Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Diabetes Res Clin Pract ; 188: 109908, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35533745

ABSTRACT

AIM: Identification of monogenic diabetes (MgD) conveys benefits for patients' care. Algorithms for selecting the patients to be genetically tested have been established in EuroCaucasians, but not in non-EuroCaucasian individuals. We assessed the diagnosis rate, the phenotype of MgD, and the relevance of selection criteria, according to ancestry in patients referred for a suspected MgD. METHODS: Seven genes (GCK, HNF1A, HNF4A, HNF1B, ABCC8, KCNJ11, INS) were analyzed in 1975 adult probands (42% non-EuroCaucasians), selected on the absence of diabetes autoantibodies and ≥2 of the following criteria: age ≤40 years and body mass index <30 kg/m2 at diagnosis, and a family history of diabetes in ≥2 generations. RESULTS: Pathogenic/likely pathogenic variants were identified in 6.2% of non-EuroCaucasian and 23.6% of EuroCaucasian patients (OR 0.21, [0.16-0.29]). Diagnosis rate was low in all non-EuroCaucasian subgroups (4.1-11.8%). Common causes of MgD (GCK, HNF1A, HNF4A), but not rare causes, were less frequent in non-EuroCaucasians than in EuroCaucasians (4.1%, vs. 21.1%, OR 0.16 [0.11-0.23]). Using ethnicity-specific body mass index cutoffs increased the diagnosis rate in several non-EuroCaucasian subgroups. CONCLUSION: The diagnosis rate of MgD is low in non-EuroCaucasian patients, but may be improved by tailoring selection criteria according to patients'ancestry.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/genetics , Genetic Testing , Humans , Mutation , Phenotype
4.
Diabetes Metab ; 42(1): 33-7, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25753245

ABSTRACT

AIM: Low plasma levels of high-sensitivity C-reactive protein (hs-CRP) have been suggested to differentiate hepatocyte nuclear factor 1 alpha-maturity-onset diabetes of the young (HNF1A-MODY) from type 2 diabetes (T2D). Yet, differential diagnosis of HNF1A-MODY and familial young-onset type 2 diabetes (F-YT2D) remains a difficult challenge. Thus, this study assessed the added value of hs-CRP to distinguish between the two conditions. METHODS: This prospective multicentre study included 143 HNF1A-MODY patients, 310 patients with a clinical history suggestive of HNF1A-MODY, but not confirmed genetically (F-YT2D), and 215 patients with T2D. The ability of models, including clinical characteristics and hs-CRP to predict HNF1A-MODY was analyzed, using the area of the receiver operating characteristic (AUROC) curve, and a grey zone approach was used to evaluate these models in clinical practice. RESULTS: Median hs-CRP values were lower in HNF1A-MODY (0.25mg/L) than in F-YT2D (1.14mg/L) and T2D (1.70mg/L) patients. Clinical parameters were sufficient to differentiate HNF1A-MODY from classical T2D (AUROC: 0.99). AUROC analyses to distinguish HNF1A-MODY from F-YT2D were 0.82 for clinical features and 0.87 after including hs-CRP. For the grey zone analysis, the lower boundary was set to miss<1.5% of true positives in non-tested subjects, while the upper boundary was set to perform 50% of genetic tests in individuals with no HNF1A mutation. On comparing HNF1A-MODY with F-YT2D, 65% of patients were classified in between these categories - in the zone of diagnostic uncertainty - even after adding hs-CRP to clinical parameters. CONCLUSION: hs-CRP does not improve the differential diagnosis of HNF1A-MODY and F-YT2D.


