About   Help   FAQ
Phenotypes associated with this allele
Allele Symbol
Allele Name
Allele ID
Insrtm1Khn
targeted mutation 1, Ronald Kahn
MGI:2384332
Summary 19 genotypes
Jump to Allelic Composition Genetic Background Genotype ID
cn1
Insrtm1Khn/Insrtm1Khn
Leprtm1.1Chua/Leprtm1.1Chua
Tg(Pomc1-cre)16Lowl/0
involves: 129 * 129S4/SvJae * 129S6/SvEvTac * C57BL/6J * FVB/N MGI:5471586
cn2
Insrtm1Khn/Insrtm1Khn
Speer6-ps1Tg(Alb-cre)21Mgn/Speer6-ps1+
involves: 129S4/SvJae * C57BL/6 * C57BL/6J MGI:6791351
cn3
Insrtm1Khn/Insrtm1Khn
Tg(Ins2-cre)25Mgn/0
involves: 129S4/SvJae * C57BL/6 * DBA/2 MGI:3775383
cn4
Insrtm1Khn/Insrtm1Khn
Pdx1tm1Ted/Pdx1+
Speer6-ps1Tg(Alb-cre)21Mgn/Speer6-ps1+
involves: 129S4/SvJae * C57BL/6 * DBA/2 MGI:3583116
cn5
Insrtm1Khn/Insrtm1Khn
Speer6-ps1Tg(Alb-cre)21Mgn/Speer6-ps1+
involves: 129S4/SvJae * C57BL/6 * DBA/2 MGI:3775385
cn6
Insrtm1Khn/Insrtm1Khn
Speer6-ps1Tg(Alb-cre)21Mgn/Speer6-ps1+
Tg(Ins2-cre)25Mgn/0
involves: 129S4/SvJae * C57BL/6 * DBA/2 MGI:3775382
cn7
Insrtm1Khn/Insrtm1Khn
Tg(Ckmm-cre)5Khn/0
involves: 129S4/SvJae * C57BL/6 * FVB MGI:3775309
cn8
Insrtm1Khn/Insrtm1Khn
Tg(Pdgfrb-cre)#Rha/0
involves: 129S4/SvJae * C57BL/6J MGI:7327617
cn9
Insrtm1Khn/Insrtm1Khn
Speer6-ps1Tg(Alb-cre)21Mgn/Speer6-ps1+
involves: 129S4/SvJae * C57BL/6J * DBA/2 * FVB MGI:3039262
cn10
Insrtm1Khn/Insrtm1Khn
Tg(Ckmm-cre)5Khn/0
involves: 129S4/SvJae * C57BL/6J * FVB MGI:2389585
cn11
Insrtm1Khn/Insrtm1Dac
Tg(Ckmm-cre)5Khn/0
involves: 129S4/SvJae * C57BL/6J * FVB MGI:2389586
cn12
Insrtm1Khn/Insrtm1Khn
Tg(Nes-cre)1Kln/0
involves: 129S4/SvJae * C57BL/6J * SJL MGI:3583775
cn13
Insrtm1Khn/Insrtm1Khn
Tg(Nes-cre)1Kln/0
involves: 129S4/SvJae * C57BL/6 * SJL MGI:3629045
cn14
Insrtm1Khn/Insrtm1Khn
Tg(Myhca-cre)1Abel/0
involves: 129S4/SvJae * FVB MGI:3620045
cn15
Insrtm1Khn/Insrtm1Khn
Tg(Slc2a4-cre)546Dac/0
involves: 129S4/SvJaeSor * C57BL/6 * FVB MGI:4941586
cn16
Igf1rtm1.1Mhz/Igf1r+
Insrtm1Khn/Insrtm1Khn
Tg(Ckmm-cre)5Khn/0
involves: 129/Sv * 129S4/SvJae * C57BL/6 * FVB MGI:3775310
cn17
Igf1rtm1.1Mhz/Igf1rtm1.1Mhz
Insrtm1Khn/Insrtm1Khn
Tg(Ckmm-cre)5Khn/0
involves: 129/Sv * 129S4/SvJae * C57BL/6 * FVB MGI:3775311
cn18
Igf1rtm1.1Mhz/Igf1rtm1.1Mhz
Insrtm1Khn/Insr+
Tg(Ckmm-cre)5Khn/0
involves: 129/Sv * 129S4/SvJae * C57BL/6 * FVB MGI:3775313
cx19
Igf1rtm1.1Mhz/Igf1rtm1.1Mhz
Insrtm1Khn/Insrtm1Khn
involves: 129/Sv * 129S4/SvJae * C57BL/6 * FVB MGI:3775314


