1,347 results on '"Ellard, S."'
Search Results
402. Glucokinase mutations in a phenotypically selected multiethnic group of women with a history of gestational diabetes
- Author
-
Kousta, E., primary, Ellard, S., additional, Allen, L. I. S., additional, Saker, P. J., additional, Huxtable, S. J., additional, Hattersley, A. T., additional, and McCarthy, M. I., additional
- Published
- 2001
- Full Text
- View/download PDF
403. Predictive genetic testing in maturity‐onset diabetes of the young (MODY)
- Author
-
Shepherd, M., primary, Ellis, I., additional, Ahmad, A. M., additional, Todd, P. J., additional, Bowen‐Jones, D., additional, Mannion, G., additional, Ellard, S., additional, Sparkes, A. C., additional, and Hattersley, A. T., additional
- Published
- 2001
- Full Text
- View/download PDF
404. beta-cell genes and diabetes: molecular and clinical characterization of mutations in transcription factors.
- Author
-
Frayling, T M, primary, Evans, J C, additional, Bulman, M P, additional, Pearson, E, additional, Allen, L, additional, Owen, K, additional, Bingham, C, additional, Hannemann, M, additional, Shepherd, M, additional, Ellard, S, additional, and Hattersley, A T, additional
- Published
- 2001
- Full Text
- View/download PDF
405. beta-cell genes and diabetes: quantitative and qualitative differences in the pathophysiology of hepatic nuclear factor-1alpha and glucokinase mutations.
- Author
-
Pearson, E R, primary, Velho, G, additional, Clark, P, additional, Stride, A, additional, Shepherd, M, additional, Frayling, T M, additional, Bulman, M P, additional, Ellard, S, additional, Froguel, P, additional, and Hattersley, A T, additional
- Published
- 2001
- Full Text
- View/download PDF
406. Growth-hormone treatment and risk of diabetes
- Author
-
Spyer, G, primary, Ellard, S, additional, and Hattersley, A, additional
- Published
- 2000
- Full Text
- View/download PDF
407. A severe clinical phenotype results from the co-inheritance of type 2 susceptibility genes and a hepatocyte nuclear factor-1alpha mutation
- Author
-
Tack, C. J., primary, Ellard, S., additional, and Hattersley, A. T., additional
- Published
- 2000
- Full Text
- View/download PDF
408. A high prevalence of glucokinase mutations in gestational diabetic subjects selected by clinical criteria
- Author
-
Ellard, S., primary, Beards, F., additional, Allen, L. I. S., additional, Shepherd, M., additional, Ballantyne, E., additional, Harvey, R., additional, and Hattersley, A. T., additional
- Published
- 2000
- Full Text
- View/download PDF
409. Fetal genotype is the primary determinant of fetal growth in gestational diabetes due to a mutation in the maternal glucokinase gene
- Author
-
Hannemann, M.M., primary, Spyer, G., additional, Sturley, R., additional, MacLeod, K.M., additional, Ellard, S., additional, and Hattersley, A.T., additional
- Published
- 2000
- Full Text
- View/download PDF
410. HNF4A mutation: switch from hyperinsulinaemic hypoglycaemia to maturity-onset diabetes of the young, and incretin response.
- Author
-
Arya, V. B., Rahman, S., Senniappan, S., Flanagan, S. E., Ellard, S., and Hussain, K.
- Subjects
BLOOD sugar analysis ,TYPE 2 diabetes diagnosis ,HYPERINSULINISM ,BIRTH weight ,CARRIER state (Communicable diseases) ,DIABETES ,PEOPLE with diabetes ,GENES ,GLUCOSE tolerance tests ,HYPOGLYCEMIA ,INTRAVENOUS therapy ,INCRETINS ,GENETIC mutation ,WHITE people ,DISEASE complications ,DIAGNOSIS - Abstract
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. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
411. The evolving course of HNF4A hyperinsulinaemic hypoglycaemia-a case series.
- Author
-
McGlacken‐Byrne, S. M., Hawkes, C. P., Flanagan, S. E., Ellard, S., McDonnell, C. M., and Murphy, N. P.
- Subjects
GENETIC disorder diagnosis ,HYPOGLYCEMIA ,TYPE 2 diabetes diagnosis ,LIVER ,HYPERINSULINISM ,GENES ,GENETIC mutation ,PEDIATRICS ,PHENOTYPES ,DIAGNOSIS ,ANATOMY - Abstract
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. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
412. Parent-offspring trios: a resource to facilitate the identification of type 2 diabetes genes.
- Author
-
Frayling, T M, primary, Walker, M, additional, McCarthy, M I, additional, Evans, J C, additional, Allen, L I, additional, Lynn, S, additional, Ayres, S, additional, Millauer, B, additional, Turner, C, additional, Turner, R C, additional, Sampson, M J, additional, Hitman, G A, additional, Ellard, S, additional, and Hattersley, A T, additional
- Published
- 1999
- Full Text
- View/download PDF
413. Non-penetrance in a MODY 3 family with a mutation in the hepatic nuclear factor 1α gene: implications for predictive testing
- Author
-
Miedzybrodzka, Z, primary, Hattersley, AT, additional, Ellard, S, additional, Pearson, D, additional, de Silva, D, additional, Harvey, R, additional, and Haites, N, additional
- Published
- 1999
- Full Text
- View/download PDF
414. Allelic drop-out in exon 2 of the hepatocyte nuclear factor-1alpha gene hinders the identification of mutations in three families with maturity-onset diabetes of the young.
- Author
-
Ellard, S, primary, Bulman, M P, additional, Frayling, T M, additional, Allen, L I, additional, Dronsfield, M J, additional, Tack, C J, additional, and Hattersley, A T, additional
- Published
- 1999
- Full Text
- View/download PDF
415. Mutations in hepatocyte nuclear factor 1beta are not a common cause of maturity-onset diabetes of the young in the U.K.