Subject(s)
C-Reactive Protein/analysis , Diabetes Mellitus, Type 2/classification , Diabetes Mellitus, Type 2/diagnosis , Adolescent , Adult , Aged , Aged, 80 and over , Child , Diabetes Mellitus, Type 2/epidemiology , Diagnosis, Differential , Female , Humans , Male , Middle Aged , Prospective Studies , ROC Curve , Young Adult
5.
Clin Genet ; 87(5): 448-54, 2015 May.
Article in English | MEDLINE | ID: mdl-24814349

ABSTRACT

ABCC8 encodes a subunit of the ß-cell potassium channel (KATP ) whose loss of function is responsible for congenital hyperinsulinism (CHI). Patients with two recessive mutations of ABCC8 typically have severe diffuse forms of CHI unresponsive to diazoxide. Some dominant ABCC8 mutations are responsible for a subset of diffuse diazoxide-unresponsive forms of CHI. We report the analysis of 21 different ABCC8 mutations identified in 25 probands with diazoxide-unresponsive diffuse CHI and carrying a single mutation in ABCC8. Nine missense ABCC8 mutations were subjected to in vitro expression studies testing traffic efficiency and responses of mutant channels to activation by MgADP and diazoxide. Eight of the 9 missense mutations exhibited normal trafficking. Seven of the 8 mutants reaching the plasma membrane had dramatically reduced response to MgADP or to diazoxide (<10% of wild-type response). In our cohort, dominant KATP mutations account for 22% of the children with diffuse unresponsive-diazoxide CHI. Their clinical phenotype being indistinguishable from that of children with focal CHI and diffuse CHI forms due to two recessive KATP mutations, we show that functional testing is essential to make the most reliable diagnosis and offer appropriate genetic counseling.


Subject(s)
Alleles , Congenital Hyperinsulinism/drug therapy , Congenital Hyperinsulinism/genetics , Diazoxide/therapeutic use , Drug Resistance/genetics , Mutation , Sulfonylurea Receptors/genetics , Amino Acid Substitution , Congenital Hyperinsulinism/diagnosis , DNA Mutational Analysis , Female , Genes, Recessive , Humans , Infant , Infant, Newborn , Male , Phenotype , Sulfonylurea Receptors/metabolism , Treatment Outcome
7.
Diabetes Metab ; 38(2): 179-82, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22326206

ABSTRACT

AIM: ATP-sensitive potassium channels are important regulators of insulin secretion. They consist of four sulphonylurea receptor (encoded by ABCC8) and four inwardly rectifying protein (encoded by KCNJ11) subunits. Activating ABCC8 mutations lead to decreased insulin secretion and to diabetes. Wide phenotype variability is associated with single ABCC8 mutations, ranging from transient or permanent neonatal diabetes (ND) with or without developmental delay (DEND syndrome) to very mild phenotypes. This report describes the case of a Caucasian infant diagnosed with ND at the age of 2 months due to a novel ABCC8 missense mutation. METHODS: ABCC8 was analyzed by sequence analysis. The mutation was present in the patient and her family and was found to be associated with phenotypes ranging from ND to asymptomatic impaired fasting glucose (IFG). RESULTS: A novel His863Tyr ABCC8 mutation was identified in a 2-month-old girl diagnosed with ND. After an initial insulin treatment, treatment with glibenclamide was initiated and the treatment with insulin discontinued. The same mutation was found in her father, who had been fortuitously diagnosed with diabetes and had an HbA(1c) level of 9% (74.8 mmol/mol). The patient's brother and mother both had normal fasting glucose, and were not found to be carriers of the mutation. However, the same mutation was found in her grandmother, who had been asymptomatic and discovered IFG (6.9 mmol/L) with an HbA(1c) of 6.8% (50.8 mmol/mol). CONCLUSION: This case describes a novel ABCC8 mutation and offers a further illustration of the highly variable phenotypes associated with an identical mutation present across three generations.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Diabetes Mellitus/genetics , Mutation, Missense , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels/genetics , Receptors, Drug/genetics , Diabetes Mellitus/drug therapy , Female , Genetic Diseases, Inborn , Humans , Infant , Pedigree , Phenotype , Sulfonylurea Receptors
8.
J Clin Endocrinol Metab ; 96(12): 3785-93, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21956412

