Skip to main content
Top

06-22-2018 | Monogenic diabetes | Review | Article

Monogenic Diabetes in Children and Adolescents: Recognition and Treatment Options

Journal: Current Diabetes Reports

Authors: May Sanyoura, Louis H. Philipson, Rochelle Naylor

Publisher: Springer US

Abstract

Purpose of Review

We provide a review of monogenic diabetes in young children and adolescents with a focus on recognition, management, and pharmacological treatment.

Recent Findings

Monogenic forms of diabetes account for approximately 1–2% of diabetes in children and adolescents, and its incidence has increased in recent years due to greater awareness and wider availability of genetic testing. Monogenic diabetes is due to single gene defects that primarily affect beta cell function with more than 30 different genes reported. Children with antibody-negative, C-peptide-positive diabetes should be evaluated and genetically tested for monogenic diabetes. Accurate genetic diagnosis impacts treatment in the most common types of monogenic diabetes, including the use of sulfonylureas in place of insulin or other glucose-lowering agents or discontinuing pharmacologic treatment altogether.

Summary

Diagnosis of monogenic diabetes can significantly improve patient care by enabling prediction of the disease course and guiding appropriate management and treatment.
Literature
1.
Flannick J, Johansson S, Njølstad PR. Common and rare forms of diabetes mellitus: towards a continuum of diabetes subtypes. Nat Publ Group. 2016;12(7):1–13. https://​doi.​org/​10.​1038/​nrendo.​2016.​50.​ CrossRef
2.
Vaxillaire M, Froguel P. Monogenic forms of diabetes mellitus: an update. Endocrinol Nutr. 2009;56S4:26–9. https://​doi.​org/​10.​1016/​S1575-0922(09)73513-2.PubMedCrossRef
3.
Alkorta-Aranburu G, Carmody D, Cheng YW, Nelakuditi V, Ma L, Dickens JT, et al. Phenotypic heterogeneity in monogenic diabetes: the clinical and diagnostic utility of a gene panel-based next-generation sequencing approach. Mol Genet Metab. 2014;113(4):315–20. https://​doi.​org/​10.​1016/​j.​ymgme.​2014.​09.​007.PubMedPubMedCentralCrossRef
4.
• Hattersley AT, Patel KA. Precision diabetes: learning from monogenic diabetes. Diabetologia. 2017;60(5):1–9. https://​doi.​org/​10.​1007/​s00125-017-4226-2. Provides a review of the success of a precision medicine approach using monogenic forms of diabetes as an example. CrossRef
5.
Kleinberger JW, Pollin TI. Personalized medicine in diabetes mellitus: current opportunities and future prospects. Ann N Y Acad Sci. 2015;1346(1):45–56. https://​doi.​org/​10.​1111/​nyas.​12757.PubMedPubMedCentralCrossRef
6.
Ehtisham S, Hattersley AT, Dunger DB, Barrett TG, Group BSfPEaDCT. First UK survey of paediatric type 2 diabetes and MODY. Arch Dis Child. 2004;89(6):526–9. https://​doi.​org/​10.​1136/​adc.​2003.​027821.PubMedPubMedCentralCrossRef
7.
Fendler W, Borowiec M, Baranowska-Jazwiecka A, Szadkowska A, Skala-Zamorowska E, Deja G, et al. Prevalence of monogenic diabetes amongst Polish children after a nationwide genetic screening campaign. Diabetologia. 2012;55(10):2631–5. https://​doi.​org/​10.​1007/​s00125-012-2621-2.PubMedPubMedCentralCrossRef
8.
Irgens HU, Molnes J, Johansson BB, Ringdal M, Skrivarhaug T, Undlien DE, et al. Prevalence of monogenic diabetes in the population-based Norwegian Childhood Diabetes Registry. Diabetologia. 2013;56(7):1512–9. https://​doi.​org/​10.​1007/​s00125-013-2916-y.
9.
Pihoker C, Gilliam LK, Ellard S, Dabelea D, Davis C, Dolan LM, et al. Prevalence, characteristics and clinical diagnosis of maturity onset diabetes of the young due to mutations in HNF1A, HNF4A, and glucokinase: results from the SEARCH for Diabetes in Youth. J Clin Endocrinol Metab. 2013;98(10):4055–62. https://​doi.​org/​10.​1210/​jc.​2013-1279.
10.