- Author
-
Beards, F, primary, Frayling, T, additional, Bulman, M, additional, Horikawa, Y, additional, Allen, L, additional, Appleton, M, additional, Bell, G I, additional, Ellard, S, additional, and Hattersley, A T, additional
- Published
- 1998
- Full Text
- View/download PDF
416. A missense mutation in the hepatocyte nuclear factor 4 alpha gene in a UK pedigree with maturity-onset diabetes of the young
- Author
-
Bulman, M. P., primary, Dronsfield, M. J., additional, Frayling, T., additional, Appleton, M., additional, Bain, S. C., additional, Ellard, S., additional, and Hattersley, A. T., additional
- Published
- 1997
- Full Text
- View/download PDF
417. Mutations in the hepatocyte nuclear factor-1alpha gene are a common cause of maturity-onset diabetes of the young in the U.K
- Author
-
Frayling, T. M., primary, Bulamn, M. P., additional, Ellard, S., additional, Appleton, M., additional, Dronsfield, M. J., additional, Mackie, A. D., additional, Baird, J. D., additional, Kaisaki, P. J., additional, Yamagata, K., additional, Bell, G. I., additional, Bain, S. C., additional, and Hattersley, A. T., additional
- Published
- 1997
- Full Text
- View/download PDF
418. A study of the aneugenic activity of trichlorfon detected by centromere-specific probes in human lymphoblastoid cell lines
- Author
-
DOHERTY, A, primary, ELLARD, S, additional, PARRY, E, additional, and PARRY, J, additional
- Published
- 1996
- Full Text
- View/download PDF
419. The detection and evaluation of aneugenic chemicals
- Author
-
Parry, James M, primary, Parry, E.M, additional, Boumer, R, additional, Doherty, A, additional, Ellard, S, additional, O'Donovan, J, additional, Hoebee, B, additional, de Stoppelaar, J.M, additional, Mohn, G.R, additional, Önfelt, A, additional, Renglin, A, additional, Schultz, N, additional, Soderpalm-Bemdes, C, additional, Jensen, K.G, additional, Kirsch-Volders, M, additional, Elhajouji, A, additional, Van Hummelen, P, additional, Degrassi, F, additional, Antoccia, A, additional, Cimini, D, additional, Izzo, M, additional, Tanzarella, C, additional, Adler, I.-D, additional, Kliesch, U, additional, Schriever-Schwemmer, G, additional, Gasser, P, additional, Crebelli, R, additional, Carere, A, additional, Andreoli, C, additional, Benigni, R, additional, Leopardi, P, additional, Marcon, F, additional, Zinjo, Z, additional, Natarajan, A.T, additional, Boei, J.J.W.A, additional, Kappas, A, additional, Voutsinas, G, additional, Zarani, F.E, additional, Patrinelli, A, additional, Pachierotti, F, additional, Tiveron, C, additional, and Hess, P, additional
- Published
- 1996
- Full Text
- View/download PDF
420. Expanding the Clinical Spectrum Associated With GLIS3Mutations
- Author
-
Dimitri, P., Habeb, A. M., Garbuz, F., Millward, A., Wallis, S., Moussa, K., Akcay, T., Taha, D., Hogue, J., Slavotinek, A., Wales, J. K. H., Shetty, A., Hawkes, D., Hattersley, A. T., Ellard, S., and De Franco, E.
- Abstract
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 GLIS3mutations.Methods:GLIS3gene 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 GLIS3who 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 GLIS3phenotype, further extending the spectrum of abnormalities associated with GLIS3mutations and providing novel insights into the role of GLIS3in 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 GLIS3mutation, suggesting that even patients with a severe GLIS3phenotype may have a longer life expectancy than originally described.
- Published
- 2015
- Full Text
- View/download PDF
421. Improved genetic testing for monogenic diabetes using targeted next-generation sequencing.
- Author
-
Ellard, S., Lango Allen, H., Franco, E., Flanagan, S., Hysenaj, G., Colclough, K., Houghton, J., Shepherd, M., Hattersley, A., Weedon, M., and Caswell, R.
- Abstract
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. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
422. Phase II study of temsirolimus (CCI-779) in women with recurrent, unresectable, locally advanced or metastatic carcinoma of the cervix. A trial of the NCIC Clinical Trials Group (NCIC CTG IND 199).
- Author
-
Tinker, A.V., Ellard, S., Welch, S., Moens, F., Allo, G., Tsao, M.S., Squire, J., Tu, D., Eisenhauer, E.A., and MacKay, H.
- Subjects
- *
CERVICAL cancer , *METASTASIS , *CLINICAL trials , *METHYLATION , *PARTIAL response continuous phase modulation , *BIOMARKERS - Abstract
Abstract: Objective: HPV infection has been associated with deregulation of the PI3K-Akt-mTOR pathway in invasive cervical carcinomas. This 2-stage phase II study assessed the activity of the mTOR inhibitor, temsirolimus, in patients with measurable metastatic and/or locally advanced, recurrent carcinoma of the cervix. Methods: Temsirolimus 25mg i.v. was administered weekly in 4week cycles. One response among the first 18 patients was required to proceed to the second stage of accrual. Correlative molecular studies were performed on archival tumor tissue. Results: Thirty-eight patients were enrolled. Thirty-seven patients were evaluable for toxicity and 33 for response. One patient experienced a partial response (3.0%). Nineteen patients had stable disease (57.6%) [median duration 6.5months (range 2.4–12.0mo)]. The 6-month progression free survival rate was 28% (95% CI: 14–43%). The median progression free survival was 3.52months [95% CI (1.81–4.70)]. Adverse effects were mild–moderate in most cases and similar to other temsirolimus studies. No toxicity>grade 3 was observed. Assessment of PTEN and PIK3CA by IHC, copy number analyses and PTEN promoter methylation status did not reveal subsets associated with disease stability. Conclusion: Single agent temsirolimus has modest activity in cervical carcinoma with about two-thirds of patients exhibiting stable disease. Molecular markers for treatment benefit remain to be identified. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
423. Atypical phenotype associated with reported GCK exon 10 deletions: clinical judgement is needed alongside appropriate genetic investigations.
- Author
-
Thanabalasingham, G., Kaur, K., Talbot, F., Colclough, K., Mathews, A., Taylor, J., Ellard, S., and Owen, K. R.