ABSTRACT

BACKGROUND: Morphological studies of the pancreas in persistent hyperinsulinemic hypoglycemia of infancy (PHHI) have focused on the diagnosis of focal vs. diffuse forms, a distinction that determines the optimal surgical management. ABCC8 or KCNJ11 genomic mutations are present in most of them. AIM: Our aim was to report a new form of PHHI with peculiar morphological and clinical characteristics. RESEARCH DESIGN AND METHODS: Histopathological review of 217 pancreatic PHHI specimens revealed 16 cases morphologically different from diffuse and focal forms. They were analyzed by conventional microscopy, quantitative morphometry, immunohistochemistry, and in situ hybridization. RESULTS: Their morphological peculiarity was the coexistence of two types of islet: large islets with cytoplasm-rich ß-cells and occasional enlarged nuclei and shrunken islets with ß-cells exhibiting little cytoplasm and small nuclei. In small islets, ß-cells had abundant insulin content but limited amount of Golgi proinsulin. Large islets had low insulin storage and high proinsulin production and were mostly confined to a few lobules. No evidence for K(ATP) channels involvement or 11p15 deletion was found. Genomic mutations for ABCC8, KCNJ11, and GCK were absent. Patients had normal birth weight and late hypoglycemia onset and improved with diazoxide. Ten were cured by limited pancreatectomy. Six recurred after surgery and were medically controlled. CONCLUSION: This new form of PHHI is characterized by a morphological mosaicism. Pathologists should recognize this mosaicism on intraoperative frozen sections because it is often curable by partial pancreatectomy. The currently unknown genetic background does not involve the classical genomic mutations responsible for diffuse and focal PHHI.


Subject(s)
Congenital Hyperinsulinism/pathology , Islets of Langerhans/pathology , Congenital Hyperinsulinism/genetics , Congenital Hyperinsulinism/surgery , Female , Humans , Infant , Infant, Newborn , Islets of Langerhans/surgery , Male , Microsatellite Repeats , Mosaicism , Mutation , Pancreatectomy , Treatment Outcome
9.
Clin Microbiol Infect ; 17(2): 135-9, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20148920

ABSTRACT

Disseminated Mycobacterium avium complex (MAC) infection is a rare but severe disease mostly seen in patients with AIDS. It has been previously described in patients suffering from other kinds of immunodeficiency (e.g. primary immunodeficiency diseases in children or hairy cell leukaemia). We report two cases of disseminated MAC disease in young women with extended granulomatosis that revealed a new form of severe immunodeficiency syndrome. Both clinical observations initially appeared to be very similar to WHIM syndrome (Warts, Hypogammaglobulinemia, Infection, Myelokathexis), a rare immunodeficiency disease correlated with CXC chemokine receptor 4 (CXCR4) mutation leading to an impaired internalization of the receptor upon its ligand CXCL12. We investigated the CXCR4 status of the lymphocytes in both patients and found a severe defect in CXCL12-promoted internalization but no mutation of its gene. Moreover, myelokathexis was not noted in bone marrow biopsies and therefore a diagnosis of WHIM syndrome could not be assessed. This immunodeficiency syndrome associated with CXCR4 dysfunction was responsible for severe MAC infection in our patients, with a fatal outcome in one case. It may be possible that these patients would have benefited from early antimycobacterial infection or azythromycin prophylaxis.