•• Shepherd M, Shields B, Hammersley S, Hudson M, McDonald TJ, Colclough K, et al. Systematic population screening, using biomarkers and genetic testing, identifies 2.5% of the U.K. pediatric diabetes population with monogenic diabetes. Diabetes Care. 2016;39(11):1879–88. https://​doi.​org/​10.​2337/​dc16-0645. The study proposes the use of biomarkers in addition to clinical criteria as a useful approach to better identify cases of monogenic diabetes. PubMedPubMedCentralCrossRef
11.
Kanakatti Shankar R, Pihoker C, Dolan LM, Standiford D, Badaru A, Dabelea D, et al. Permanent neonatal diabetes mellitus: prevalence and genetic diagnosis in the SEARCH for Diabetes in Youth Study. 2013;14(3):174–80. https://​doi.​org/​10.​1111/​pedi.​12003.
12.
Tattersall RB, Fajans SS. A difference between the inheritance of classical juvenile-onset and maturity-onset type diabetes of young people. Diabetes. 1975;24(1):44–53. https://​doi.​org/​10.​2337/​diabetes.​24.​1.​44.PubMedCrossRef
13.
Fajans SS, Bell GI. MODY: history, genetics, pathophysiology, and clinical decision making. Diabetes Care. 2011;34(8):1878–84. https://​doi.​org/​10.​2337/​dc11-0035.PubMedPubMedCentralCrossRef
14.
Bellanné-Chantelot C, Lévy DJ, Carette C, Saint-Martin C, Riveline J-P, Larger E, et al. Clinical characteristics and diagnostic criteria of maturity-onset diabetes of the young (MODY) due to molecular anomalies of the HNF1A gene. J Clin Endocrinol Metab. 2011;96(8):E1346–51. https://​doi.​org/​10.​1210/​jc.​2011-0268.PubMedCrossRef
15.
Shields BM, Hicks S, Shepherd MH, Colclough K, Hattersley AT, Ellard S. Maturity-onset diabetes of the young (MODY): how many cases are we missing? Diabetologia. 2010;53(12):2504–8. https://​doi.​org/​10.​1007/​s00125-010-1799-4.PubMedCrossRef
16.
•• Kleinberger JW, Copeland KC, Gandica RG, Haymond MW, Levitsky LL, Linder B, et al. Monogenic diabetes in overweight and obese youth diagnosed with type 2 diabetes: the TODAY clinical trial. Nat Publ Group. 2017:1–8. https://​doi.​org/​10.​1038/​gim.​2017.​150. This study shows that a monogenic cause should be considered in children and adolescents with atypical diabetes regardless of BMI.
17.
Harjutsalo V, Lammi N, Karvonen M, Groop P-H. Age at onset of type 1 diabetes in parents and recurrence risk in offspring. Diabetes. 2010;59(1):210–4. https://​doi.​org/​10.​2337/​db09-0344.PubMedCrossRef
18.
Warram JH, Krolewski AS, Gottlieb MS, Kahn CR. Differences in risk of insulin-dependent diabetes in offspring of diabetic mothers and diabetic fathers. N Engl J Med. 1984;311(3):149–52. https://​doi.​org/​10.​1056/​NEJM198407193110​304.PubMedCrossRef
19.
McDonald TJ, Shields BM, Lawry J, Owen KR, Gloyn AL, Ellard S, et al. High-sensitivity CRP discriminates HNF1A-MODY from other subtypes of diabetes. Diabetes Care. 2011;34(8):1860–2. https://​doi.​org/​10.​2337/​dc11-0323.PubMedPubMedCentralCrossRef
20.
Bacon S, Kyithar MP, Rizvi SR, Donnelly E, McCarthy A, Burke M, et al. Successful maintenance on sulphonylurea therapy and low diabetes complication rates in a HNF1A-MODY cohort. Diabet Med. 2016;33(7):976–84. https://​doi.​org/​10.​1111/​dme.​12992.PubMedCrossRef
21.
Pearson ER, Liddell WG, Shepherd M, Corrall RJ, Hattersley AT. Sensitivity to sulphonylureas in patients with hepatocyte nuclear factor-1alpha gene mutations: evidence for pharmacogenetics in diabetes. Diabet Med. 2000;17(7):543–5.PubMedCrossRef
22.
Shepherd M, Pearson ER, Houghton J, Salt G, Ellard S, Hattersley AT. No deterioration in glycemic control in HNF-1alpha maturity-onset diabetes of the young following transfer from long-term insulin to sulphonylureas. Diabetes Care. 2003;26(11):3191–2.PubMedCrossRef
23.
Shepherd M, Shields B, Ellard S, Rubio-Cabezas O, Hattersley AT. A genetic diagnosis of HNF1Adiabetes alters treatment and improves glycaemic control in the majority of insulin-treated patients. Diabet Med. 2009;26(4):437–41. https://​doi.​org/​10.​1111/​j.​1464-5491.​2009.​02690.​x.PubMedCrossRef
24.