- Subjects
TYPE 2 diabetes diagnosis ,HYPERGLYCEMIA ,GENETIC testing ,BIOCHEMISTRY ,BLOOD sugar ,CELL physiology ,DIABETES ,GENES ,GENETICS ,GLYCOSYLATED hemoglobin ,TYPE 1 diabetes ,TYPE 2 diabetes ,WEIGHT loss ,WHITE people ,PHENOTYPES ,DECISION making in clinical medicine ,BODY mass index ,SYMPTOMS ,DIAGNOSIS - Abstract
Background Maturity-onset diabetes of the young ( MODY) caused by heterozygous mutations in the glucokinase ( GCK) gene typically presents with lifelong, stable, mild fasting hyperglycaemia. With the exception of pregnancy, patients with GCK- MODY usually do not require pharmacological therapy. We report two unrelated patients whose initial genetic test results indicated a deletion of GCK exon 10, but whose clinical phenotypes were not typical of GCK- MODY. Case reports In case 1, the patient was hyperglycaemic at diagnosis (glucose > 30 mmol/l) and elevated glucose levels > 10 mmol/l persisted after withdrawal of insulin therapy. The patient in case 2 was also hyperglycaemic at diagnosis [HbA
1c > 86 mmol/mol (10%)], which improved with the introduction of oral hypoglycaemic agents. These clinical features were not consistent with GCK- MODY. Both patients had a single nucleotide variant that prevented multiplex ligation-dependent probe analysis, which generated a false positive result of a GCK exon 10 deletion. Conclusion False positive genetic results in these two unrelated cases were attributable to the presence of a rare single nucleotide variant that prevented ligation of the probe in the multiplex ligation-dependent probe analysis kit used and falsely indicated deletion of exon 10 within GCK. Both cases had clinical features that did not tally with the typical GCK- MODY phenotype. These cases emphasize the need to interpret the results of definitive genetic tests within the specific clinical context. Increased medical sequencing is likely to lead to more reports of novel mutations of uncertain significance. If genetic investigations do not agree with the clinical picture, clinicians should exercise caution when making therapeutic changes based on these results. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
424. Biallelic PDX1 (insulin promoter factor 1) mutations causing neonatal diabetes without exocrine pancreatic insufficiency.
- Author
-
Franco, E., Shaw‐Smith, C., Flanagan, S. E., Edghill, E. L., Wolf, J., Otte, V., Ebinger, F., Varthakavi, P., Vasanthi, T., Edvardsson, S., Hattersley, A. T., and Ellard, S.
- Subjects
GENETICS of diabetes ,PANCREATIC physiology ,GENETIC testing ,PANCREAS ,EXOCRINE pancreatic insufficiency ,DIABETES ,PEOPLE with diabetes ,GENETICS ,INSULIN ,MOLECULAR biology ,GENETIC mutation ,PEDIATRICS ,DIAGNOSIS ,ANATOMY - Abstract
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. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
425. Clinical presentation of 6q24 transient neonatal diabetes mellitus (6q24 TNDM) and genotype-phenotype correlation in an international cohort of patients.
- Author
-
Docherty, L., Kabwama, S., Lehmann, A., Hawke, E., Harrison, L., Flanagan, S., Ellard, S., Hattersley, A., Shield, J., Ennis, S., Mackay, D., and Temple, I.
- Abstract
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. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
426. HNF1B deletions in patients with young-onset diabetes but no known renal disease.
- Author
-
Edghill, E. L., Stals, K., Oram, R. A., Shepherd, M. H., Hattersley, A. T., and Ellard, S.
- Subjects
DIAGNOSIS of diabetes ,KIDNEY disease diagnosis ,LIVER ,GENETIC polymorphisms ,GENETICS ,GENETIC mutation ,ANATOMY - Abstract
Diabet. Med. 30, 114-117 (2013) Abstract Aims Hepatocyte nuclear factor 1β ( 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. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
427. Hypoglycaemia following diabetes remission in patients with 6q24 methylation defects: expanding the clinical phenotype.
- Author
-
Flanagan, S., Mackay, D., Greeley, S., McDonald, T., Mericq, V., Hassing, J., Richmond, E., Martin, W., Acerini, C., Kaulfers, A., Flynn, D., Popovic, J., Sperling, M., Hussain, K., Ellard, S., and Hattersley, A.
- Published
- 2013
- Full Text
- View/download PDF
428. Outcomes of women with early-stage breast cancer receiving adjuvant trastuzumab.
- Author
-
Seal, M. D., Speers, C. H., O'Reilly, S., Gelmon, K. A., Ellard, S. L., and Chia, S. K.
- Subjects
TRASTUZUMAB ,TREATMENT effectiveness ,HER2 protein ,BREAST cancer ,IMMUNOHISTOCHEMISTRY ,DISEASES in women - Abstract
Introduction Large randomized trials assessing the benefit of adjuvant trastuzumab in early-stage breast cancer positive for the human epidermal growth factor receptor 2 (HER2) have demonstrated a significant improvement in survival. The objective of the present study was to describe the outcomes of women who received adjuvant trastuzumab for HER2-positive breast cancer in British Columbia since publicly funded population-based use was initiated in July 2005. Methods Women from British Columbia, newly diagnosed with stage I-III breast cancer between July 2004 and December 2006, who were positive for HER2 overexpression by immunohistochemistry (3+) or amplification by fluorescence in situ hybridization (ratio ⩾ 2.0) were included in the study. Data were collected from the prospectively assembled BC Cancer Agency Outcomes Unit, with cases linked to the provincial pharmacy data repository to determine the proportion of women who received adjuvant trastuzumab. Results Our retrospective study identified 703 HER2-positive patients, of whom 480 (68%) received trastuzumab. In patients receiving trastuzumab, the 2-year relapse-free survival was 96.1% [95% confidence interval (CI): 93.6% to 97.7%] and the overall survival was 99.3% (95% CI: 97.9% to 99.8%). Among node-negative and -positive patients, the 2-year relapse-free survival was 97.8% and 94.8% respectively (p = 0.09) for the trastuzumab-treated group and 90.9% and 77.3% (p = 0.01) for the group not receiving trastuzumab (n = 223). Site of first distant metastasis was the central nervous system in 19.5% of the entire cohort and in 37.5% of patients treated with trastuzumab. Discussion This population-based analysis of adjuvant trastuzumab use among Canadian women demonstrates highly favorable outcomes at the 2-year follow-up. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
429. Pseudohypoaldosteronism type 2 presenting with hypertension and hyperkalaemia due to a novel mutation in the WNK4 gene.
- Author
-
Brooks, A.M., Owens, M., Sayer, J.A., Salzmann, M., Ellard, S., and Vaidya, B.
- Subjects
HYPERALDOSTERONISM ,HYPERTENSION ,HYPERKALEMIA ,HUMAN genetic variation ,BLOOD pressure ,GENETIC mutation ,GENETIC testing - Published
- 2012
- Full Text
- View/download PDF
430. JAG1 mutations are found in approximately one third of patients presenting with only one or two clinical features of Alagille syndrome.