Subject(s)
Immunologic Deficiency Syndromes/complications , Immunologic Deficiency Syndromes/diagnosis , Mycobacterium avium Complex/isolation & purification , Mycobacterium avium-intracellulare Infection/diagnosis , Mycobacterium avium-intracellulare Infection/immunology , Receptors, CXCR4/immunology , Fatal Outcome , Female , Histocytochemistry , Humans , Mesenteric Lymphadenitis/diagnostic imaging , Mesenteric Lymphadenitis/pathology , Microscopy , Mycobacterium avium-intracellulare Infection/pathology , Positron-Emission Tomography , Radiography , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Skin/pathology , Young Adult
10.
Diabet Med ; 27(12): 1454-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21105491

ABSTRACT

OBJECTIVE: We have dissected the rare molecular anomalies that may affect hepatocyte nuclear factor-1a (HNF1A) and hepatocyte nuclear factor-4a (HNF4A) in patients with familial young-onset diabetes for whom HNF1A mutations have been excluded by sequence analysis. METHODS: Eighty-four unrelatedHNF1A-negative patients with diabetes diagnosed before the age of 40 years, a family history of diabetes and the absence of features suggestive of Type 2 diabetes were included. We analysed by sequencing the HNF4A promoter and coding regions, the HNF1A promoter region and specific regions of HNF1A(B) and HNF1A(C) isoforms and searched for large deletions of HNF1A and HNF4A by multiplex ligation-dependent probe amplification (MLPA). RESULTS: We identified five novel HNF4A mutations (5 / 84, 6%), including four missense and one in-frame deletion, and one mutation of the HNF1A promoter (1 / 84). Sequence analysis of isoform-specific coding regions of HNF1A did not reveal any mutation. We next identified two whole gene deletions of HNF1A and HNF4A, respectively (2 / 84, 2.4%). CONCLUSIONS: Altogether, the search for rare molecular events in HNF1A and HNF4A led us to elucidate 8 / 84 (9.5%) of our HNF1A-negative cases.This study shows that genetic aetiologies remain to be elucidated in familial young-onset diabetes. It also highlights the difficulty of the differential diagnosis with Type 2 diabetes because of the wide clinical expression of monogenic young-onset diabetes and the absence of specific biomarkers.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 4/genetics , Mutation/genetics , Adult , Age of Onset , Diabetes Mellitus, Type 2/diagnosis , Family , Female , Genotype , Hepatocyte Nuclear Factor 1-alpha/physiology , Hepatocyte Nuclear Factor 4/physiology , Humans , Male , Molecular Sequence Data , Polymorphism, Genetic , Retrospective Studies
11.
J Med Genet ; 47(11): 752-9, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20685672

ABSTRACT

BACKGROUND: Congenital hyperinsulinism (CHI) is characterised by an over secretion of insulin by the pancreatic ß-cells. This condition is mostly caused by mutations in ABCC8 or KCNJ11 genes encoding the SUR1 and KIR6.2 subunits of the ATP-sensitive potassium (K(ATP)) channel. CHI patients are classified according to their responsiveness to diazoxide and to their histopathological diagnosis (either focal, diffuse or atypical forms). Here, we raise the benefits/limits of the genetic diagnosis in the clinical management of CHI patients. METHODS: ABCC8/KCNJ11 mutational spectrum was established in 109 diazoxide-unresponsive CHI patients for whom an appropriate clinical management is essential to prevent brain damage. Relationships between genotype and radiopathological diagnosis were analysed. RESULTS: ABCC8 or KCNJ11 defects were found in 82% of the CHI cases. All patients with a focal form were associated with a single K(ATP) channel molecular event. In contrast, patients with diffuse forms were genetically more heterogeneous: 47% were associated with recessively inherited mutations, 34% carried a single heterozygous mutation and 19% had no mutation. There appeared to be a predominance of paternally inherited mutations in patients diagnosed with a diffuse form and carrying a sole K(ATP) channel mutation. CONCLUSIONS: The identification of recessively inherited mutations related to severe and diffuse forms of CHI provides an informative genetic diagnosis and allows prenatal diagnosis. In contrast, in patients carrying a single K(ATP) channel mutation, genetic analysis should be confronted with the PET imaging to categorise patients as focal or diffuse forms in order to get the appropriate therapeutic management.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Congenital Hyperinsulinism/genetics , Mutation , Potassium Channels, Inwardly Rectifying/genetics , Receptors, Drug/genetics , Congenital Hyperinsulinism/diagnosis , Congenital Hyperinsulinism/drug therapy , DNA Mutational Analysis , Diazoxide/therapeutic use , Drug Resistance , Female , Genotype , Humans , Infant , Infant, Newborn , Male , Sulfonylurea Receptors , Vasodilator Agents/therapeutic use
12.
Rev Med Interne ; 31(6): e5-6, 2010 Jun.
Article in French | MEDLINE | ID: mdl-20206420