Greeley SAW, John PM, Winn AN, Ornelas J, Lipton RB, Philipson LH, et al. The cost-effectiveness of personalized genetic medicine: the case of genetic testing in neonatal diabetes. Diabetes Care. 2011;34(3):622–7. https://​doi.​org/​10.​2337/​dc10-1616.PubMedPubMedCentralCrossRef
25.
Naylor RN, John PM, Winn AN, Carmody D, Greeley SAW, Philipson LH, et al. Cost-effectiveness of MODY genetic testing: translating genomic advances into practical health applications. Diabetes Care. 2014;37(1):202–9. https://​doi.​org/​10.​2337/​dc13-0410.PubMedCrossRef
26.
•• Nguyen HV, Finkelstein EA, Mital S, Gardner DS-L. Incremental cost-effectiveness of algorithm-driven genetic testing versus no testing for Maturity Onset Diabetes of the Young (MODY) in Singapore. J Med Genet. 2017;54(11):747–53. https://​doi.​org/​10.​1136/​jmedgenet-2017-104670. This study provides another important example of the cost-effectiveness of genetic testing for MODY. PubMedCrossRef
27.
Schnyder S, Mullis PE, Ellard S, Hattersley AT, Flück CE. Genetic testing for glucokinase mutations in clinically selected patients with MODY: a worthwhile investment. Swiss Med Wkly. 2005;135(23–24):352–6.PubMed
28.
Kleinberger JW, Pollin TI. Undiagnosed MODY: Time for action. Current Diabetes Reports. 2015;15(12):458–11. https://​doi.​org/​10.​1007/​s11892-015-0681-7.​ CrossRef
29.
Naylor R, Philipson LH. Who should have genetic testing for maturity-onset diabetes of the young? Clin Endocrinol. 2011;75(4):422–6. https://​doi.​org/​10.​1111/​j.​1365-2265.​2011.​04049.​x.CrossRef
30.
Shepherd M, Sparkes AC, Hattersley AT. Genetic testing in maturity onset diabetes of the young (MODY): a new challenge for the diabetic clinic. Practical Diabetes International. Pract Diabetes Int 2001;18(1):16–21. https://​doi.​org/​10.​1002/​pdi.​108.
31.
Carroll R, Murphy R. Monogenic diabetes: a diagnostic algorithm for clinicians. Genes (Basel). 2013;4(4):522–35. https://​doi.​org/​10.​3390/​genes4040522.CrossRef
32.
Vaxillaire M, Froguel P. Monogenic diabetes: implementation of translational genomic research towards precision medicine. Journal of Diabetes. 2016;8(6):782–95. https://​doi.​org/​10.​1111/​1753-0407.​12446.PubMedCrossRef
33.
McDonald TJ, Ellard S. Maturity onset diabetes of the young: identification and diagnosis. Ann Clin Biochem. 2013;50(Pt 5):403–15. https://​doi.​org/​10.​1177/​0004563213483458​.​ PubMedCrossRef
34.
Matschinsky FM. Regulation of pancreatic β-cell glucokinase: from basics to therapeutics. Diabetes. 2002;51(suppl 3):S394–404. https://​doi.​org/​10.​2337/​diabetes.​51.​2007.​S394.PubMedCrossRef
35.
Stride A, Vaxillaire M, Tuomi T, Barbetti F, Njølstad PR, Hansen T, et al. The genetic abnormality in the beta cell determines the response to an oral glucose load. Diabetologia. 2002;45(3):427–35. https://​doi.​org/​10.​1007/​s00125-001-0770-9.PubMedCrossRef
36.
Steele AM, Wensley KJ, Ellard S, Murphy R, Shepherd M, Colclough K, et al. Use of HbA1c in the identification of patients with hyperglycaemia caused by a glucokinase mutation: observational case control studies. PLoS One. 2013;8(6):e65326. https://​doi.​org/​10.​1371/​journal.​pone.​0065326.PubMedPubMedCentralCrossRef
37.
Spégel P, Ekholm E, Tuomi T, Groop L, Mulder H, Filipsson K. Metabolite profiling reveals normal metabolic control in carriers of mutations in the glucokinase gene (MODY2). Diabetes. 2013;62(2):653–61. https://​doi.​org/​10.​2337/​db12-0827.PubMedPubMedCentralCrossRef
38.
Steele AM, Shields BM, Wensley KJ, Colclough K, Ellard S, Hattersley AT. Prevalence of vascular complications among patients with glucokinase mutations and prolonged, mild hyperglycemia. JAMA. 2014;311(3):279–86. https://​doi.​org/​10.​1001/​jama.​2013.​283980.PubMedCrossRef
39.
Chakera AJ, Steele AM, Gloyn AL, Shepherd MH, Shields B, Ellard S, et al. Recognition and management of individuals with hyperglycemia because of a heterozygous glucokinase mutation. Diabetes Care. 2015;38(7):1383–92. https://​doi.​org/​10.​2337/​dc14-2769.PubMedCrossRef
