Skip to main content

OPINION article

Front. Endocrinol., 02 July 2018
Sec. Clinical Diabetes

Hidden MODY—Looking for a Needle in a Haystack

  • 1Second Department of Internal Medicine, Third Faculty of Medicine, Center for Research of Diabetes, Metabolism and Nutrition, University Hospital Královské Vinohrady, Prague, Czechia
  • 2Department of Internal Medicine, Second Faculty of Medicine, Charles University, Prague, Czechia

Introduction

MODY (Maturity onset diabetes of the young) is a specific type of diabetes caused by mutation in a single gene, involved in the development and function of the β-cells, inherited in an autosomal dominant manner (1). Out of fourteen, up to date discovered, MODY genes (2) the most often affected ones include GCK (gene encoding glucokinase enzyme) and HNF1A (encoding the transcription factor—hepatocyte nuclear factor 1α), which altogether account for approximately 80% of all MODY cases (3). Mutations in other genes (e.g., HNF4A or HNF1B—hepatocyte nuclear factor 4α and 1β), occur rarely (4).

Although MODY represents a rather scarce diabetes type (1), searching for MODY among much more prevalent forms of diabetes is important and desirable for its clear impact on clinical practice—for appropriate diabetes management with the most suitable treatment (accompanied with improved quality of life) (5, 6), for assessing the real risk of development and progression of specific diabetic complications in each MODY type, as well as for early diagnosis in the patient's relatives and offspring.

Nevertheless, overwhelming majority of MODY patients worldwide remains misdiagnosed (3, 7). Moreover, no unitary and up-to-date diagnostic guidelines have been established so far, and also it is not obvious, which approach to correct identification of MODY patients is optimal.

The aim of this communication is to present our experiences with searching for patients with MODY in the context of current diagnostic proceedings and actual study outputs available.

How to Recognize MODY Among Diabetic Patients?

MODY is traditionally characterized by the following features (8):

• early onset of diabetes (i.e., <25 years of age);

• insulin-independence (i.e., preserved endogenous insulin secretion >5 years of diabetes duration);

• and autosomal dominant inheritance (i.e., family history of glycaemic disorders at least in two consecutive generations).

Thus, a young patient diagnosed with diabetes before the age of 25 years with a marked family history of diabetes in one parental line and signs of ongoing insulin secretion (the absence of ketoacidosis/DKA at the time of diabetes onset and later on /under circumstances significantly increasing its risk or detectable C-peptide for years and obviously low doses of insulin required) should be suspected of MODY and recommended to molecular genetic testing (8); especially when islet autoimmunity and insulin resistance are not simultaneously present, so the patient doesn't fit either to type 1 or 2 type diabetes.

Clinical Characteristics of MODY

GCK-MODY is suspected in patients diagnosed with mild and asymptomatic fasting hyperglycaemia (typically 5.4–8.3 mmol/L) with relatively low glycated hemoglobin/HbA1c level (ranging usually 40–60 mmol/mol) (8, 9). In most cases it is detected incidentally during routine blood tests in adolescence or adulthood (although hyperglycaemia is present already from birth) (10). In younger age, however, the most prevalent type 1 diabetes (11) should be excluded; whereas the likelihood of type 2 diabetes grows with older age or obesity. In our experience, suspicion of GCK-MODY in contrast to type 1 diabetes at the time of diagnosis increases in the absence of DKA and islet autoantibodies, normal level of fasting C-peptide and presence of diabetes in one parent, whereas in contrast to type 2 diabetes GCK-MODY is suspected in rather normal body mass index/BMI, the absence of insulin resistance (e.g., dyslipidemia, hypertension) or modest rise in 2-h glucose level on oGTT/oral glucose tolerance test (< 4.6 mmol/L in 95% of cases, 12). On the contrary, signs that point out to GCK-MODY in patients with already established diagnosis of diabetes (labeled falsely as type 1 or type 2), in our opinion, include non-progressive character of glycaemic disorder (relatively stable glycaemia and HbA1c), non-decreasing (normal) endogenous insulin secretion, low efficacy of oral antidiabetics (e.g., metformin) administered or relatively low doses of insulin required and absence of vascular complications after many years of diabetes duration. Clinical picture of GCK-MODY could be, however, modified in some adult patients by their genetic background (e.g., polymorphisms in other diabetes-related genes), which could accentuate some features more typical for type 2 diabetes.