ABSTRACT

A 55-year-old woman presented with a recent diabetes mellitus associated with pancreatic and renal malformations. This atypical diabetes raised the possibility of maturity onset diabetes of the young (MODY) and a genetic research was performed. These malformations led to MODY5 diagnosis that was confirmed by the presence of HNF1beta gene mutation.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Hepatocyte Nuclear Factor 1-beta/genetics , Kidney/abnormalities , Pancreas/abnormalities , Biomarkers/metabolism , Female , Genetic Counseling , Humans , Middle Aged , Mutation , Phenotype
13.
J Clin Endocrinol Metab ; 94(8): 3025-30, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19470619

ABSTRACT

CONTEXT: Maternally inherited diabetes and deafness (MIDD) is a rare form of diabetes with a matrilineal transmission, sensorineural hearing loss, and macular pattern dystrophy due to an A to G transition at position 3243 of mitochondrial DNA (mtDNA) (m.3243A>G). The phenotypic heterogeneity of MIDD may be the consequence of different levels of mutated mtDNA among mitochondria in a given tissue. OBJECTIVE: The aim of the present study was thus to ascertain the correlation between the severity of the phenotype in patients with MIDD and the level of heteroplasmy in the blood leukocytes. PARTICIPANTS: The GEDIAM prospective multicenter register was initiated in 1995. Eighty-nine Europid patients from this register, with MIDD and the mtDNA 3243A>G mutation, were included. Patients with MELAS (mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes) or with mitochondrial diabetes related to other mutations or to deletions of mtDNA were excluded. RESULTS: A significant negative correlation was found between levels of heteroplasmy and age of the patients at the time of sampling for molecular analysis, age at the diagnosis of diabetes, and body mass index. After adjustment for age at sampling for molecular study and gender, the correlation between heteroplasmy levels and age at the diagnosis of diabetes was no more significant. The two other correlations remained significant. A significant positive correlation between levels of heteroplasmy and HbA(1c) was also found and remained significant after adjustment for age at molecular sampling and gender. CONCLUSIONS: These results support the hypothesis that heteroplasmy levels are at least one of the determinants of the severity of the phenotype in MIDD.


Subject(s)
DNA, Mitochondrial/genetics , Deafness/genetics , Diabetes Mellitus/genetics , Leukocytes/metabolism , Mitochondrial Diseases/genetics , Point Mutation , Adult , Age Factors , Body Mass Index , Female , Glycated Hemoglobin/analysis , Humans , Male , Middle Aged , Prospective Studies , Sex Characteristics
14.
Diabetes Metab ; 35(3): 233-5, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19342262

ABSTRACT

Activating mutations in genes KCNJ11 and ABCC8, which form the ATP-sensitive K+channel (K(ATP) channel), have been shown to cause transient or permanent neonatal diabetes. We describe here a rather different phenotype: two cases of adult diabetic patients-considered and treated as insulin-dependent diabetic patients since adolescence-who, in fact, turned out to be heterozygous for an ABCC8 mutation and able to successfully discontinue insulin while taking sulphonylurea treatment.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Autoantibodies/blood , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Mutation , Potassium Channels, Inwardly Rectifying/genetics , Receptors, Drug/genetics , Adolescent , Adult , Female , Hepatocyte Nuclear Factor 1-alpha/genetics , Humans , Infant, Newborn , Infant, Newborn, Diseases/genetics , Male , Middle Aged , Sulfonylurea Receptors
16.
J Clin Endocrinol Metab ; 93(12): 4941-7, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18796520