- Author
-
Guegan, K, Stals, K, Day, M, Turnpenny, P, and Ellard, S
- Subjects
GENETIC mutation ,CHROMOSOME abnormalities ,LIVER biopsy ,BILE ducts ,MOLECULAR genetics - Abstract
Guegan K, Stals K, Day M, Turnpenny P, Ellard S. JAG1 mutations are found in approximately one third of patients presenting with only one or two clinical features of Alagille syndrome. Alagille syndrome is a multisystem disorder characterized by highly variable expressivity, most frequently caused by heterozygous JAG1 gene mutations. Classic diagnostic criteria combine the presence of bile duct paucity on liver biopsy with three of five systems affected; liver, heart, skeleton, eye and dysmorphic facies. The aim of this study was to determine the prevalence and distribution of JAG1 mutations in patients referred for routine clinical diagnostic testing. Clinical data were available for 241 patients from 135 families. The index cases were grouped according to the number of systems affected (heart, liver, skeletal, eye and facies) and the mutation frequency calculated for each group. JAG1 mutations were identified in 59/135 (44%) probands. The highest mutation detection rates were observed in patients with the most frequent presenting features of Alagille syndrome; ranging from 20% (one system) to 86% (five systems). The overall mutation pick-up rate in a clinical diagnostic setting was lower than in previous research studies. Identification of a JAG1 gene mutation is particularly useful for those patients with atypical or mild Alagille syndrome who do not meet classic diagnostic criteria as it provides a definite molecular diagnosis and allows accurate genetic counselling for the family. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
431. Systematic assessment of etiology in adults with a clinical diagnosis of young-onset type 2 diabetes is a successful strategy for identifying maturity-onset diabetes of the young.
- Author
-
Thanabalasingham G, Pal A, Selwood MP, Dudley C, Fisher K, Bingley PJ, Ellard S, Farmer AJ, McCarthy MI, Owen KR, Thanabalasingham, Gaya, Pal, Aparna, Selwood, Mary P, Dudley, Christina, Fisher, Karen, Bingley, Polly J, Ellard, Sian, Farmer, Andrew J, McCarthy, Mark I, and Owen, Katharine R
- Abstract
Objective: Misdiagnosis of maturity-onset diabetes of the young (MODY) remains widespread, despite the benefits of optimized management. This cross-sectional study examined diagnostic misclassification of MODY in subjects with clinically labeled young adult-onset type 1 and type 2 diabetes by extending genetic testing beyond current guidelines.Research Design and Methods: Individuals were selected for diagnostic sequencing if they displayed features atypical for their diagnostic label. From 247 case subjects with clinically labeled type 1 diabetes, we sequenced hepatocyte nuclear factor 1 α (HNF1A) and hepatocyte nuclear factor 4 α (HNF4A) in 20 with residual β-cell function ≥ 3 years from diagnosis (random or glucagon-stimulated C-peptide ≥ 0.2 nmol/L). From 322 with clinically labeled type 2 diabetes, we sequenced HNF1A and HNF4A in 80 with diabetes diagnosed ≤ 30 years and/or diabetes diagnosed ≤ 45 years without metabolic syndrome. We also sequenced the glucokinase (GCK) in 40 subjects with mild fasting hyperglycemia.Results: In the type 1 diabetic group, two HNF1A mutations were found (0.8% prevalence). In type 2 diabetic subjects, 10 HNF1A, two HNF4A, and one GCK mutation were identified (4.0%). Only 47% of MODY case subjects identified met current guidelines for diagnostic sequencing. Follow-up revealed a further 12 mutation carriers among relatives. Twenty-seven percent of newly identified MODY subjects changed treatment, all with improved glycemic control (HbA(1c) 8.8 vs. 7.3% at 3 months; P = 0.02).Conclusions: The systematic use of widened diagnostic testing criteria doubled the numbers of MODY case subjects identified compared with current clinical practice. The yield was greatest in young adult-onset type 2 diabetes. We recommend that all patients diagnosed before age 30 and with presence of C-peptide at 3 years' duration are considered for molecular diagnostic analysis. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
432. Partial ABCC8 gene deletion mutations causing diazoxide-unresponsive hyperinsulinaemic hypoglycaemia.
- Author
-
Flanagan, SE, Damhuis, A, Banerjee, I, Rokicki, D, Jefferies, C, Kapoor, RR, Hussain, K, and Ellard, S
- Subjects
HYPOGLYCEMIA ,ANTIHYPERTENSIVE agents ,INSULIN shock ,PANCREAS ,BIOCHEMISTRY ,CELL physiology ,GENEALOGY ,GENES ,GENETIC techniques ,MOLECULAR biology ,GENETIC mutation ,PEDIATRICS ,DIAGNOSIS ,ANATOMY - Abstract
Flanagan SE, Damhuis A, Banerjee I, Rokicki D, Jefferies C, Kapoor RR, Hussain K, Ellard S. Partial ABCC8 gene deletion mutations causing diazoxide-unresponsive hyperinsulinaemic hypoglycaemia. Inactivating mutations in the pancreatic beta cell ATP-sensitive potassium (K
ATP ) channel genes are identified by sequencing in approximately 80% of patients with diazoxide-unresponsive hyperinsulinaemic hypoglycaemia (HH). Genetic testing is clinically important as the mode of inheritance of a KATP channel mutation(s) provides information on the histological subtype. For example in patients with a single paternally inherited mutation a focal lesion is possible and once confirmed, the patient can undergo a curative lesionectomy. By contrast, recessive inheritance indicates diffuse disease, which requires near-total pancreatectomy, if medical management is unsuccessful. We investigated ABCC8 and KCNJ11 gene dosage in 29 probands from a cohort of 125 with diazoxide-unresponsive HH where sequencing did not provide a genetic diagnosis. We identified heterozygous partial ABCC8 deletions in four probands. In two cases with focal pancreatic disease, a paternally inherited deletion was found. Two other probands with diffuse pancreatic disease were compound heterozygotes for a deletion and a recessively acting mutation that had been identified by sequencing. Family member studies confirmed compound heterozygosity for the deletion and the missense mutation in two affected siblings of one proband. Heterozygous deletions of the ABCC8 gene are a rare, but important cause of diazoxide-unresponsive HH. Dosage analysis should be undertaken in all patients when sequencing analysis does not confirm the genetic diagnosis as confirmation of the mode of inheritance can guide clinical management and will provide important information regarding recurrence risk. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
433. Heterozygous ABCC8 mutations are a cause of MODY.
- Author
-
Bowman, P., Flanagan, S., Edghill, E., Damhuis, A., Shepherd, M., Paisey, R., Hattersley, A., and Ellard, S.
- Abstract
Aims/hypothesis: The ABCC8 gene encodes the sulfonylurea receptor 1 (SUR1) subunit of the pancreatic beta cell ATP-sensitive potassium (K) 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/m, 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. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
434. Urinary C-peptide creatinine ratio is a practical outpatient tool for identifying hepatocyte nuclear factor 1-{alpha}/hepatocyte nuclear factor 4-{alpha} maturity-onset diabetes of the young from long-duration type 1 diabetes.