HNF1A-MODY is characterized by progressive glucose dysregulation, clinically presented at first with postprandial, later fasting hyperglycaemia, usually during adolescence or early adulthood (1, 8); thus, a large glucose increment of > 5 mmol/L can be observed on oGTT (12) in the early stages of the disease, whereas fasting blood glucose levels remain normal (14). Due to low renal threshold for glucose, glycosuria following a carbohydrate load often occurs, even before the clinical features of diabetes become apparent (14). Individuals with HNF1A-MODY similarly to other patients with diabetes can present with typical hyperglycaemia-related symptoms as polyuria, polydipsia and weight loss. Early signs that make diagnosis of HNF1A-MODY (in contrast to type 1 diabetes) possible include family history of diabetes, manifestation of diabetes without DKA, absence of islet antibodies, detectable C-peptide levels; similarly, younger age <30 years (11), lower BMI, presence of hypertension and triglycerides (15), higher HDL-cholesterol (>1.3 mmol/L) (16), eventually non-elevated fasting C-peptide level and marked sensitivity to SUR/sulphonylurea derivatives (5) are more indicative for HNF1A-MODY compared to type 2 diabetes.

In our opinion, level of fasting C-peptide measured at the time of diabetes manifestation is the most beneficial in discriminating MODY from type 1 diabetes if detected either very low (<150 pmol/L)—suggesting type 1 diabetes, or within normal range (300–600 pmol/L)—favoring MODY. Normal C-peptide, however, needn't necessarily exclude type 1 diabetes since endogenous insulin production in type 1 diabetes may be detectable for a variable length of time, including honeymoon period (17), particularly in LADA (late-onset autoimmune diabetes in adults). Thus, in case of uncertainty, to differentiate type 1 diabetes in the early stages we prefer the assessment of stimulated (post-meal) C-peptide (with levels >200 pmol/L showing to MODY as described (18, 26). Persistence of fasting C-peptide production 3–5 years after diagnosis further increases suspicion of MODY (26). On the contrary, we consider the detection of islet autoantibodies most useful soon after the diabetes diagnosis, as their prevalence in type 1 diabetes declines with the duration of the disease (19), whereas, in our experience, they could be detectable in some MODY patients even more than a decade after the diabetes onset, particularly in patients with poor glycaemic control (20).

Although significantly lower hsCRP level has been identified as the sign discriminating the patients with HNF1A-MODY from those with type 2 diabetes (21), we did not find this parameter useful in our clinical experience. Furthermore, some recent studies (comparing HNF1A-MODY to young-onset type 2 diabetes) challenged this paradigm, indicating that hsCRP does not improve diagnosis (7, 22). Moreover, hsCRP is not useful if elevated (>10 mg/L) as this usually indicates the presence of confounding inflammation, and should be repeated after a few weeks (23).

HNF4A-MODY shares similar clinical characteristic with HNF1A-MODY, except for the low renal threshold and possibly later diagnosis (24). We consider this type of MODY the most difficult to distinguish from type 2 diabetes, since HNF4A-MODY patients at the diagnosis may be older and may present with dyslipidaemia similar to patients with type 2 diabetes, particularly higher LDL- and lower HDL-cholesterol (4, 24). Differential diagnosis may facilitate higher birth weight (>4.4 kg at term), fetal macrosomy (56% of HNF4A-MODY individuals) and history of transient neonatal hyperinsulinaemic hypoglycaemia (15% of cases) (25).

Obstacles in Searching for MODY

Despite well-defined clinical presentation of MODY, reliability on clinical features alone seems insufficient in identification of MODY among other and more common types of diabetes. Less than 50% of all MODY cases actually meet traditional features of MODY (26). Moreover, clinical heterogeneity of MODY and possible overlapping clinical characteristics with type 1 and type 2 diabetes make the correct identification of MODY an uneasy task.

