Horm Metab Res 2009; 41(4): 320-326
DOI: 10.1055/s-0028-1102922
Humans, Clinical

© Georg Thieme Verlag KG Stuttgart · New York

Diagnostic Difficulties in Glucokinase Hyperinsulinism

T. Meissner 1 , J. Marquard 1 , N. Cobo-Vuilleumier 2 , M. Maringa 3 , P. Rodríguez-Bada 2 , M. A. García-Gimeno 4 , E. Baixeras 2 , J. Weber 5 , K. Olek 6 , P. Sanz 4 , E. Mayatepek 1 , A. L. Cuesta-Muñoz 2
  • 1Department of General Pediatrics, University Children's Hospital, Düsseldorf, Germany
  • 2Center for the Study of Pancreatic beta-cell Diseases, IMABIS Foundation Carlos Haya Hospital, Malaga, Spain
  • 3Practice, Human Genetics, Bonn, Germany
  • 4Instituto de Biomedicina de Valencia, CSIC and CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
  • 5Integragen, Laboratory, Bonn, Germany
  • 6Parentage Certificate, Laboratory, Gelsenkirchen, Germany
Further Information

Publication History

received 15.08.2008

accepted 14.10.2008

Publication Date:
03 December 2008 (online)

Abstract

Glucokinase hyperinsulinism is a rare variant of congenital hyperinsulinism caused by activating mutations in the glucokinase gene and has been reported so far to be a result of overactivity of glucokinase within the pancreatic β-cell. Here we report on a new patient with difficulties to diagnose persistent hyperinsulinism and discuss diagnostic procedures of this as well as the other reported individuals. After neonatal hypoglycemia, the patient was reevaluated at the age of 3 years for developmental delay. Morning glucose after overnight fast was 2.5–3.6 mmol/l. Fasting tests revealed supressed insulin secretion at the end of fasting (1.4–14.5 pmol/l). In addition, diagnostic data of the patients reported so far were reviewed. A novel heterozygous missense mutation in exon 10 c.1354G>C (p.Val452Leu) was found and functional studies confirmed the activating mutation. There was no single consistent diagnostic criterion found for our patient and glucokinase hyperinsulinism individuals in general. Often at the time of hypoglycemia low insulin levels were found. Therefore insulin concentrations at hypoglycemia, or during fasting test as well as reactive hypoglycemia after an oral glucose tolerance test were not conclusive for all patients. A glucose lowering effect in extra-pancreatic tissues independent from hyperinsulinism that results in diagnostic difficulties may contribute to underestimation of glucokinase hyperinsulinism. Mutational analysis of the GCK-gene should be performed in all individuals with unclear episodes of hypoglycemia even without documented hyperinsulinism during hypogly-cemia. Delay of diagnosis might result in mental handicap of the affected individuals.

References

  • 1 Meissner T, Beinbrech B, Mayatepek E. Congenital hyperinsulinism: molecular basis of a heterogeneous disease.  Hum Mutat. 1999;  13 351-361
  • 2 Lindley KJ, Dunne MJ. Contemporary strategies in the diagnosis and management of neonatal hyperinsulinaemic hypoglycaemia.  Early Hum Dev. 2005;  81 61-72
  • 3 Aynsley-Green A, Hussain K, Hall J, Saudubray JM, Nihoul-Fekete C, Lonlay-Debeney P De, Brunelle F, Otonkoski T, Thornton P, Lindley KJ. Practical management of hyperinsulinism of infancy.  Arch Dis Child Fetal Neonatal. 2000;  82 98-107
  • 4 Thomas PM, Cote GJ, Wohllk N, Haddad B, Mathew PM, Rabl W, Aguilar-Bryan L, Gagel RF, Bryan J. Mutations in the sulfonylurea receptor gene in familial hyperinsulinemic hypoglycemia of infancy.  Science. 1995;  268 426-429
  • 5 Nestorowicz A, Inagaki N, Gonoi T, Schoor KP, Wilson BA, Glaser B, Landau H, Stanley CA, Thornton PS, Seino S, Permutt MA. A nonsense mutation in the inward rectifier potassium channel gene, Kir6.2, is associated with familial hyperinsulinism.  Diabetes. 1997;  46 1743-1748
  • 6 Stanley CA, Lieu YK, Hsu BY, Ming JE, Glaser B, Poncz M. Hyperin-sulinism and hyperamonemia in infants with regulatory mutations of the glutamate dehydrogenase gene.  N Engl J Med. 1998;  338 1352-1357
  • 7 Clayton PT, Eaton S, Aynsley-Green A, Edginton M, Hussain K, Krywawych S, Datta V, Malingré HEM, Berger R, Berg IET van den. Hyperinsulinism in short-chain l-3-hydroxyacyl-CoA dehydrogenase deficiency reveals the importance of β-oxidation in insulin secretion.  J Clin Invest. 2001;  108 457-465
  • 8 Glaser B, Kesavan P, Heyman M, Davis E, Cuesta A, Buchs A, Stanley CA, Thornton PS, Permutt MA, Matschinsky FM, Herold HC. Familial hyperinsulinism caused by an activating glucokinase mutation.  N Engl J Med. 1998;  338 226-230
  • 9 Christesen HB, Jacobsen BB, Odili S, Buettger C, Cuesta-Munoz A, Hansen T, Brusgaard K, Massa O, Magnuson MA, Shiota C, Matschinsky FM, Barbetti F. The second activating glucokinase mutation (A456 V): implications for glucose homeostasis and diabetes therapy.  Diabetes. 2002;  51 1240-1246
  • 10 Gloyn AL, Noordam K, Willemsen MA, Ellard S, Lam WW, Campbell IW, Midgley P, Shiota C, Buettger C, Magnuson MA, Matschinsky FM, Hattersley AT. Insights into the biochemical and genetic basis of glucokinase activation from naturally occurring hypoglycemia mutations.  Diabetes. 2003;  52 2433-2434
  • 11 Cuesta-Munoz AL, Huopio H, Otonkoski T, Gomez-Zumaquero JM, Nanto-Salonen K, Rahier J, Lopez-Enriquez S, Garcia-Gimeno MA, Sanz P, Soriguer FC, Laakso M. Severe persistent hyperinsulinemic hypoglycemia due to a de novo glucokinase mutation.  Diabetes. 2004;  53 2164-2168
  • 12 Dullaart RP, Hoogenberg K, Rouwe CW, Stulp BK. Family with autosomal dominant hyperinsulinism associated with A456V mutation in the glucokinase gene.  J Intern Med. 2004;  255 143-145
  • 13 Wabitsch M, Lahr G, Bunt M Van de, Marchant C, Lindner M, Puttkamer J von, Fenneberg A, Debatin KM, Klein R, Ellard S, Clark A, Gloyn AL. Heterogeneity in disease severity in a family with a novel G68V GCK activating mutation causing persistent hyperinsulinaemic hypoglycaemia of infancy.  Diabet Med. 2007;  24 1393-1399
  • 14 Christesen HB, Tribble ND, Molven A, Siddiqui J, Sandal T, Brusgaard K, Ellard S, Njølstad PR, Alm J, Brock Jacobsen B, Hussain K, Gloyn AL. Activating glucokinase (GCK) mutations as a cause of medically responsive congenital hyperinsulinism: prevalence in children and characterisation of a novel GCK mutation.  Eur J Endocrinol. 2008;  159 27-34
  • 15 Duvanel CB, Fawer CL, Cotting J, Hohlfeld P, Matthieu JM. Long-term effects of neonatal hypoglycemia on brain growth and psychomotor development in small-for-gestational-age preterm infants.  J Pediatr. 1999;  134 492-498
  • 16 Matschinsky FM. Regulation of pancreatic β-cellglucokinase: from basics to therapeutics.  Diabetes. 2002;  51 394-404
  • 17 Davis EA, Cuesta-Munoz A, Raoul M, Buettger C, Sweet I, Moates M, Magnuson MA, Matschinsky FM. Mutants of glucokinase cause hypoglycaemia- and hyperglycaemia syndromes and their analysis illuminates fundamental quantitative concepts of glucose homeostasis.  Diabetologia. 1999;  42 1175-1186
  • 18 Froguel P, Zouali H, Vionnet N, Velho G, Vdaire M, Sun F, Lesage S, Stoffel M, Takeda J, Passa P, Permutt A, Beckmann JS, Bell GI, Cohen D. Familial hyperglycemia due to mutations in glucokinase.  N Engl J Med. 1993;  328 697-702
  • 19 Njolstad PR, Sovic O, Cuesta-Munoz A, Bjorkhaug L, Massa O, Barbetti F, Undlien DE, Shiota C, Magnuson MA, Molven A, Matschinsky FM, Bell GI. Neonatal diabetes mellitus due to complete glucokinase deficiency.  N Engl J Med. 2001;  344 1588-1592
  • 20 Matschinsky FM, Magnuson MA, Zelent D, Jetton TL, Doliba N, Han Y, Taub R, Grimsby J. The network of glucokinase-expressing cells in glucose homeostasis and the potential of glucokinase activators for diabetes therapy.  Diabetes. 2006;  55 1-12

Correspondence

T. Meissner

Department of General Pediatrics

University Children's Hospital

Moorenstr. 5

40225 Düsseldorf

Germany

Phone: +49/211/811 76 38

Fax: +49/211/811 95 12

Email: thomas.meissner@med.uni-duesseldorf.de

A. L. Cuesta-MuñozMD, PhD 

IMABIS Foundation and Center

for the Study of Pancreatic

Beta-cell Diseases Carlos

Haya University Hospital

Avda. Carlos Haya no 82

29010 Málaga

Spain

Phone: 34/95/129 14 46

Fax: 34/95/261 40 12

Email: alcm@fundacionimabis.org

    >