ABSTRACT

CONTEXT: Focal forms of congenital hyperinsulinism are due to a constitutional heterozygous mutation of paternal origin in the ABCC8 gene, more often than the KCNJ11 gene, located in the 11p15.1 region. This mutation is associated with the loss of the maternally inherited 11p15.1 to 11p15.5 region in the lesion. We investigated the possible occurrence of a compensatory duplication of the paternal 11p15.1-11p15.5 region. MATERIALS AND METHODS: A combined immunohistochemistry and fluorescent in situ hybridization study on beta-cell interphase nuclei with probes covering two genes located in this region (ABCC8 and CDKN1C genes) was performed in four cases of focal forms of hyperinsulinism. RESULTS: beta-Cells in the lesions of four cases of focal congenital hyperinsulinism were diploid for chromosomes 11 and 13. The 11p15.1 to 11p15.2 and 11p15.4 to 11p15.5 regions containing ABCC8 and CDKN1C genes, respectively, were present with two copies. Loss of the maternal allele was confirmed in these focal lesions with microsatellite markers flanking the ABCC8 and CDKN1C genes, whereas a heterozygous mutation in the ABCC8 gene was inherited from the father. CONCLUSIONS: There is a duplication of the paternal allele on chromosome 11 in the focal forms of hyperinsulinism lesion. The paternal isodisomy observed rendered the beta-cells homozygous for ABCC8 mutation and harbored a K-channel defect in the lesion similar to that observed in diffuse forms of congenital hyperinsulinism.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Hyperinsulinism/congenital , Hyperinsulinism/genetics , Uniparental Disomy/genetics , ATP-Binding Cassette Transporters/genetics , Alleles , Chromosomes, Human, Pair 13/genetics , DNA/biosynthesis , DNA/genetics , Fathers , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Infant, Newborn , Insulin-Secreting Cells/metabolism , Male , Microsatellite Repeats , Ploidies , Potassium Channels, Inwardly Rectifying/genetics , Receptors, Drug/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sulfonylurea Receptors
17.
Pediatr Blood Cancer ; 51(5): 701-3, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18661496

ABSTRACT

A 12-year-old daughter of consanguineous Moroccan parents was diagnosed with cyclic neutropenia, based on a combination of recurrent gingivostomatitis, a fluctuating neutrophil count, and several episodes of severe neutropenia. No ELA2 gene mutations were found. At age 19 years she presented with edema of the limbs, proteinuria and renal failure. Renal amyloidosis AA was diagnosed by biopsy. Gene mutations associated with family Mediterranean fever (FMF) were sought, and a homozygous mutation (M694V) was found in the MFEV gene. This is the novel finding of FMF that masqueraded as cyclic neutropenia.


Subject(s)
Familial Mediterranean Fever/diagnosis , Familial Mediterranean Fever/genetics , Familial Mediterranean Fever/physiopathology , Neutropenia/pathology , Adult , Amyloidosis/genetics , Child , Chronic Disease , Cytoskeletal Proteins/genetics , DNA Mutational Analysis , Diagnosis, Differential , Female , Fever/genetics , Humans , Kidney Diseases/genetics , Mutation , Pyrin , Stomatitis, Herpetic/genetics
18.
Diabetologia ; 51(9): 1664-70, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18581092

