16.
Clin Endocrinol (Oxf) ; 62(2): 169-75, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15670192

RESUMO

OBJECTIVE: Diagnostic molecular genetic testing for multiple endocrine neoplasia type 1 (MEN1) has been available since the identification of the MEN1 gene in 1997. Mutation screening of the MEN1 gene has been recommended for patients who meet clinical criteria for MEN1 (at least two of the following: parathyroid hyperplasia, pancreatic endocrine tumour or pituitary adenoma) and those in whom a diagnosis of MEN1 is suspected. We examined the appropriateness of these clinical criteria. PATIENTS AND METHODS: A total of 292 patients were referred for diagnostic testing. The coding region of the MEN1 gene was sequenced in 186 index cases and mutation testing was requested for 106 subjects, including 83 asymptomatic relatives. RESULTS: MEN1 gene mutations were identified in 68/186 index cases (37%). Twenty-nine of the 60 MEN1 mutations reported are novel. The likelihood of finding a mutation was correlated with the number of MEN1-related tumours (mutation detection rate of 79%, 37% and 15% in patients with three, two and one main MEN1-related tumours; P < or = 0.00001) and increased in the presence of a family history (mutation detection rate of 91%, 69% and 29%vs. 69%, 23% and 0% in sporadic cases with three, two or one main MEN1-related tumours, respectively; P < or = 0.00001). The pick-up rate in the 83% of subjects who met proposed criteria for diagnostic testing was 42%, but in those who did not meet these criteria this fell to 0%. CONCLUSIONS: The likelihood of finding an MEN1 mutation depends on the clinical features of the patient and their family. This large series supports present referral criteria for diagnostic mutation screening, but suggests that patients with sporadic isolated tumours rarely have MEN1 mutations.


Assuntos
Testes Genéticos , Neoplasia Endócrina Múltipla Tipo 1/genética , Seleção de Pacientes , Proteínas Proto-Oncogênicas/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Análise Mutacional de DNA , Éxons , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , Valor Preditivo dos Testes , Encaminhamento e Consulta , Estatísticas não Paramétricas
17.
Pediatr Pulmonol ; 39(2): 103-7, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15573388

RESUMO

The improving longevity of cystic fibrosis (CF) subjects has resulted in an increased prevalence and duration of cystic fibrosis-related diabetes (CFRD). Microvascular complications were reported in CFRD. Microalbuminuria is well-established as a sensitive indicator of progression to diabetic nephropathy in non-CF diabetes, but confounding factors may make it less sensitive for CF subjects. We performed a cross-sectional study to look for the presence of microalbuminuria in samples from 40 CF subjects (34 without diabetes; CFND) attending the Exeter CF Clinic, compared with 43 nondiabetic, non-CF controls. The albumin-creatinine ratio (ACR) was raised in CF subjects both with (P < 0.001) and without (P < 0.0001) diabetes compared to controls. This reflected an increase in urinary albumin and a reduction in urinary creatinine in CF subjects. In single samples, microalbuminuria was present in 66.7%, 32.4%, and 15.4% of subjects in CFRD, CFND, and control groups. Repeat samples showed that 12% of CFND subjects and 17% of CFRD subjects met the criteria for a diagnosis of persistent microalbuminuria. In conclusion, CF subjects, even when not diabetic, have increased urinary albumin excretion due to chronic infection, and reduced urinary creatinine excretion due to low muscle mass. This results in subjects, who are not developing diabetic nephropathy, meeting the conventional criteria for microalbuminuria. We feel that further studies are required to clarify whether this measure is a useful tool to predict progression to diabetic nephropathy in subjects with CFRD.


Assuntos
Albuminúria/urina , Fibrose Cística/complicações , Diabetes Mellitus/diagnóstico , Programas de Rastreamento , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Albuminúria/complicações , Albuminúria/diagnóstico , Biomarcadores/urina , Criança , Pré-Escolar , Creatinina/urina , Estudos Transversais , Fibrose Cística/urina , Diabetes Mellitus/etiologia , Diabetes Mellitus/urina , Feminino , Seguimentos , Humanos , Masculino , Programas de Rastreamento/métodos , Pessoa de Meia-Idade , Nefelometria e Turbidimetria
18.
Diabet Med ; 21(7): 691-6, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15209760

RESUMO

AIMS: Diabetes mellitus is an increasingly important complication of cystic fibrosis (CF). The association with increased morbidity of cystic fibrosis-related diabetes (CFRD) has emphasized the need for accurate monitoring of glycaemia in all CF patients. The diagnosis has relied on conventional thresholds in an oral glucose tolerance test (OGTT) derived from epidemiological studies in non-CF subjects. However, it has not been established if these values are equivalent in CF and non-CF populations. METHODS: We compared glycaemia in 21 non-diabetic CF subjects with 21-age and BMI-matched non-CF controls using HbA(1c), OGTT and a subcutaneous continuous glucose monitoring system (CGMS) that measures interstitial glucose levels. RESULTS: All conventional measures of glycaemia were similar in the two groups: HbA(1c) mean CF vs. controls (5.5 vs. 5.3%P = 0.4), fasting glucose (4.8 vs. 4.7 mmol/l P = 0.7) and 2-h glucose (5.8 vs. 5.7 mmol/l P = 0.8). However, these conventional measures did not accurately reflect glycaemia 30-, 60- and 90-min. Glucose values and area under the curve in OGTT were all higher in CF subjects than controls (P = 0.01-0.0001). Mean CGMS value [5.9 (0.8) vs. 5.1 (0.5) mmol/l, P = 0.004], and the proportion of subjects with peak CGMS values > 11.1 mmol/l (33 vs. 5%P = 0.00001) were also higher in CF subjects than controls. These results remained significantly different when only subjects with normal glucose tolerance in the two groups were studied. CONCLUSIONS: We have shown that overall glycaemia, as shown by both the response during an OGTT and CGMS, is higher in CF subjects who have similar HbA(1c), fasting and 2-h glucose values. These results question whether it is appropriate to use the diagnostic thresholds and OGTT time points derived from the non-CF population for a diagnosis of diabetes in patients with cystic fibrosis.