40.
Dickens LT, Naylor RN. Clinical management of women with monogenic diabetes during pregnancy. Curr Diab Rep. 2018;18(3):12. https://​doi.​org/​10.​1007/​s11892-018-0982-8.PubMedCrossRef
41.
Stride A, Shields B, Gill-Carey O, Chakera AJ, Colclough K, Ellard S, et al. Cross-sectional and longitudinal studies suggest pharmacological treatment used in patients with glucokinase mutations does not alter glycaemia. Diabetologia. 2014;57(1):54–6. https://​doi.​org/​10.​1007/​s00125-013-3075-x.PubMedCrossRef
42.
Carmody D, Naylor RN, Bell CD, Berry S, Montgomery JT, Tadie EC, et al. GCK-MODY in the US National Monogenic Diabetes Registry: frequently misdiagnosed and unnecessarily treated. Acta Diabetol. 2016;53(5):703–8. https://​doi.​org/​10.​1007/​s00592-016-0859-8.PubMedPubMedCentralCrossRef
43.
Wang H, Maechler P, Antinozzi PA, Hagenfeldt KA, Wollheim CB. Hepatocyte nuclear factor 4alpha regulates the expression of pancreatic beta -cell genes implicated in glucose metabolism and nutrient-induced insulin secretion. J Biol Chem. 2000;275(46):35953–9. https://​doi.​org/​10.​1074/​jbc.​M006612200.PubMedCrossRef
44.
Stoffel M, Duncan SA. The maturity-onset diabetes of the young (MODY1) transcription factor HNF4alpha regulates expression of genes required for glucose transport and metabolism. Proc Natl Acad Sci U S A. 1997;94(24):13209–14.PubMedPubMedCentralCrossRef
45.
Colclough K, Bellanné-Chantelot C, Saint-Martin C, Flanagan SE, Ellard S. Mutations in the genes encoding the transcription factors hepatocyte nuclear factor 1 alpha and 4 alpha in maturity-onset diabetes of the young and hyperinsulinemic hypoglycemia. Hum Mutat. 2013;34(5):669–85. https://​doi.​org/​10.​1002/​humu.​22279.PubMedCrossRef
46.
Pontoglio M, Prié D, Cheret C, Doyen A, Leroy C, Froguel P, et al. HNF1alpha controls renal glucose reabsorption in mouse and man. EMBO Rep. 2000;1(4):359–65. https://​doi.​org/​10.​1093/​embo-reports/​kvd071.PubMedPubMedCentralCrossRef
47.
Bellanné-Chantelot C, Carette C, Riveline J-P, Valéro R, Gautier J-F, Larger E, et al. The type and the position of HNF1A mutation modulate age at diagnosis of diabetes in patients with maturity-onset diabetes of the young (MODY)-3. Diabetes. 2008;57(2):503–8. https://​doi.​org/​10.​2337/​db07-0859.PubMedCrossRef
48.
Shih DQ, Bussen M, Sehayek E, Ananthanarayanan M, Shneider BL, Suchy FJ, et al. Hepatocyte nuclear factor-1alpha is an essential regulator of bile acid and plasma cholesterol metabolism. Nat Genet. 2001;27(4):375–82. https://​doi.​org/​10.​1038/​86871.PubMedCrossRef
49.
Steele AM, Shields BM, Shepherd M, Ellard S, Hattersley AT, Pearson ER. Increased all-cause and cardiovascular mortality in monogenic diabetes as a result of mutations in the HNF1A gene. Diabet Med. 2010;27(2):157–61. https://​doi.​org/​10.​1111/​j.​1464-5491.​2009.​02913.​x.PubMedCrossRef
50.
Thanabalasingham G, Shah N, Vaxillaire M, Hansen T, Tuomi T, Gašperíková D, et al. A large multi-centre European study validates high-sensitivity C-reactive protein (hsCRP) as a clinical biomarker for the diagnosis of diabetes subtypes. Diabetologia. 2011;54(11):2801–10. https://​doi.​org/​10.​1007/​s00125-011-2261-y.PubMedCrossRef
51.
Jeannot E, Mellottee L, Bioulac-Sage P, Balabaud C, Scoazec J-Y, Tran Van Nhieu J, et al. Spectrum of HNF1A somatic mutations in hepatocellular adenoma differs from that in patients with MODY3 and suggests genotoxic damage. Diabetes. 2010;59(7):1836–44. https://​doi.​org/​10.​2337/​db09-1819.PubMedPubMedCentralCrossRef
52.
Reznik Y, Dao T, Coutant R, Chiche L, Jeannot E, Clauin S, et al. Hepatocyte nuclear factor-1 alpha gene inactivation: cosegregation between liver adenomatosis and diabetes phenotypes in two maturity-onset diabetes of the young (MODY)3 families. J Clin Endocrinol Metab. 2004;89(3):1476–80. https://​doi.​org/​10.​1210/​jc.​2003-031552.PubMedCrossRef
53.