ABSTRACT

AIMS/HYPOTHESIS: We assessed the prevalence and determinants of retinal and renal complications in patients with maternally inherited diabetes and deafness (MIDD). METHODS: This was a multicentre prospective study comparing the prevalence of retinopathy and renal disease in 74 patients with MIDD and 134 control patients matched for sex, age and clinical presentation at onset of diabetes, duration of diabetes and current treatment. Comparisons were adjusted for HbA(1c) and hypertension. RESULTS: In MIDD patients, HbA(1c) (7.6 +/- 1.6 vs 8.5 +/- 2.0%, p < 0.002), systolic blood pressure (126.6 +/- 16.2 vs 133.1 +/- 17.3 mmHg, p < 0.007) and prevalence of hypertension (33.8 vs 64.2%, p < 0.0001) were lower than in control patients. Prevalence of diabetic retinopathy was 3.7-fold lower in MIDD patients (6/74, 8 vs 40/134, 29.6%, p < 0.0001). Differences between groups remained significant after adjustment for hypertension, systolic blood pressure and HbA(1c). In MIDD, urinary albumin excretion (314.8 vs 80.1 mg/24 h, p = 0.035) and creatinine plasma levels (103.5 vs 82.2 micromol/l, p = 0.0178) were higher and GFR was lower. Impaired renal function (GFR <60 ml/min) was four- to sixfold more frequent in MIDD. Differences between MIDD and control diabetic patients further increased when adjusted for HbA(1c) and systolic blood pressure (p < 0.0001). Adjustment for treatment with an ACE inhibitor or angiotensin II receptor antagonist did not modify the results. CONCLUSIONS/INTERPRETATION: This study indicates that diabetic retinopathy is less prevalent in MIDD than in control diabetes. This suggests that retinal alterations due to mitochondrial disease may have a protective role. By contrast, nephropathy is far more frequent in MIDD, suggesting the presence of a specific renal disease independent of diabetic nephropathy.


Subject(s)
DNA, Mitochondrial/genetics , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 2/genetics , Diabetic Nephropathies/genetics , Diabetic Retinopathy/genetics , Kidney Diseases/genetics , Mitochondrial Diseases/genetics , Mutation , Retinal Diseases/genetics , Blood Pressure , DNA, Mitochondrial/chemistry , Diabetic Angiopathies/genetics , Female , Glomerular Filtration Rate , Glycated Hemoglobin/analysis , Humans , Kidney Diseases/epidemiology , Phenotype , Retinal Diseases/epidemiology
19.
Diabetologia ; 51(4): 546-53, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18297260

ABSTRACT

AIMS/HYPOTHESIS: Mutations in the GCK and HNF1A genes are the most common cause of the monogenic forms of diabetes known as 'maturity-onset diabetes of the young'. GCK encodes the glucokinase enzyme, which acts as the pancreatic glucose sensor, and mutations result in stable, mild fasting hyperglycaemia. A progressive insulin secretory defect is seen in patients with mutations in the HNF1A and HNF4A genes encoding the transcription factors hepatocyte nuclear factor-1 alpha and -4 alpha. A molecular genetic diagnosis often changes management, since patients with GCK mutations rarely require pharmacological treatment and HNF1A/4A mutation carriers are sensitive to sulfonylureas. These monogenic forms of diabetes are often misdiagnosed as type 1 or 2 diabetes. Best practice guidelines for genetic testing were developed to guide testing and reporting of results. METHODS: A workshop was held to discuss clinical criteria for testing and the interpretation of molecular genetic test results. The participants included 22 clinicians and scientists from 13 countries. Draft best practice guidelines were formulated and edited using an online tool (http://www.coventi.com). RESULTS: An agreed set of clinical criteria were defined for the testing of babies, children and adults for GCK, HNF1A and HNF4A mutations. Reporting scenarios were discussed and consensus statements produced. CONCLUSIONS/INTERPRETATION: Best practice guidelines have been established for monogenic forms of diabetes caused by mutations in the GCK, HNF1A and HNF4A genes. The guidelines include both diagnostic and predictive genetic tests and interpretation of the results.


Subject(s)
Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/genetics , Mutation , Practice Guidelines as Topic/standards , Adolescent , Child , Chromosome Mapping , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 4/genetics , Humans , Polymorphism, Single Nucleotide , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...