In accordance with Shields et al. (11) we consider having a parent with diabetes to be a very strong predictor of MODY compared with type 1 diabetes (increasing the probability of MODY by 23 times in those initially classified as type 1 diabetes). Though, in patients with MODY and type 2 diabetes, a positive family history of diabetes is reported in the same frequency (26, 27). On the other hand, the absence of diabetes in family members does not exclude MODY. Recent study has revealed much higher frequency of spontaneous de-novo mutations in GCK, HNF1A and HNF4A genes than previously assumed (28); moreover in GCK-MODY, discrete fasting hyperglycaemia may not be yet recognized in a parent, thus, family history could be seemingly negative. In a major part of our MODY patients at least one family member (parent, sibling) was identified. The only exception represents patients with HNF1B-MODY (in whom de-novo mutations occur more often) or accidental cases with unknown or prematurely dead relatives, eventually with so far unexamined glycaemia in parents.

Variable age at the time of diabetes onset, which is actually determined by many factors [e.g., the type, position and penetration of causal mutation in HNF1A and HNF4A genes (29), presence of risk polygenic single nucleotide polymorphisms of type 2 diabetes (30) or exposure to hyperglycaemia in utero (13)] represents another problem in differential diagnosis. For example, high penetrance of HNF1A mutation confers development of diabetes before 25 years of age in only 63% mutation carriers, but before the age of 35 in nearly 80% of individuals (4), whereas variable penetrance of HNF4A mutations cause that some of their carriers do not develop diabetes until the fourth decade of life (18). Thus, in patients with HNF1A/HNF4A mutation, diabetes may present later than before traditionally defined 25 years of age. Conversely, proportion of GCK-MODY in older populations (>35 years) is low and mild hyperglycaemia in adults is more likely to reflect type 2 diabetes than GCK-MODY (26, 31).

Due to globally increasing prevalence of obesity, expected non-obese habitus and absence of insulin resistance needn't necessarily apply to all MODY cases. Overweight, obesity and insulin resistance seem to be underestimated among MODY patients, at least in some populations (32, 33). However, in our cohort of patients referred for molecular genetic testing, BMI was usually just below the normal range among those with confirmed mutation in one of the MODY gene, probably because of the pre-selection of lean individuals for the testing. Nevertheless, in our own clinical practice, particularly overweight occurs more frequently in the older age groups.

Conflicting data exists on autoantibody positivity in MODY patients. Contrary to originally described low prevalence of islet autoantibodies in MODY (34), our own clinical experience (20), similarly to other authors (26, 35) shows that the prevalence of autoantibodies (especially GADA) in patients with confirmed MODY mutations is significantly higher. Thus, we fully agree with the recommendation that in the case of strong clinical suspicion, the presence of islet autoantibodies should not preclude genetic testing (23). Another diagnostic pitfall is the presence of DKA (36, 37).

Since strict adherence to traditional clinical criteria is insufficient in identifying MODY patients, other algorithms (10, 23, 26, 38), predictive models (11) or selected biomarkers (39) have been tested. Widening of the clinical criteria for genetic testing beyond current guidelines (to all patients diagnosed up to the age of 30 years, regardless of the family history of diabetes, GADA positivity, or insulin resistance) almost doubled the number of MODY diagnoses (26). We strongly recommend using Shield's predictive model (11), particularly in case of clinical uncertainty whether to indicate molecular genetic testing or not. Finally, biomarker-based screening pathway (combining assessment of endogenous insulin secretion with measurement of GADA and IA2A) increased the discriminatory ability and facilitated identification of even atypical cases and rarer forms of monogenic diabetes, which traditional criteria may miss (39) although as we have just pointed out the results obtained should be interpreted with caution. In Table 1, we tried to summarize typical clinical and laboratory features, together with some diagnostic pitfalls discussed above.

TABLE 1
www.frontiersin.org

Table 1. Rational approach to identify MODY patients—gradual evaluation of routinely available clinical and laboratory data increasing suspicion of MODY.