Tuomi T, Honkanen EH, Isomaa B, Sarelin L, Groop LC. Improved prandial glucose control with lower risk of hypoglycemia with nateglinide than with glibenclamide in patients with maturity-onset diabetes of the young type 3. Diabetes Care. 2006;29(2):189–94.PubMedCrossRef
54.
Becker M, Galler A, Raile K. Meglitinide analogues in adolescent patients with HNF1A-MODY (MODY 3). Pediatrics. 2014;133(3):e775–9. https://​doi.​org/​10.​1542/​peds.​2012-2537.PubMedCrossRef
55.
Ostoft SH, Bagger JI, Hansen T, Pedersen O, Faber J, Holst JJ, et al. Glucose-lowering effects and low risk of hypoglycemia in patients with maturity-onset diabetes of the young when treated with a GLP-1 receptor agonist: a double-blind, randomized, crossover trial. Diabetes Care. 2014;37(7):1797–805. https://​doi.​org/​10.​2337/​dc13-3007.​ PubMedCrossRef
56.
Hohendorff J, Szopa M, Skupien J, Kapusta M, Zapala B, Platek T, et al. A single dose of dapagliflozin, an SGLT-2 inhibitor, induces higher glycosuria in GCK- and HNF1A-MODY than in type 2 diabetes mellitus. Endocrine. 2017;57(2):272–9. https://​doi.​org/​10.​1007/​s12020-017-1341-2.PubMedPubMedCentralCrossRef
57.
Carette C, Dubois-Laforgue D, Saint-Martin C, Clauin S, Beaufils S, Larger E, et al. Familial young-onset forms of diabetes related to HNF4A and rare HNF1A molecular aetiologies. Diabet Med. 2010;27(12):1454–8. https://​doi.​org/​10.​1111/​j.​1464-5491.​2010.​03115.​x.
58.
Yamagata K, Furuta H, Oda N, Kaisaki PJ, Menzel S, Cox NJ, et al. Mutations in the hepatocyte nuclear factor-4alpha gene in maturity-onset diabetes of the young (MODY1). Nature. 1996;384(6608):458–60. https://​doi.​org/​10.​1038/​384458a0.PubMedCrossRef
59.
Dusátková P, Pruhova S, Sumnik Z, Kolousková S, Obermannova B, Cinek O, et al. HNF1A mutation presenting with fetal macrosomia and hypoglycemia in childhood prior to onset of overt diabetes. J Pediatr Endocrinol Metab. 2011;24(5–6):377–9.PubMed
60.
Pearson ER, Boj SF, Steele AM, Barrett T, Stals K, Shield JP, et al. Macrosomia and hyperinsulinaemic hypoglycaemia in patients with heterozygous mutations in the HNF4A gene. PLoS Med. 2007;4(4):e118. https://​doi.​org/​10.​1371/​journal.​pmed.​0040118.PubMedPubMedCentralCrossRef
61.
Pearson ER, Pruhova S, Tack CJ, Johansen A, Castleden HAJ, Lumb PJ, et al. Molecular genetics and phenotypic characteristics of MODY caused by hepatocyte nuclear factor 4α mutations in a large European collection. Diabetologia. 2005;48(5):878–85. https://​doi.​org/​10.​1007/​s00125-005-1738-y.PubMedCrossRef
62.
Bellanne-Chantelot C, Clauin S, Chauveau D, Collin P, Daumont M, Douillard C, et al. Large genomic rearrangements in the hepatocyte nuclear factor-1 (TCF2) gene are the most frequent cause of maturity-onset diabetes of the young type 5. Diabetes. 2005;54(11):3126–32. https://​doi.​org/​10.​2337/​diabetes.​54.​11.​3126.PubMedCrossRef
63.
Chambers C, Fouts A, Dong F, Colclough K, Wang Z, Batish SD, et al. Characteristics of maturity onset diabetes of the young in a large diabetes center. 2015;17(5):360–7. https://​doi.​org/​10.​1111/​pedi.​12289.
64.
Pearson ER, Badman MK, Lockwood CR, Clark PM, Ellard S, Bingham C, et al. Contrasting diabetes phenotypes associated with hepatocyte nuclear factor-1a and -1b mutations. Diabetes Care. 2004;27(5):1102–7. https://​doi.​org/​10.​2337/​diacare.​27.​5.​1102.PubMedCrossRef
65.
Bellanné-Chantelot C, Chauveau D, Gautier J-F, Dubois-Laforgue D, Clauin S, Beaufils S, et al. Clinical spectrum associated with hepatocyte nuclear factor-1beta mutations. Ann Intern Med. 2004;140(7):510–7.PubMedCrossRef
66.
Clissold RL, Hamilton AJ, Hattersley AT, Ellard S, Bingham C. HNF1B-associated renal and extra-renal disease—an expanding clinical spectrum. Nat Publ Group. 2014;11(2):102–12. https://​doi.​org/​10.​1038/​nrneph.​2014.​232.CrossRef
67.
Weber S, Moriniere V, Knuppel T, Charbit M, Dusek J, Ghiggeri GM, et al. Prevalence of mutations in renal developmental genes in children with renal hypodysplasia: results of the ESCAPE study. J Am Soc Nephrol. 2006;17(10):2864–70. https://​doi.​org/​10.​1681/​ASN.​2006030277.PubMedCrossRef