- Author
-
Besser RE, Shepherd MH, McDonald TJ, Shields BM, Knight BA, Ellard S, Hattersley AT, Besser, Rachel E J, Shepherd, Maggie H, McDonald, Timothy J, Shields, Beverley M, Knight, Bridget A, Ellard, Sian, and Hattersley, Andrew T
- Abstract
Objective: Hepatocyte nuclear factor 1-α (HNF1A)/hepatocyte nuclear factor 4-α (HNF4A) maturity-onset diabetes of the young (MODY) is frequently misdiagnosed as type 1 diabetes, and patients are inappropriately treated with insulin. Blood C-peptide can aid in the diagnosis of MODY, but practical reasons limit its widespread use. Urinary C-peptide creatinine ratio (UCPCR), a stable measure of endogenous insulin secretion, is a noninvasive alternative. We aimed to compare stimulated UCPCR in adults with HNF1A/4A MODY, type 1 diabetes, and type 2 diabetes.Research Design and Methods: Adults with diabetes for ≥ 5 years, without renal impairment, were studied (HNF1A MODY [n = 54], HNF4A MODY [n = 23], glucokinase MODY [n = 20], type 1 diabetes [n = 69], and type 2 diabetes [n = 54]). The UCPCR was collected in boric acid 120 min after the largest meal of the day and mailed for analysis. Receiver operating characteristic (ROC) curves were used to identify optimal UCPCR cutoffs to differentiate HNF1A/4A MODY from type 1 and type 2 diabetes.Results: UCPCR was lower in type 1 diabetes than HNF1A/4A MODY (median [interquartile range]) (<0.02 nmol/mmol [<0.02 to <0.02] vs. 1.72 nmol/mmol [0.98-2.90]; P < 0.0001). ROC curves showed excellent discrimination (area under curve [AUC] 0.98) and identified a cutoff UCPCR of ≥ 0.2 nmol/mmol for differentiating HNF1A/4A MODY from type 1 diabetes (97% sensitivity, 96% specificity). UCPCR was lower in HNF1A/4A MODY than in type 2 diabetes (1.72 nmol/mmol [0.98-2.90] vs. 2.47 nmol/mmol [1.4-4.13]); P = 0.007). ROC curves showed a weak distinction between HNF1A/4A MODY and type 2 diabetes (AUC 0.64).Conclusions: UCPCR is a noninvasive outpatient tool that can be used to discriminate HNF1A and HNF4A MODY from long-duration type 1 diabetes. To differentiate MODY from type 1 diabetes of >5 years' duration, UCPCR could be used to determine whether genetic testing is indicated. [ABSTRACT FROM AUTHOR]- Published
- 2011
- Full Text
- View/download PDF
435. A phase IB study of ABT-751 in combination with docetaxel in patients with advanced castration-resistant prostate cancer.
- Author
-
Michels, J., Ellard, S. L., Le, L., Kollmannsberger, C., Murray, N., Tomlinson Guns, E. S., Carr, R., and Chi, K. N.
- Abstract
Background: This study investigated the safety, pharmacokinetics (PK) and clinical antitumor activity of ABT-751, a novel sulfonamide antimitotic and vascular disrupting agent, in combination with docetaxel (Taxotere) in patients with castration-resistant prostate cancer (CRPC). Patients and methods: Patients received docetaxel (60–75 mg/m2) i.v. on day 1 and ABT-751 (100–200 mg) orally daily for 14 days, repeated every 3 weeks for up to 10 times on four escalating dose levels (DLs). Results: Thirty-two patients received a median of 8.5 treatment cycles (range 1–10). One of six patients on DL 3 (D 60 mg/m2 + A 200 mg) and 4 (D 75 mg/m2 + A 200 mg) experienced dose-limiting toxicity, and both DLs were expanded. Overall, severe adverse events occurred more commonly on DL 4 than 3 (47% versus 18% of patients). PK data for docetaxel and ABT-751 were similar to reported literature. Best post-treatment prostate-specific antigen decline of ‡50% occurred in 60% and objective responses occurred in 45% of patients. Median overall survival was 24 months (95% confidence interval 8.3–37.7 months). Conclusions: The combination of ABT-751 and docetaxel is safe and active in CRPC. Based on the cumulative safety analysis, the recommended phase II dose of ABT-751 is 200 mg daily with docetaxel 60 mg/m2 for this patient population. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
436. Incidence of neonatal diabetes in Austria -- calculation based on the Austrian Diabetes Register.
- Author
-
Wiedemann B, Schober E, Waldhoer T, Koehle J, Flanagan SE, Mackay DJG, Steichen E, Meraner D, Zimmerhackl L, Hattersley AT, Ellard S, and Hofer S
- Abstract
Background: Neonatal diabetes mellitus (NDM) is a rare monogenic form of diabetes which is diagnosed in the first 6 months of life. Several studies in the last few years provide information on genetic causes for NDM. Objective: The aim of this study was to identify all patients with diabetes in the first 6 months of life through the Austrian Diabetes Register, which is available since 1989. A retrospective data analyses was performed to calculate the current incidence of NDM. Subjects and Methods: Ten patients were registered with diabetes onset within the first 6 months of life in the Austrian Diabetes Register. Evaluation of detailed clinical data was performed by sending a questionnaire to all diabetes centers. Results: Ten patients from nine different families with NDM were diagnosed in Austria from 1989 until September 2007. Seven patients (one male, six females) had transient NDM (TNDM), three (two males, one female) showed a permanent course [permanent neonatal diabetes mellitus (PNDM)]. One had immunodeficiency, polyendocrinopathy and enteropathy X-linked (IPEX) syndrome and another showed aplasia of the pancreas; no genetic etiology was found in the third case. In three out of seven patients with a transient course of NDM a genetic diagnosis was possible. Two female siblings had activating point mutations in the ABCC8 gene, although one patient had paternal uniparental isodisomy of chromosome 6q24. One patient's family did not consent to genetic testing. Conclusions: The incidence of NDM in Austria is 1/160 949, with an incidence of 1/ 536 499 for PNDM and 1/229 928 for TNDM. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
437. Identification of a novel beta-cell glucokinase (GCK) promoter mutation (-71G>C) that modulates GCK gene expression through loss of allele-specific Sp1 binding causing mild fasting hyperglycemia in humans.