Conclusion

Substantial heterogeneity and overlapping in clinical features of MODY with other common types of diabetes considerably contribute to omission and misclassification of MODY in routine clinical praxis. Currently no uniform diagnostic algorithm exists and it is not evident which approach is optimal in identification of MODY patients. A comprehensive evaluation of patient history, disease manifestation and progression, treatment outcomes and routine laboratory parameters (considering the possible exceptions listed above) represents, in our experience, a rational approach in recognizing new patients with MODY and also individuals scattered for many years between patients with other types of diabetes.

Author Contributions

JU prepared the draft of the paper, LB and JB revisited it critically for important intellectual content.

Funding

The paper was supported by PROGRES Q36.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

Special thank to Rob Smith for language editing.

Abbreviations

BMI, body mass index; DKA, diabetic ketoacidosis; GADA, autoantibodies to glutamic acid decarboxylase; GCK, glucokinase; HbA1c, glycated haemoglobin; HDL, high density lipoprotein; HNF1A, hepatocyte nuclear factor 1 alpha; HNF1B, hepatocyte nuclear factor 1 beta; HNF4A, hepatocyte nuclear factor 4 alpha; hsCRP, high sensitive C-reactive protein; IA2A, autoantibodies to IA-2; LADA, Late-onset Autoimmune Diabetes in Adults; LDL, low density lipoprotein; MODY, Maturity Onset Diabetes of the Young; OGTT, oral glucose tolerance test; SUR, sulphonylurea derivatives.

References

1. Fajans SS, Bell GI. MODY: history, genetics, pathophysiology, and clinical decision making. Diabetes Care (2011) 34:1878–84. doi: 10.2337/dc11-0035

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Omim A. Online Mendelian Inheritance in Man. Maturity-Onset Diabetes of the Young; MODY. (2016). Available online at: http://mirror.omim.org/entry/606391 (Accessed March 19, 2018).

3. 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) 53:2504–8. doi: 10.1007/s00125-010-1799-4

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Murphy R, Ellard S, Hattersley AT. Clinical implications of a molecular genetic classification of monogenic β-cell diabetes. Nat Clin Pract Endocrinol Metab. (2008) 4:200–13. doi: 10.1038/ncpendmet0778

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Pearson ER, Liddell WG, Shepherd M, Hattersley AT. Sensitivity to sulphonylureas in patients with hepatocyte nuclear factor-1alpha gene mutations: evidence for pharmacogenetics in diabetes. Diab Med. (2000) 17:543–5. doi: 10.1046/j.1464-5491.2000.00305.x

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Brunerova L, Rahelic D, Ceriello A, Broz J. Use of oral antidiabetic drugs in the treatment of maturity-onset diabetes of the young (MODY): a minireview. Diabetes Metab Res Rev. (2018) 34:e2940. doi: 10.1002/dmrr.2940

CrossRef Full Text | Google Scholar

7. 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) 98:4055–62. doi: 10.1210/jc.2013-1279

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Ellard S, Bellanne-Chantelot C, Hattersley AT. Best practice guidelines for the molecular genetic diagnosis of maturity-onset diabetes of the young. Diabetologia (2008) 51:546–53. doi: 10.1007/s00125-008-0942-y

PubMed Abstract | CrossRef Full Text | Google Scholar

9. 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:e65326. doi: 10.1371/journal.pone.0065326

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Carroll RW, Murphy R. Monogenic diabetes: a diagnostic algorithm for clinicians. Genes (2013) 4:522–35. doi: 10.3390/genes4040522

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Shields BM, McDonald TJ, Ellard S, Campbell MJ, Hyde C, Hattersley AT. The development and validation of a clinical prediction model to determine the probability of MODY in patients with young-onset diabetes. Diabetologia (2012) 55:1265–72. doi: 10.1007/s00125-011-2418-8

PubMed Abstract | CrossRef Full Text | Google Scholar

12. 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) 45:427–35. doi: 10.1007/s00125-001-0770-9

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Stride A, Shepherd M, Frayling TM, Bulman MP, Ellard S, Hattersley AT. Intrauterine hyperglycaemia is associated with an earlier diagnosis of diabetes in HNF-1α gene mutation carriers. Diabetes Care (2002) 25:2287–91. doi: 10.2337/diacare.25.12.2287