68.
Heidet L, Decramer S, Pawtowski A, Moriniere V, Bandin F, Knebelmann B, et al. Spectrum of HNF1B mutations in a large cohort of patients who harbor renal diseases. Clin J Am Soc Nephrol. 2010;5(6):1079–90. https://​doi.​org/​10.​2215/​CJN.​06810909.​ PubMedPubMedCentralCrossRef
69.
Bingham C, Hattersley AT. Renal cysts and diabetes syndrome resulting from mutations in hepatocyte nuclear factor-1beta. Nephrol Dial Transplant. 2004;19(11):2703–8. https://​doi.​org/​10.​1093/​ndt/​gfh348.​ PubMedCrossRef
70.
Edghill EL, Bingham C, Ellard S, Hattersley AT. Mutations in hepatocyte nuclear factor-1beta and their related phenotypes. J Med Genet. 2006;43(1):84–90. https://​doi.​org/​10.​1136/​jmg.​2005.​032854.​ PubMedCrossRef
71.
Yu DD, Guo SW, Jing YY, Dong YL, Wei LX. A review on hepatocyte nuclear factor-1beta and tumor. Cell Biosci. 2015;5:58. https://​doi.​org/​10.​1186/​s13578-015-0049-3.PubMedPubMedCentralCrossRef
72.
Sun M, Tong P, Kong W, Dong B, Huang Y, Park IY, et al. HNF1B loss exacerbates the development of chromophobe renal cell carcinomas. Cancer Res. 2017;77(19):5313–26. https://​doi.​org/​10.​1158/​0008-5472.​CAN-17-0986.PubMedCrossRefPubMedCentral
73.
Wang CC, Mao TL, Yang WC, Jeng YM. Underexpression of hepatocyte nuclear factor-1beta in chromophobe renal cell carcinoma. Histopathology. 2013;62(4):589–94. https://​doi.​org/​10.​1111/​his.​12026.PubMedCrossRef
74.
Ferre S, Bongers EM, Sonneveld R, Cornelissen EA, van der Vlag J, van Boekel GA, et al. Early development of hyperparathyroidism due to loss of PTH transcriptional repression in patients with HNF1beta mutations? J Clin Endocrinol Metab. 2013;98(10):4089–96. https://​doi.​org/​10.​1210/​jc.​2012-3453.PubMedCrossRef
75.
Loirat C, Bellanne-Chantelot C, Husson I, Deschenes G, Guigonis V, Chabane N. Autism in three patients with cystic or hyperechogenic kidneys and chromosome 17q12 deletion. Nephrol Dial Transplant. 2010;25(10):3430–3. https://​doi.​org/​10.​1093/​ndt/​gfq380.PubMedCrossRef
76.
Raile K, Klopocki E, Holder M, Wessel T, Galler A, Deiss D, et al. Expanded clinical spectrum in hepatocyte nuclear factor 1b-maturity-onset diabetes of the young. J Clin Endocrinol Metab. 2009;94(7):2658–64. https://​doi.​org/​10.​1210/​jc.​2008-2189.PubMedCrossRef
77.
Clissold RL, Shaw-Smith C, Turnpenny P, Bunce B, Bockenhauer D, Kerecuk L, et al. Chromosome 17q12 microdeletions but not intragenic HNF1B mutations link developmental kidney disease and psychiatric disorder. Kidney Int. 2016;90(1):203–11. https://​doi.​org/​10.​1016/​j.​kint.​2016.​03.​027.PubMedPubMedCentralCrossRef
78.
Brissova M, Shiota M, Nicholson WE, Gannon M, Knobel SM, Piston DW, et al. Reduction in pancreatic transcription factor PDX-1 impairs glucose-stimulated insulin secretion. J Biol Chem. 2002;277(13):11225–32. https://​doi.​org/​10.​1074/​jbc.​M111272200.PubMedCrossRef
79.
Ohlsson H, Karlsson K, Edlund T. IPF1, a homeodomain-containing transactivator of the insulin gene. EMBO J. 1993;12(11):4251–9.PubMedPubMedCentralCrossRef
80.
Fajans SS, Bell GI, Paz VP, Below JE, Cox NJ, Martin C, et al. Obesity and hyperinsulinemia in a family with pancreatic agenesis and MODY caused by the IPF1 mutation Pro63fsX60. Transl Res. 2010;156(1):7–14. https://​doi.​org/​10.​1016/​j.​trsl.​2010.​03.​003.PubMedPubMedCentralCrossRef
81.
Hansen L. Missense mutations in the human insulin promoter factor-1 gene and their relation to maturity-onset diabetes of the young and late-onset type 2 diabetes mellitus in Caucasians. J Clin Endocrinol Metab. 2000;85(3):1323–6. https://​doi.​org/​10.​1210/​jc.​85.​3.​1323.​ PubMedCrossRef
82.
Flanagan SE, Patch AM, Mackay DJG, Edghill EL, Gloyn AL, Robinson D, et al. Mutations in ATP-sensitive K+ channel genes cause transient neonatal diabetes and permanent diabetes in childhood or adulthood. Diabetes. 2007;56(7):1930–7. https://​doi.​org/​10.​2337/​db07-0043.PubMedCrossRef