- Author
-
Gasperíková D, Tribble ND, Staník J, Hucková M, Misovicová N, van de Bunt M, Valentínová L, Barrow BA, Barák L, Dobránsky R, Bereczková E, Michálek J, Wicks K, Colclough K, Knight JC, Ellard S, Klimes I, Gloyn AL, Gasperíková, Daniela, and Tribble, Nicolas D
- Abstract
Objective: Inactivating mutations in glucokinase (GCK) cause mild fasting hyperglycemia. Identification of a GCK mutation has implications for treatment and prognosis; therefore, it is important to identify these individuals. A significant number of patients have a phenotype suggesting a defect in glucokinase but no abnormality of GCK. We hypothesized that the GCK beta-cell promoter region, which currently is not routinely screened, could contain pathogenic mutations; therefore, we sequenced this region in 60 such probands.Research Design and Methods: The beta-cell GCK promoter was sequenced in patient DNA. The effect of the identified novel mutation on GCK promoter activity was assessed using a luciferase reporter gene expression system. Electrophoretic mobility shift assays (EMSAs) were used to determine the impact of the mutation on Sp1 binding.Results: A novel -71G>C mutation was identified in a nonconserved region of the human promoter sequence in six apparently unrelated probands. Family testing established cosegregation with fasting hyperglycemia (> or = 5.5 mmol/l) in 39 affected individuals. Haplotype analysis in the U.K. family and four of the Slovakian families demonstrated that the mutation had arisen independently. The mutation maps to a potential transcriptional activator binding site for Sp1. Reporter assays demonstrated that the mutation reduces promoter activity by up to fourfold. EMSAs demonstrated a dramatic reduction in Sp1 binding to the promoter sequence corresponding to the mutant allele.Conclusions: A novel beta-cell GCK promoter mutation was identified that significantly reduces gene expression in vitro through loss of regulation by Sp1. To ensure correct diagnosis of potential GCK-MODY (maturity-onset diabetes of the young) cases, analysis of the beta-cell GCK promoter should be included. [ABSTRACT FROM AUTHOR]- Published
- 2009
- Full Text
- View/download PDF
438. A genetic diagnosis of HNF1A diabetes alters treatment and improves glycaemic control in the majority of insulin-treated patients.
- Author
-
Shepherd, M., Shields, B., Ellard, S., Rubio-Cabezas, O., and Hattersley, A. T.
- Subjects
DIABETES risk factors ,GENETIC disorder diagnosis ,PEOPLE with diabetes ,INSULIN therapy ,MOLECULAR genetics - Abstract
Background and aims Hepatocyte nuclear factor-1 alpha ( HNF1A) gene mutations are the commonest cause of monogenic diabetes, but patients are often misdiagnosed as having Type 1 diabetes and started on insulin treatment. Patients with HNF1A diabetes are particularly sensitive to the glucose-lowering effect of sulphonylureas, which are the pharmacological treatment of choice. We aimed to assess if patients do change from insulin to sulphonylurea treatment when HNF1A diabetes is confirmed and the impact of this treatment change on long-term glycaemic control. Methods We investigated the clinical course of 43 patients who were insulin treated from diagnosis for a median 4 years (range 1–14) before an HNF1A gene mutation was identified. Results Thirty-four patients (79%) stopped insulin following genetic testing and transferred to sulphonylureas. Twenty-four of them (71%) remained off insulin at a median 39 months (range 17–90) post-transfer. The 10 patients who recommenced insulin had a trend towards a longer duration of diabetes (18 vs. 7 years, P = 0.066) compared with those remaining on tablets. The median glycated haemoglobin (HbA
1c ) was good (6.9%; interquartile range 6.3–8.0%) in the patients who remained off insulin and 19/24 patients (79%) achieved HbA1c < 7.5% or improved their pre-genetic diagnosis HbA1c by > 1.0%. Transfer off insulin was not attempted in eight patients: one of these was planning pregnancy and two chose to remain on insulin. Conclusion In this observational study we found that a molecular genetic diagnosis of HNF1A diabetes does alter treatment in clinical practice, with 79% attempting transfer to sulphonylureas. Transfer to sulphonylureas was successful in the majority of patients without deterioration in glycaemic control. [ABSTRACT FROM AUTHOR]- Published
- 2009
- Full Text
- View/download PDF
439. Diabetes susceptibility in the Canadian Oji-Cree population is moderated by abnormal mRNA processing of HNF1A G319S transcripts.
- Author
-
Harries LW, Sloman MJ, Sellers EA, Hattersley AT, Ellard S, Harries, Lorna W, Sloman, Melissa J, Sellers, Elizabeth A C, Hattersley, Andrew T, and Ellard, Sian
- Abstract
Objective: The G319S HNF1A variant is associated with an increased risk of type 2 diabetes in the Canadian Oji-Cree population. We hypothesized that the variant site at the 3' end of exon 4 might influence splicing and characterized mRNA transcripts to investigate the mutational mechanism underlying this susceptibility to diabetes.Research Design and Methods: We established lymphoblastoid cell lines from a G319S homozygote and controls. HNF1A transcripts were characterized in the cell lines and pancreatic tissue by sequence analysis of RT-PCR products and quantification using real-time PCR. Susceptibility to mRNA surveillance was investigated using cycloheximide.Results: Full-length G319S mRNA accounted for 24% of mRNA transcripts in the homozygous G319S cell line. A novel isoform lacking the terminal 12 bases of exon 4 was upregulated (55% of mRNA transcripts) compared with control cell lines (33%) and human pancreatic tissue (17%). Two abnormal transcripts present only in the G319S cell line included premature termination codons as a result of the inclusion of seven nucleotides from intron 4 or the deletion of exon 8. Cycloheximide treatment increased the levels of both transcripts.Conclusions: The G319S variant results in the production of two abnormal transcripts and an alteration in the relative balance of normal splicing products. This is predicted to lead to a reduction in total HNF1A transcript levels, but residual hepatocyte nuclear factor-1alpha protein activity in G319S homozygotes may still reach up to 66% of normal levels. A combination of abnormal splicing and reduced activity of the G319S protein may explain the diabetes susceptibility. [ABSTRACT FROM AUTHOR]- Published
- 2008
- Full Text
- View/download PDF
440. Best practice guidelines for the molecular genetic diagnosis of maturity-onset diabetes of the young.
- Author
-
Ellard, S., Bellanné-Chantelot, C., and Hattersley, A.
- Abstract
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. 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 (). 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. 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. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
441. Effective treatment with oral sulfonylureas in patients with diabetes due to sulfonylurea receptor 1 (SUR1) mutations.