CrossRef Full Text | Google Scholar

14. Stride A, Ellard S, Clark P, Shakespeare L, Salzmann M, Shepherd M, et al. Beta-cell dysfunction, insulin sensitivity, and glycosuria precede diabetes in hepatocyte nuclear factor-1alpha mutation carriers. Diabetes Care (2005) 28:1751–6. doi: 10.2337/diacare.28.7.1751

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Owen KR, Shepherd M, Stride A, Ellard S, Hattersley AT. Heterogeneity in young adult onset aetiology alters clinical characteristics. Diabet Med. (2002) 19:758–61. doi: 10.1046/j.1464-5491.2002.00766.x

PubMed Abstract | CrossRef Full Text | Google Scholar

16. McDonald TJ, McEneny J, Pearson ER, Thanabalasingham G, Szopa M, Shields BM. Lipoprotein composition in HNF1A-MODY: differentiating between HNF1A-MODY and type 2 diabetes. Clin Chim Acta (2012) 413:927–32. doi: 10.1016/j.cca.2012.02.005

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Leighton E, Sainsbury CA, Jones GC. A practical review of C-peptide testing in diabetes. Diabetes Ther. (2017) 8:475–87. doi: 10.1007/s13300-017-0265-4

PubMed Abstract | CrossRef Full Text | Google Scholar

18. McDonald TJ, Ellard S. Maturity onset diabetes of the young: identification and diagnosis. Ann Clin Biochem. (2013) 50(Pt 5):403–15. doi: 10.1177/0004563213483458

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Bingley PJ. Clinical applications of diabetes antibody testing. J Clin Endocrinol Metab. (2010) 95:25–33. doi: 10.1210/jc.2009-1365

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Urbanová J, Rypáčková B, Procházková Z, Kučera P, Cerná M, Anděl M, et al. Positivity for islet cell autoantibodies in patients with monogenic diabetes is associated with later diabetes onset and higher HbA1c level. Diabetic Med. (2014) 31:466–71. doi: 10.1111/dme.12314

CrossRef Full Text

21. 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) 54:2801–10. doi: 10.1007/s00125-011-2261-y

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Bellanne-Chantelot C, Coste J, Ciangura C, Fonfrède M, Saint-Martin C, Bouché C, et al. High-sensitivity C-reactive protein does not improve the differential diagnosis of HNF1A-MODY and familial young-onset type 2 diabetes: a grey zone analysis. Diabetes Metab. (2015) 42:33–7. doi: 10.1016/j.diabet.2015.02.001

CrossRef Full Text | Google Scholar

23. Mughal SA, Thanabalasingham G, Owen RK. Biomarkers currently used for the diagnosis of maturity-onset diabetes of the young. Diabetes Manage. (2013) 3:71–80. doi: 10.2217/dmt.12.82

CrossRef Full Text | Google Scholar

24. Pearson ER, Pruhova S, Tack CJ, Johansen A, Castleden HA, Lumb PJ, et al. Molecular genetics and phenotypic characteristics of MODY caused by hepatocyte nuclear factor 4alpha mutations in a large European collection. Diabetologia (2005) 48:878–85. doi: 10.1007/s00125-005-1738-y

PubMed Abstract | CrossRef Full Text | Google Scholar

25. 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:e118. doi: 10.1371/journal.pmed.0040118

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Thanabalasingham G, Pal A, Selwood MP, Dudley C, Fisher K, Bingley PJ, et al. 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. Diabetes Care (2012) 35:1206–12. doi: 10.2337/dc11-1243

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Amed S, Dean HJ, Panagiotopoulos C, Sellers EA, Hadjiyannakis S, Laubscher TA, et al. Type 2 diabetes, medication-induced diabetes, and monogenic diabetes in Canadian children. Diabetes Care (2010) 33:786–91. doi: 10.2337/dc09-1013

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Stanik J, Dusatkova P, Cinek O, Valentinova L, Huckova M, Skopkova M, et al. De novo mutations of GCK, HNF1A and HNF4A may be more frequent in MODY than previously assumed. Diabetologia (2014) 57:480–4. doi: 10.1007/s00125-013-3119-2

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Bellanné-Chantelot C, Carette C, Riveline JP, Valéro R, Gautier JF, 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) 57:503–8. doi: 10.2337/db07-0859

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Lango Allen H, Johansson S, Ellard S, Shields B, Hertel JK, Raeder H, et al. Polygenic risk variants for type 2 diabetes susceptibility modify age at diagnosis in monogenic HNF1A diabetes. Diabetes (2010) 59:266–71. doi: 10.2337/db09-0555

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Gloyn AL, van de Bunt M, Stratton IM, Lonie L, Tucker L, Ellard S, et al. Prevalence of GCK mutations in individuals screened for fasting hyperglycaemia. Diabetologia (2009) 52:172–4. doi: 10.1007/s00125-008-1188-4

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Kleinberger JW, Pollin TI. Undiagnosed MODY: time for action. Curr Diab Rep. (2015) 15:110. doi: 10.1007/s11892-015-0681-7

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Kawakita R, Hosokawa Y, Fujimaru R, Tamagawa N, Urakami T, Takasawa K, et al. Molecular and clinical characterization of glucokinase maturity-onset diabetes of the young (GCK-MODY) in Japanese patients. Diabet Med. (2014) 31:1357–62. doi: 10.1111/dme.12487

PubMed Abstract | CrossRef Full Text | Google Scholar

34. McDonald TJ, Colclough K, Brown R, Shields B, Shepherd M, Bingley P, et al. Islet autoantibodies can discriminate maturity-onset diabetes of the young (MODY) from Type 1 diabetes. Diabet Med. (2011) 28:1028–33. doi: 10.1111/j.1464-5491.2011.03287.x

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Schober E, Rami B, Grabert M, Thon A, Kapellen T, Reinehr T, et al. Phenotypical aspects of maturity-onset diabetes of the young (MODY diabetes) in comparison with Type 2 diabetes mellitus (T2DM) in children and adolescents: experience from a large multicenter database. Diabetic Med. (2009) 26:466–73. doi: 10.1111/j.1464-5491.2009.02720.x

CrossRef Full Text

36. Pruhová Š, Dušátková P, Neumann D, Hollay E, Cinek O, Lebl J, et al. Two cases of diabetic ketoacidosis in HNF1A-MODY linked to severe dehydration: is it time to change the diagnostic criteria for MODY? Diabetes Care (2013) 36:2573–4. doi: 10.2337/dc13-0058

PubMed Abstract | CrossRef Full Text

37. Kyithar MP, Bacon S, Pannu KK, Rizvi SR, Colclough K, Ellard S, et al. Identification of HNF1A-MODY and HNF4A-MODY in Irish families: phenotypic characteristics and therapeutic implications. Diabetes Metab. (2011) 37:512–9. doi: 10.1016/j.diabet.2011.04.002

CrossRef Full Text

38. Kim SH. Maturity-onset diabetes of the young: what do clinicians need to know? Diabetes Metab J. (2015) 39:468–77. doi: 10.4093/dmj.2015.39.6.468

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Shields BM, Shepherd M, Hudson M, McDonald TJ, Colclough K, Peters J, et al. Population-based assessment of a biomarker-based screening pathway to aid diagnosis of monogenic diabetes in young-onset patients. Diabetes Care (2017) 40:1017–25. doi: 10.2337/dc17-0224

CrossRef Full Text

Keywords: MODY, GCK-MODY, HNF1A-MODY, HNF4A-MODY, diagnostics, differential diagnosis

Citation: Urbanová J, Brunerová L and Brož J (2018) Hidden MODY—Looking for a Needle in a Haystack. Front. Endocrinol. 9:355. doi: 10.3389/fendo.2018.00355

Received: 24 March 2018; Accepted: 13 June 2018;
Published: 02 July 2018.

Edited by:

Gaetano Santulli, Columbia University, United States

Reviewed by:

Gianpaolo De Filippo, Bicêtre Hospital, France

Copyright © 2018 Urbanová, Brunerová and Brož. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Ludmila Brunerová, brunerova@seznam.cz

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.