83.
Gloyn AL, Pearson ER, Antcliff JF, Proks P, Bruining GJ, Slingerland AS, et al. Activating mutations in the gene encoding the ATP-sensitive potassium-channel subunit Kir6.2 and permanent neonatal diabetes. 2009;350(18):1838–49. https://​doi.​org/​10.​1056/​NEJMoa032922.
84.
Kapoor RR, Flanagan SE, James CT, McKiernan J, Thomas AM, Harmer SC, et al. Hyperinsulinaemic hypoglycaemia and diabetes mellitus due to dominant ABCC8/KCNJ11 mutations. Diabetologia. 2011;54(10):2575–83. https://​doi.​org/​10.​1007/​s00125-011-2207-4.PubMedPubMedCentralCrossRef
85.
Bonnefond A, Philippe J, Durand E, Dechaume A, Huyvaert M, Montagne L, et al. Whole-exome sequencing and high throughput genotyping identified KCNJ11 as the thirteenth MODY gene. PLoS One. 2012;7(6):e37423–8. https://​doi.​org/​10.​1371/​journal.​pone.​0037423.PubMedPubMedCentralCrossRef
86.
Bowman P, Flanagan SE, Edghill EL, Damhuis A, Shepherd MH, Paisey R, et al. Heterozygous ABCC8 mutations are a cause of MODY. Diabetologia. 2011;55(1):123–7. https://​doi.​org/​10.​1007/​s00125-011-2319-x.PubMedCrossRef
87.
Ovsyannikova AK, Rymar OD, Shakhtshneider EV, Klimontov VV, Koroleva EA, Myakina NE, et al. ABCC8-related maturity-onset diabetes of the young (MODY12): clinical features and treatment perspective. Diabetes Ther. 2016;7(3):591–600. https://​doi.​org/​10.​1007/​s13300-016-0192-9.PubMedPubMedCentralCrossRef
88.
Molven A, Ringdal M, Nordbø AM, Ræder H, Støy J, Lipkind GM, et al. Mutations in the insulin gene can cause MODY and autoantibody-negative type 1 diabetes. Diabetes. 2008;57(4):1131–5. https://​doi.​org/​10.​2337/​db07-1467.PubMedCrossRef
89.
Boesgaard TW, Pruhova S, Andersson EA, Cinek O, Obermannova B, Lauenborg J, et al. Further evidence that mutations in INS can be a rare cause of maturity-onset diabetes of the young (MODY). BMC Med Genet. 2010;11(1):42. https://​doi.​org/​10.​1186/​1471-2350-11-42.​ PubMedPubMedCentralCrossRef
90.
Edghill EL, Flanagan SE, Patch A-M, Boustred C, Parrish A, Shields B, et al. Insulin mutation screening in 1,044 patients with diabetes: mutations in the INS gene are a common cause of neonatal diabetes but a rare cause of diabetes diagnosed in childhood or adulthood. Diabetes. 2008;57(4):1034–42. https://​doi.​org/​10.​2337/​db07-1405.PubMedCrossRef
91.
Meur G, Simon A, Harun N, Virally M, Dechaume A, Bonnefond A, et al. Insulin gene mutations resulting in early-onset diabetes: marked differences in clinical presentation, metabolic status, and pathogenic effect through endoplasmic reticulum retention. Diabetes. 2010;59(3):653–61. https://​doi.​org/​10.​2337/​db09-1091.PubMedCrossRef
92.
Barrett TG. Differential diagnosis of type 1 diabetes: which genetic syndromes need to be considered? Pediatr Diabetes. 2007;8(Suppl 6):15–23. https://​doi.​org/​10.​1111/​j.​1399-5448.​2007.​00278.​x.PubMedCrossRef
93.
Y-i G, Nonaka I, Horai S. A mutation in the tRNALeu(UUR) gene associated with the MELAS subgroup of mitochondrial encephalomyopathies. Nature. 1990;348(6302):651–3. https://​doi.​org/​10.​1038/​348651a0.CrossRef
94.
Murphy R, Turnbull DM, Walker M, Hattersley AT. Clinical features, diagnosis and management of maternally inherited diabetes and deafness (MIDD) associated with the 3243A>G mitochondrial point mutation. Diabet Med. 2008;25(4):383–99. https://​doi.​org/​10.​1111/​j.​1464-5491.​2008.​02359.​x.PubMedCrossRef
95.
Sivitz WI, Yorek MA. Mitochondrial dysfunction in diabetes: from molecular mechanisms to functional significance and therapeutic opportunities. Antioxid Redox Signal. 2010;12(4):537–77. https://​doi.​org/​10.​1089/​ars.​2009.​2531.PubMedPubMedCentralCrossRef
96.
Supale S, Li N, Brun T, Maechler P. Mitochondrial dysfunction in pancreatic β cells. Trends Endocrinol Metab. 2012;23(9):477–87. https://​doi.​org/​10.​1016/​j.​tem.​2012.​06.​002.​ PubMedCrossRef