Genotype
MGI:5471586
cn1
Allelic
Composition
Insrtm1Khn/Insrtm1Khn
Leprtm1.1Chua/Leprtm1.1Chua
Tg(Pomc1-cre)16Lowl/0
Genetic
Background
involves: 129 * 129S4/SvJae * 129S6/SvEvTac * C57BL/6J * FVB/N
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Insrtm1Khn mutation (1 available); any Insr mutation (94 available)
Leprtm1.1Chua mutation (1 available); any Lepr mutation (121 available)
Tg(Pomc1-cre)16Lowl mutation (1 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
adipose tissue
• trend towards increased intra-abdominal adiposity at 4 months of age
• perigonadal adipocyte hypertrophy at 4 months of age
• perigonadal adipocyte hypertrophy at 4 months of age
• an increase in macrophage accumulation is seen in the perigonadal fat indicating inflammation
• percent body fat is increased in females at 4 months of age, however absolute body weight is not increased at this time
• an increase in macrophage accumulation is seen in the perigonadal fat, and macrophage marker analysis and cytokine expression indicates low-grade inflammation in adipose tissue

endocrine/exocrine glands
• 45% of mice lack either corpora lutea or preovulatory follicles
• 45% of mice lack either corpora lutea or preovulatory follicles
• low-grade inflammation as indicated by increased IL-6 expression in the ovary

growth/size/body
• percent lean body mass is decreased at 4 months of age

homeostasis/metabolism
• females are hyperglycemic before (at 3 months of age) and during pregnancy (at gestational days 12 and 15), without evidence of gestational glucose intolerance
• circulating levels of adipokine leptin are increased at 4 months of age

immune system
• an increase in macrophage accumulation is seen in the perigonadal fat, and macrophage marker analysis and cytokine expression indicates low-grade inflammation in adipose tissue
• low-grade inflammation as indicated by increased IL-6 expression in the ovary
• low-grade inflammation as indicated by increased IL-1beta expression in the liver

liver/biliary system
• low-grade inflammation as indicated by increased IL-1beta expression in the liver

reproductive system
• 45% of mice lack either corpora lutea or preovulatory follicles
• 45% of mice lack either corpora lutea or preovulatory follicles
• low-grade inflammation as indicated by increased IL-6 expression in the ovary
• females exhibit reduced ovulation but show normal cycling suppression from fasting

Mouse Models of Human Disease
DO ID OMIM ID(s) Ref(s)
polycystic ovary syndrome DOID:11612 OMIM:184700
J:192274




Genotype
MGI:6791351
cn2
Allelic
Composition
Insrtm1Khn/Insrtm1Khn
Speer6-ps1Tg(Alb-cre)21Mgn/Speer6-ps1+
Genetic
Background
involves: 129S4/SvJae * C57BL/6 * C57BL/6J
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Insrtm1Khn mutation (1 available); any Insr mutation (94 available)
Speer6-ps1Tg(Alb-cre)21Mgn mutation (6 available); any Speer6-ps1 mutation (4 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
homeostasis/metabolism
• at 2 months of age, mice show hyperglycemia in the fed state
• however, blood glucose levels become normal by 6 months of age
• at 2 months of age, mice show hyperinsulinemia in the fed state that is more severe than in Mad2l1bptm1.1Itl/Mad2l1bptm1.1Itl Speer6-ps1Tg(Alb-cre)21Mgn/Speer6-ps1+ mice
• at 2 months of age, mice show severe glucose intolerance
• surprisingly, glucose intolerance is not severe at 6 months of age
• at 2 months of age, liver glycogen levels are decreased to a greater extent than in Mad2l1bptm1.1Itl/Mad2l1bptm1.1Itl Speer6-ps1Tg(Alb-cre)21Mgn/Speer6-ps1+ mice
• at 2 months of age, mice show severe insulin intolerance
• surprisingly, insulin intolerance is not severe at 6 months of age
• insulin treatment fails to stimulate glycogen synthase activity in hepatocytes, unlike in wild-type hepatocytes