- Author
-
Rafiq M, Flanagan SE, Patch AM, Shields BM, Ellard S, Hattersley AT, Neonatal Diabetes International Collaborative Group, Rafiq, Meena, Flanagan, Sarah E, Patch, Ann-Marie, Shields, Beverley M, Ellard, Sian, and Hattersley, Andrew T
- Abstract
Objective: Neonatal diabetes can result from mutations in the Kir6.2 or sulfonylurea receptor 1 (SUR1) subunits of the ATP-sensitive K(+) channel. Transfer from insulin to oral sulfonylureas in patients with neonatal diabetes due to Kir6.2 mutations is well described, but less is known about changing therapy in patients with SUR1 mutations. We aimed to describe the response to sulfonylurea therapy in patients with SUR1 mutations and to compare it with Kir6.2 mutations.Research Design and Methods: We followed 27 patients with SUR1 mutations for at least 2 months after attempted transfer to sulfonylureas. Information was collected on clinical features, treatment before and after transfer, and the transfer protocol used. We compared successful and unsuccessful transfer patients, glycemic control before and after transfer, and treatment requirements in patients with SUR1 and Kir6.2 mutations.Results: Twenty-three patients (85%) successfully transferred onto sulfonylureas without significant side effects or increased hypoglycemia and did not need insulin injections. In these patients, median A1C fell from 7.2% (interquartile range 6.6-8.2%) on insulin to 5.5% (5.3-6.2%) on sulfonylureas (P = 0.01). When compared with Kir6.2 patients, SUR1 patients needed lower doses of both insulin before transfer (0.4 vs. 0.7 units x kg(-1) x day(-1); P = 0.002) and sulfonylureas after transfer (0.26 vs. 0.45 mg x kg(-1) x day(-1); P = 0.005).Conclusions: Oral sulfonylurea therapy is safe and effective in the short term in most patients with diabetes due to SUR1 mutations and may successfully replace treatment with insulin injections. A different treatment protocol needs to be developed for this group because they require lower doses of sulfonylureas than required by Kir6.2 patients. [ABSTRACT FROM AUTHOR]- Published
- 2008
- Full Text
- View/download PDF
442. Mosaic paternal uniparental isodisomy and an ABCC8 gene mutation in a patient with permanent neonatal diabetes and hemihypertrophy.
- Author
-
Shield JP, Flanagan SE, Mackay DJ, Harries LW, Proks P, Girard C, Ashcroft FM, Temple IK, Ellard S, Shield, Julian P H, Flanagan, Sarah E, Mackay, Deborah J, Harries, Lorna W, Proks, Peter, Girard, Christophe, Ashcroft, Frances M, Temple, I Karen, and Ellard, Sian
- Abstract
Objective: Activating mutations in the KCNJ11 and ABCC8 genes encoding the Kir6.2 and SUR1 subunits of the pancreatic ATP-sensitive K(+) channel are the most common cause of permanent neonatal diabetes. In contrast to KCNJ11, where only dominant heterozygous mutations have been identified, recessively acting ABCC8 mutations have recently been found in some patients with neonatal diabetes. These genes are co-located on chromosome 11p15.1, centromeric to the imprinted Beckwith-Wiedemann syndrome (BWS) locus at 11p15.5. We investigated a male with hemihypertrophy, a condition classically associated with neonatal hyperinsulinemia and hypoglycemia, who developed neonatal diabetes at age 5 weeks.Research Design and Methods: The KCNJ11 and ABCC8 genes and microsatellite markers on chromosome 11 were analyzed in DNA samples from the patient and his parents.Results: A paternally inherited activating mutation (N72S) in the ABCC8 gene was identified in the proband. The mutation was present at 70% in the patient's leukocytes and 50% in buccal cells. Microsatellite analysis demonstrated mosaic segmental paternal uniparental isodisomy (UPD) of 11pter-11p14 in the proband that encompassed the ABCC8 gene and the BWS locus.Conclusions: We report a patient with neonatal diabetes, hemihypertrophy, and relatively high birth weight resulting from telomeric segmental paternal UPD of chromosome 11, which unmasks a recessively acting gain-of-function mutation in the ABCC8 gene and causes deregulation of imprinted genes at the BWS locus on 11p15.5. [ABSTRACT FROM AUTHOR]- Published
- 2008
- Full Text
- View/download PDF
443. Mutations in the ABCC8 gene encoding the SUR1 subunit of the KATP channel cause transient neonatal diabetes, permanent neonatal diabetes or permanent diabetes diagnosed outside the neonatal period.
- Author
-
Patch, A. M., Flanagan, S. E., Boustred, C., Hattersley, A. T., and Ellard, S.
- Subjects
DIABETES ,POTASSIUM channels ,PATIENTS ,EXONS (Genetics) ,PHENOTYPES ,ADENOSINE triphosphate - Abstract
Aim: Mutations in the ABCC8 gene encoding the SUR1 subunit of the pancreatic ATP-sensitive potassium channel cause permanent neonatal diabetes mellitus (PNDM) and transient neonatal diabetes mellitus (TNDM). We reviewed the existing literature, extended the number of cases and explored genotype–phenotype correlations. Methods: Mutations were identified by sequencing in patients diagnosed with diabetes before 6 months without a KCNJ11 mutation. Results: We identified ABCC8 mutations in an additional nine probands (including five novel mutations L135P, R306H, R1314H, L438F and M1290V), bringing the total of reported families to 48. Both dominant and recessive mutations were observed with recessive inheritance more common in PNDM than TNDM (9 vs. 1; p < 0.01). The remainder of the PNDM probands (n = 12) had de novo mutations. Seventeen of twenty-five children with TNDM inherited their heterozygous mutation from a parent. Nine of these parents had permanent diabetes (median age at diagnosis: 27.5 years, range: 13–35 years). Recurrent mutations of residues R1183 and R1380 were found only in TNDM probands and dominant mutations causing PNDM clustered within exons 2–5. Conclusions: ABCC8 mutations cause PNDM, TNDM or permanent diabetes diagnosed outside the neonatal period. There is some evidence that the location of the mutation is correlated with the clinical phenotype. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
444. Mutations in ATP-sensitive K+ channel genes cause transient neonatal diabetes and permanent diabetes in childhood or adulthood.