Assuntos
Fibrose Cística/complicações , Hiperglicemia/diagnóstico , Hiperglicemia/etiologia , Adulto , Fibrose Cística/sangue , Feminino , Teste de Tolerância a Glucose , Hemoglobinas Glicadas/análise , Humanos , Masculino , Monitorização Fisiológica/métodos , Monitorização Fisiológica/normas , Reprodutibilidade dos Testes
19.
Diabetologia ; 47(5): 937-42, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15085338

RESUMO

AIMS/HYPOTHESIS: Mutations in the hepatocyte nuclear factor-1 beta ( HNF-1 beta) gene result in disorders of renal development, typically involving renal cysts and early-onset diabetes (the RCAD syndrome/ MODY5). Sixteen mutations have been reported, including three splicing mutations of the intron 2 splice donor site. Because tissues showing abundant expression (kidney, liver, pancreas, gut, lung and gonads) are not easily accessible for analysis in living subjects, it has previously proven difficult to determine the effect of HNF-1 beta mutations at the mRNA level. This is the aim of the present study. METHODS: We have developed a nested RT-PCR assay that exploits the presence of ectopic HNF-1 beta transcripts in Epstein-Barr virus (EBV)-transformed lymphoblastoid cell lines derived from subjects carrying HNF-1 beta splice site mutations. RESULTS: We report a fourth mutation of the intron 2 splice donor site, IVS2nt+2insT. Sequence analysis of ectopic HNF-1 beta transcripts showed that both IVS2nt+2insT and IVS2nt+1G>T result in the deletion of exon 2 and are predicted to result in premature termination of the HNF-1 beta protein. Mutant transcripts were less abundant than the normal transcripts but there was no evidence of nonsense-mediated decay. CONCLUSIONS/INTERPRETATION: This is the first study to define the pathogenic consequences of mutations within the HNF-1 beta gene by mRNA analysis. This type of approach is a useful and important tool to define mutational mechanisms and determine pathogenicity.


Assuntos
Processamento Alternativo/genética , Proteínas de Ligação a DNA/genética , Diabetes Mellitus Tipo 2/genética , Doenças Renais Císticas/genética , Fatores de Transcrição/genética , Sequência de Bases , Primers do DNA , Feminino , Fator 1-beta Nuclear de Hepatócito , Humanos , Masculino , Linhagem , RNA Mensageiro/genética , Transcrição Gênica/genética
20.
Fam Pract ; 21(1): 57-62, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14760046

RESUMO

BACKGROUND: Although whole population screening for type 2 diabetes is not currently considered to be justified, targeted screening within higher risk groups may be more cost-effective, and more pragmatic. OBJECTIVES: Our aim was to investigate the feasibility and performance of a pragmatic system for identifying patients with type 2 diabetes and impaired fasting glycaemia (IFG). METHODS: A clustered observational survey of the prevalence of diabetes and IFG was carried out in randomly selected patients from four at-risk groups. Patients were identified by computerized searching of practice databases for age and body mass index (BMI) risk criteria. Sixteen practices in South West England screened 1287 Caucasian patients from four groups with progressive levels of theoretical risk (age >70 and BMI >or=33, age >65 and BMI >or=31, age >60 and BMI >or=29, and age >50 and BMI >or=27). Fasting plasma glucose was measured and repeated if abnormal to determine the prevalence of new cases in each group. BMI and age data were validated against measures taken at the clinic. RESULTS: The response rate was 60.6% and the prevalence of new cases of type 2 diabetes in each group was 4.7% [95% confidence interval (CI) 2.8-7.7], 5.7% (95% CI 4.0-8.2), 3.8% (95% CI 2.4-6.0) and 2.6% (95%CI 1.4-4.7), respectively. An additional 5.2-8.4% had IFG. CONCLUSIONS: Targeted screening by searching existing GP records for age and BMI criteria is feasible for use in general practice in the UK. Screening of patients with a BMI of >or=27 and aged >50 by fasting glucose identified a substantial prevalence of undetected type 2 diabetes and IFG. The relative costs and benefits as well as the pragmatic advantages of different systems need further evaluation.


Assuntos
Diabetes Mellitus Tipo 2/diagnóstico , Jejum/fisiologia , Hiperglicemia/diagnóstico , Programas de Rastreamento , Adulto , Fatores Etários , Idoso , Índice de Massa Corporal , Análise por Conglomerados , Intervalos de Confiança , Estudos Transversais , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/epidemiologia , Inglaterra , Medicina de Família e Comunidade , Estudos de Viabilidade , Feminino , Humanos , Hiperglicemia/sangue , Hiperglicemia/epidemiologia , Masculino , Programas de Rastreamento/estatística & dados numéricos , Sistemas Computadorizados de Registros Médicos/estatística & dados numéricos , Pessoa de Meia-Idade , Seleção de Pacientes , Medição de Risco/estatística & dados numéricos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA