Skip to main content
Top
Published in: Diabetologia 6/2006

01-06-2006 | Article

A new model of insulin-deficient diabetes: male NOD mice with a single copy of Ins1 and no Ins2

Authors: N. Babaya, M. Nakayama, H. Moriyama, R. Gianani, T. Still, D. Miao, L. Yu, J. C. Hutton, G. S. Eisenbarth

Published in: Diabetologia | Issue 6/2006

Login to get access

Abstract

Aims/hypothesis

We describe a novel model of insulin-deficient diabetes with a single copy of the gene encoding insulin 1 (Ins1) and no gene encoding insulin 2 (Ins2).

Materials and methods

We constructed five lines of mice: mice with two copies of Ins1 (NOD Ins1+/+,Ins2−/−), mice with a single copy of Ins1 (NOD Ins1+/−,Ins2−/−), mice with two copies of Ins2 (NOD Ins1−/−,Ins2+/+), mice with a single copy of Ins2 (NOD Ins1−/−,Ins2+/−) and NOD Ins1+/−,Ins2−/− mice with a transgene encoding B16:Ala proinsulin.

Results

By 10 weeks of age, all male NOD Ins1+/−,Ins2−/− mice were diabetic, whereas all female NOD Ins1+/−,Ins2−/− were not diabetic (p<0.0001). In contrast, neither male nor female NOD Ins1−/−,Ins2+/− with a single copy of Ins2 (rather than single copy of Ins1) developed early diabetes and no mice with two copies of either gene developed early diabetes. Islets of the diabetic male NOD Ins1+/−,Ins2−/− at this early age had no lymphocyte infiltration. Instead there was heterogeneous (between islet cells) weak staining for insulin. Although only male NOD Ins1+/−,Ins2−/− mice developed diabetes, both male and female NOD Ins1+/−,Ins2−/− mice had markedly decreased insulin content. In NOD Ins1+/+,Ins2−/−, there was also a significant decrease in insulin content, whereas NOD Ins1−/−,Ins2+/+ mice, and even NOD Ins1−/−,Ins2+/− mice, were normal. Male NOD Ins1+/−,Ins2−/− mice were completely rescued from diabetes by introduction of a transgene encoding proinsulin. On i.p. insulin tolerance testing, male mice had insulin resistance compared with female mice.

Conclusions/interpretation

These results suggest that Ins1 is a ‘defective gene’ relative to Ins2, and that the mouse lines created provide a novel model of sex-dimorphic insulin-deficient diabetes.
Literature
1.
go back to reference Soares MB, Schon E, Henderson A et al (1985) RNA-mediated gene duplication: the rat preproinsulin I gene is a functional retroposon. Mol Cell Biol 5:2090–2103PubMed Soares MB, Schon E, Henderson A et al (1985) RNA-mediated gene duplication: the rat preproinsulin I gene is a functional retroposon. Mol Cell Biol 5:2090–2103PubMed
2.
go back to reference Moriyama H, Abiru N, Paronen J et al (2003) Evidence for a primary islet autoantigen (preproinsulin 1) for insulitis and diabetes in the nonobese diabetic mouse. Proc Natl Acad Sci USA 100:10376–10381PubMedCrossRef Moriyama H, Abiru N, Paronen J et al (2003) Evidence for a primary islet autoantigen (preproinsulin 1) for insulitis and diabetes in the nonobese diabetic mouse. Proc Natl Acad Sci USA 100:10376–10381PubMedCrossRef
3.
go back to reference Thebault-Baumont K, Dubois-Laforgue D, Krief P et al (2003) Acceleration of type 1 diabetes mellitus in proinsulin 2-deficient NOD mice. J Clin Invest 111:851–857PubMed Thebault-Baumont K, Dubois-Laforgue D, Krief P et al (2003) Acceleration of type 1 diabetes mellitus in proinsulin 2-deficient NOD mice. J Clin Invest 111:851–857PubMed
4.
go back to reference Nakayama M, Abiru N, Moriyama H et al (2005) Prime role for an insulin epitope in the development of type 1 diabetes in NOD mice. Nature 435:220–223PubMedCrossRef Nakayama M, Abiru N, Moriyama H et al (2005) Prime role for an insulin epitope in the development of type 1 diabetes in NOD mice. Nature 435:220–223PubMedCrossRef
5.
go back to reference Nakayama M, Moriyama H, Abiru N et al (2004) Establishment of native insulin-negative NOD mice and the methodology to distinguish specific insulin knockout genotypes and a B:16 alanine preproinsulin transgene. Ann N Y Acad Sci 1037:193–198PubMedCrossRef Nakayama M, Moriyama H, Abiru N et al (2004) Establishment of native insulin-negative NOD mice and the methodology to distinguish specific insulin knockout genotypes and a B:16 alanine preproinsulin transgene. Ann N Y Acad Sci 1037:193–198PubMedCrossRef
6.
go back to reference Yu L, Robles DT, Abiru N et al (2000) Early expression of antiinsulin autoantibodies of humans and the NOD mouse: evidence for early determination of subsequent diabetes. Proc Natl Acad Sci USA 97:1701–1706PubMedCrossRef Yu L, Robles DT, Abiru N et al (2000) Early expression of antiinsulin autoantibodies of humans and the NOD mouse: evidence for early determination of subsequent diabetes. Proc Natl Acad Sci USA 97:1701–1706PubMedCrossRef
7.
go back to reference Leroux L, Desbois P, Lamotte L et al (2001) Compensatory responses in mice carrying a null mutation for Ins1 or Ins2. Diabetes 50(Suppl 1):S150–S153PubMedCrossRef Leroux L, Desbois P, Lamotte L et al (2001) Compensatory responses in mice carrying a null mutation for Ins1 or Ins2. Diabetes 50(Suppl 1):S150–S153PubMedCrossRef
8.
go back to reference Chentoufi AA, Polychronakos C (2002) Insulin expression levels in the thymus modulate insulin-specific autoreactive T-cell tolerance: the mechanism by which the IDDM2 locus may predispose to diabetes. Diabetes 51:1383–1390PubMedCrossRef Chentoufi AA, Polychronakos C (2002) Insulin expression levels in the thymus modulate insulin-specific autoreactive T-cell tolerance: the mechanism by which the IDDM2 locus may predispose to diabetes. Diabetes 51:1383–1390PubMedCrossRef
9.
go back to reference Ueda H, Ikegami H, Yamato E et al (1995) The NSY mouse: a new animal model of spontaneous NIDDM with moderate obesity. Diabetologia 38:503–508PubMedCrossRef Ueda H, Ikegami H, Yamato E et al (1995) The NSY mouse: a new animal model of spontaneous NIDDM with moderate obesity. Diabetologia 38:503–508PubMedCrossRef
10.
11.
go back to reference Ikegami H, Fujisawa T, Ogihara T (2004) Mouse models of type 1 and type 2 diabetes derived from the same closed colony: genetic susceptibility shared between two types of diabetes. ILAR J 45:268–277PubMed Ikegami H, Fujisawa T, Ogihara T (2004) Mouse models of type 1 and type 2 diabetes derived from the same closed colony: genetic susceptibility shared between two types of diabetes. ILAR J 45:268–277PubMed
12.
go back to reference Babaya N, Ikegami H, Fujisawa T et al (2005) Susceptibility to streptozotocin-induced diabetes is mapped to mouse chromosome 11. Biochem Biophys Res Commun 328:158–164PubMedCrossRef Babaya N, Ikegami H, Fujisawa T et al (2005) Susceptibility to streptozotocin-induced diabetes is mapped to mouse chromosome 11. Biochem Biophys Res Commun 328:158–164PubMedCrossRef
13.
go back to reference Kulkarni RN, Almind K, Goren HJ et al (2003) Impact of genetic background on development of hyperinsulinemia and diabetes in insulin receptor/insulin receptor substrate-1 double heterozygous mice. Diabetes 52:1528–1534PubMedCrossRef Kulkarni RN, Almind K, Goren HJ et al (2003) Impact of genetic background on development of hyperinsulinemia and diabetes in insulin receptor/insulin receptor substrate-1 double heterozygous mice. Diabetes 52:1528–1534PubMedCrossRef
14.
go back to reference Pugliese A, Zeller M, Fernandez A Jr et al (1997) The insulin gene is transcribed in the human thymus and transcription levels correlated with allelic variation at the INS VNTR-IDDM2 susceptibility locus for type 1 diabetes. Nat Genet 15:293–297PubMedCrossRef Pugliese A, Zeller M, Fernandez A Jr et al (1997) The insulin gene is transcribed in the human thymus and transcription levels correlated with allelic variation at the INS VNTR-IDDM2 susceptibility locus for type 1 diabetes. Nat Genet 15:293–297PubMedCrossRef
15.
go back to reference Vafiadis P, Bennett ST, Todd JA et al (1997) Insulin expression in human thymus is modulated by INS VNTR alleles at the IDDM2 locus. Nat Genet 15:289–292PubMedCrossRef Vafiadis P, Bennett ST, Todd JA et al (1997) Insulin expression in human thymus is modulated by INS VNTR alleles at the IDDM2 locus. Nat Genet 15:289–292PubMedCrossRef
16.
go back to reference Chen W, Bergerot I, Elliott JF et al (2001) Evidence that a peptide spanning the B–C junction of proinsulin is an early autoantigen epitope in the pathogenesis of type 1 diabetes. J Immunol 167:4926–4935PubMed Chen W, Bergerot I, Elliott JF et al (2001) Evidence that a peptide spanning the B–C junction of proinsulin is an early autoantigen epitope in the pathogenesis of type 1 diabetes. J Immunol 167:4926–4935PubMed
17.
go back to reference Brimnes MK, Jensen T, Jorgensen TN, Michelsen BK, Troelsen J, Werdelin O (2002) Low expression of insulin in the thymus of non-obese diabetic mice. J Autoimmun 19:203–213PubMedCrossRef Brimnes MK, Jensen T, Jorgensen TN, Michelsen BK, Troelsen J, Werdelin O (2002) Low expression of insulin in the thymus of non-obese diabetic mice. J Autoimmun 19:203–213PubMedCrossRef
Metadata
Title
A new model of insulin-deficient diabetes: male NOD mice with a single copy of Ins1 and no Ins2
Authors
N. Babaya
M. Nakayama
H. Moriyama
R. Gianani
T. Still
D. Miao
L. Yu
J. C. Hutton
G. S. Eisenbarth
Publication date
01-06-2006
Publisher
Springer-Verlag
Published in
Diabetologia / Issue 6/2006
Print ISSN: 0012-186X
Electronic ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-006-0241-4

Other articles of this Issue 6/2006

Diabetologia 6/2006 Go to the issue