- Author
-
Flanagan SE, Patch AM, Mackay DJ, Edghill EL, Gloyn AL, Robinson D, Shield JP, Temple K, Ellard S, Hattersley AT, Flanagan, Sarah E, Patch, Ann-Marie, Mackay, Deborah J G, Edghill, Emma L, Gloyn, Anna L, Robinson, David, Shield, Julian P H, Temple, Karen, Ellard, Sian, and Hattersley, Andrew T
- Abstract
Transient neonatal diabetes mellitus (TNDM) is diagnosed in the first 6 months of life, with remission in infancy or early childhood. For approximately 50% of patients, their diabetes will relapse in later life. The majority of cases result from anomalies of the imprinted region on chromosome 6q24, and 14 patients with ATP-sensitive K+ channel (K(ATP) channel) gene mutations have been reported. We determined the 6q24 status in 97 patients with TNDM. In patients in whom no abnormality was identified, the KCNJ11 gene and/or ABCC8 gene, which encode the Kir6.2 and SUR1 subunits of the pancreatic beta-cell K(ATP) channel, were sequenced. K(ATP) channel mutations were found in 25 of 97 (26%) TNDM probands (12 KCNJ11 and 13 ABCC8), while 69 of 97 (71%) had chromosome 6q24 abnormalities. The phenotype associated with KCNJ11 and ABCC8 mutations was similar but markedly different from 6q24 patients who had a lower birth weight and who were diagnosed and remitted earlier (all P < 0.001). K(ATP) channel mutations were identified in 26 additional family members, 17 of whom had diabetes. Of 42 diabetic patients, 91% diagnosed before 6 months remitted, but those diagnosed after 6 months had permanent diabetes (P < 0.0001). K(ATP) channel mutations account for 89% of patients with non-6q24 TNDM and result in a discrete clinical subtype that includes biphasic diabetes that can be treated with sulfonylureas. Remitting neonatal diabetes was observed in two of three mutation carriers, and permanent diabetes occurred after 6 months of age in subjects without an initial diagnosis of neonatal diabetes. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
445. Mutations in hepatocyte nuclear factor-1 ß and their related phenotypes.
- Author
-
Edghill, E. L., Bingham, C., Ellard, S., and Hattersley, A. T.
- Subjects
GENETIC mutation ,TRANSCRIPTION factors ,DIAGNOSIS ,KIDNEY diseases ,DIABETES ,CYSTS (Pathology) ,GENES ,GENETIC research ,MOLECULAR genetics ,MEDICAL genetics - Abstract
Background: Hepatocyte nuclear factor-1 beta (HNF-1β) is a widely distributed transcription factor which plays a critical role in embryonic development of the kidney, pancreas, liver, and Mullerian duct. Thirty HNF-1β 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-1β gene in 160 unrelated subjects with renal disease, 40% of whom had a personal/family history of diabetes. Results: Twenty three different heterozygous HNF-1β 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 far 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-1β mutations and confirm HNF-1β mutations as an important cause of renal disease. Despite the original description of HNF-1β as o 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-1β mutations should be considered in patients with unexplained renal cysts (including GCKD), especially when associated with diabetes, early-onset gout, or uterine abnormalities. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
446. Detection of anMEN1gene mutation depends on clinical features and supports current referral criteria for diagnostic molecular genetic testing.
- Author
-
Ellard, S., Hattersley, A. T., Brewer, C. M., and Vaidya, B.
- Subjects
- *
GENETIC mutation , *TUMORS , *ONCOLOGY , *GENETICS , *BIOLOGICAL variation , *CLINICAL medicine - Abstract
Diagnostic molecular genetic testing for multiple endocrine neoplasia type 1 (MEN1) has been available since the identification of theMEN1gene in 1997. Mutation screening of theMEN1gene 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.A total of 292 patients were referred for diagnostic testing. The coding region of theMEN1gene was sequenced in 186 index cases and mutation testing was requested for 106 subjects, including 83 asymptomatic relatives.MEN1gene mutations were identified in 68/186 index cases (37%). Twenty-nine of the 60MEN1mutations 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 ≤ 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 ≤ 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%.The likelihood of finding anMEN1mutation 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 haveMEN1mutations. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
447. Integration of the MODY link nurse project: 20-month evaluation.
- Author
-
Shepherd M, Hattersley A, and Ellard S
- Published
- 2005
448. Analysis of haematopoietic chimaerism by quantitative real-time polymerase chain reaction.
- Author
-
Harries, L W, Wickham, C L, Evans, J C, Rule, S A, Joyner, M V, and Ellard, S
- Subjects
BONE marrow transplantation ,POLYMERASE chain reaction ,BLOOD ,IMMUNE system ,HUMAN biology ,PATIENTS - Abstract
Summary:Allogeneic bone marrow transplantation (BMT) with marrow ablative conditioning is the treatment of choice for haematopoietic malignancies. The use of nonmyeloablative stem cell transplants has allowed the treatment of patients previously ineligible for BMT because of age or other disease. These reduced conditioning regimes allow the persistence initially of some recipient cells in the blood and bone marrow (haematopoietic chimaerism). Monitoring of the relative proportion of donor and recipient cells is required to assess the success of the procedure, to predict subsequent rejection or impending relapse and to guide the use of donor lymphocyte infusions. We present a quantitative real-time PCR approach for the measurement of haematopoietic chimaerism using the TaqMan™. This approach exploits the presence of single-nucleotide polymorphisms (SNPs) to distinguish cells of patient or donor origin. We have designed and validated a panel of seven allele-specific probes to quantify the contribution of patient and donor cells in the haematopoietic population from 12 patient and donor pairs. We have compared the performance of this approach with an existing method and proved it to be superior in both accuracy and sensitivity. The use of more sensitive and accurate techniques permits earlier intervention for improved clinical outcome.Bone Marrow Transplantation (2005) 35, 283-290. doi:10.1038/sj.bmt.1704764 Published online 8 November 2004 [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
449. The absorption of vitamin E is influenced by the amount of fat in a meal and the food matrix.
- Author
-
Jeanes YM, Hall WL, Ellard S, Lee E, and Lodge JK
- Published
- 2004
- Full Text
- View/download PDF
450. Abnormal splicing of hepatocyte nuclear factor-1 beta in the renal cysts and diabetes syndrome Abnormal splicing of the HNF-1β gene.
- Author
-
Harries, L. W., Ellard, S., Jones, R. W. A., Hattersley, A. T., and Bingham, C.
- Subjects
GENETIC mutation ,CYSTIC kidney disease ,INTRONS ,EXONS (Genetics) ,CELL lines ,REVERSE transcriptase ,POLYMERASE chain reaction - Abstract
Aims/hypothesis. Mutations in the hepatocyte nuclear factor-1 beta (HNF-1β) 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-1β 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-1β transcripts in Epstein-Barr virus (EBV)-transformed lymphoblastoid cell lines derived from subjects carrying HNF-1β splice site mutations. Results. We report a fourth mutation of the intron 2 splice donor site, IVS2nt+2insT. Sequence analysis of ectopic HNF-1β 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-1β 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-1β gene by mRNA analysis. This type of approach is a useful and important tool to define mutational mechanisms and determine pathogenicity. [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.