97.
Maassen JA, Janssen GMC, Hart LM. Molecular mechanisms of mitochondrial diabetes (MIDD). Ann Med. 2009;37(3):213–21. https://​doi.​org/​10.​1080/​0785389051000718​8.CrossRef
98.
Guillausseau PJ, Massin P, Dubois-Laforgue D, Timsit J, Virally M, Gin H, et al. Maternally inherited diabetes and deafness: a multicenter study. Ann Intern Med. 2001;134(9 Pt 1):721–8.PubMedCrossRef
99.
Maassen JA, ‘T Hart LM, Van Essen E, Heine RJ, Nijpels G, Jahangir Tafrechi RS, et al. Mitochondrial diabetes: molecular mechanisms and clinical presentation. Diabetes. 2004;53(Suppl 1):S103–9.PubMedCrossRef
100.
Suzuki Y, Kobayashi T, Taniyama M, Astumi Y, Oka Y, Kadowaki T, et al. Islet cell antibody in mitochondrial diabetes. Diabetes Res Clin Pract. 1997;35(2–3):163–5. https://​doi.​org/​10.​1016/​S0168-8227(97)01378-8.
101.
Suzuki Y, Taniyama M, Shimada A, Atumi Y, Matsuoka K, Oka Y. GAD antibody in mitochondrial diabetes associated with tRNA(UUR) mutation at position 3271. Diabetes Care. 2002;25(6):1097–8. https://​doi.​org/​10.​2337/​diacare.​25.​6.​1097.PubMedCrossRef
102.
van ven Ouweland JMW, Cryns K, Pennings RJE, Walraven I, Janssen GMC, Maassen JA, et al. Molecular characterization of WFS1 in patients with Wolfram syndrome. The Journal of Molecular Diagnostics : JMD. 2003;5(2):88–95.CrossRef
103.
Urano F. Wolfram syndrome iPS cells: the first human cell model of endoplasmic reticulum disease. Diabetes. 2014;63(3):844–6. https://​doi.​org/​10.​2337/​db13-1809.PubMedPubMedCentralCrossRef
104.
Bonnycastle LL, Chines PS, Hara T, Huyghe JR, Swift AJ, Heikinheimo P, et al. Autosomal dominant diabetes arising from a Wolfram syndrome 1 mutation. Diabetes. 2013;62(11):3943–50. https://​doi.​org/​10.​2337/​db13-0571.PubMedPubMedCentralCrossRef
105.
Minton JAL, Rainbow LA, Ricketts C, Barrett TG. Wolfram syndrome. Rev Endocr Metab Disord. 2003;4(1):53–9.PubMedCrossRef
106.
Cano A, Rouzier C, Monnot S, Chabrol B, Conrath J, Lecomte P, et al. Identification of novel mutations in WFS1 and genotype-phenotype correlation in Wolfram syndrome. Am J Med Genet A. 2007;143A(14):1605–12. https://​doi.​org/​10.​1002/​ajmg.​a.​31809.PubMedCrossRef
107.
Zmyslowska A, Borowiec M, Fichna P, Iwaniszewska B, Majkowska L, Pietrzak I, et al. Delayed recognition of Wolfram syndrome frequently misdiagnosed as type 1 diabetes with early chronic complications. Exp Clin Endocrinol Diabetes. 2014;122(1):35–8. https://​doi.​org/​10.​1055/​s-0033-1357160.PubMedCrossRef
108.
Zmyslowska A, Fendler W, Szadkowska A, Borowiec M, Mysliwiec M, Baranowska-Jazwiecka A, et al. Glycemic variability in patients with Wolfram syndrome is lower than in type 1 diabetes. Acta Diabetol. 2015;52(6):1057–62. https://​doi.​org/​10.​1007/​s00592-015-0757-5.PubMedPubMedCentralCrossRef
109.
Satman I, Yilmaz MT, Gürsoy N, Karşidağ K, Dinççağ N, Ovali T, et al. Evaluation of insulin resistant diabetes mellitus in Alström syndrome: a long-term prospective follow-up of three siblings. Diabetes Res Clin Pract. 2002;56(3):189–96.PubMedCrossRef
110.
Marshall JD, Maffei P, Beck S, Barrett TG, Paisey R, Naggert JK. Clinical utility gene card for: Alström syndrome—update 2013. Eur J Hum Genet. 2013;21(11):3–4. https://​doi.​org/​10.​1038/​ejhg.​2013.​61.CrossRef
111.
Marshall JD, Muller J, Collin GB, Milan G, Kingsmore SF, Dinwiddie D, et al. Alström syndrome: mutation spectrum of ALMS1. Hum Mutat. 2015;36(7):660–8. https://​doi.​org/​10.​1002/​humu.​22796.PubMedPubMedCentralCrossRef

Be confident that your patient care is up to date

Medicine Matters is being incorporated into Springer Medicine, our new medical education platform. 

Alongside the news coverage and expert commentary you have come to expect from Medicine Matters diabetes, Springer Medicine's complimentary membership also provides access to articles from renowned journals and a broad range of Continuing Medical Education programs. Create your free account »