liver/biliary system
• at 2 months of age, males show scattered focal dysplasia in the periportal area of the liver
• however, overall liver architecture is normal
• at 2 months of age, liver glycogen levels are decreased to a greater extent than in Mad2l1bptm1.1Itl/Mad2l1bptm1.1Itl Speer6-ps1Tg(Alb-cre)21Mgn/Speer6-ps1+ mice
• insulin receptor (IR) autophosphorylation and activating phosphorylation of AKT (pT308) are significantly reduced whereas inhibitory phosphorylation of GSK3 (pS9) is modestly reduced in whole-liver lysates from insulin-treated mice
• insulin-dependent glycogen synthase (GS) activation is abolished in hepatocytes




Genotype
MGI:3775383
cn3
Allelic
Composition
Insrtm1Khn/Insrtm1Khn
Tg(Ins2-cre)25Mgn/0
Genetic
Background
involves: 129S4/SvJae * C57BL/6 * DBA/2
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Insrtm1Khn mutation (1 available); any Insr mutation (94 available)
Tg(Ins2-cre)25Mgn mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
mortality/aging
• when fed a high fat diet, 30% of mice die by 16 weeks
• some mice die due to severe hyperglycemia

homeostasis/metabolism
• mice exhibit an increase in serum glucose level on a regular diet and a severe increase when fed a high fat diet
• mice exhibit impaired glucose tolerance on a normal diet and severe intolerance when fed a high fat diet
• however, mice are not insulin resistant

endocrine/exocrine glands
• mice exhibit decreased beta cell mass when fed a normal or high fat diet




Genotype
MGI:3583116
cn4
Allelic
Composition
Insrtm1Khn/Insrtm1Khn
Pdx1tm1Ted/Pdx1+
Speer6-ps1Tg(Alb-cre)21Mgn/Speer6-ps1+
Genetic
Background
involves: 129S4/SvJae * C57BL/6 * DBA/2
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Insrtm1Khn mutation (1 available); any Insr mutation (94 available)
Pdx1tm1Ted mutation (0 available); any Pdx1 mutation (35 available)
Speer6-ps1Tg(Alb-cre)21Mgn mutation (6 available); any Speer6-ps1 mutation (4 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
endocrine/exocrine glands
• these mutants fail to exhibit an adaptive islet hyperplastic response to insulin resistance; instead, pancreatic beta cell mass is reduced, and islets appear small with scattered non-beta cells




Genotype
MGI:3775385
cn5
Allelic
Composition
Insrtm1Khn/Insrtm1Khn
Speer6-ps1Tg(Alb-cre)21Mgn/Speer6-ps1+
Genetic
Background
involves: 129S4/SvJae * C57BL/6 * DBA/2
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Insrtm1Khn mutation (1 available); any Insr mutation (94 available)
Speer6-ps1Tg(Alb-cre)21Mgn mutation (6 available); any Speer6-ps1 mutation (4 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
homeostasis/metabolism
• at 10 to 12 months of age
• while mice display normoglycemia during the first 3 weeks of life, at 4 weeks mice develop hypoglycemia that lasts until week 6

endocrine/exocrine glands
• at 2 weeks beta cell mass is increased 2-fold and at 6 weeks 4-fold compared to in wild-type mice due to increased mitosis of beta cells
• at 10 to 12 months of age, mice exhibit a 27-fold increase in beta cell mass due to an 8-fold increase in proliferating beta cells with a slight increase in apoptosis




Genotype
MGI:3775382
cn6
Allelic
Composition
Insrtm1Khn/Insrtm1Khn
Speer6-ps1Tg(Alb-cre)21Mgn/Speer6-ps1+
Tg(Ins2-cre)25Mgn/0
Genetic
Background
involves: 129S4/SvJae * C57BL/6 * DBA/2
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Insrtm1Khn mutation (1 available); any Insr mutation (94 available)
Speer6-ps1Tg(Alb-cre)21Mgn mutation (6 available); any Speer6-ps1 mutation (4 available)
Tg(Ins2-cre)25Mgn mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
mortality/aging
• 82% of mice die by 6 weeks of age with some mice surviving a few weeks longer

homeostasis/metabolism
• mice develop hypoglycemia at 2 weeks of age that rapidly worsens by 6 weeks of age
• mice exhibit high fasting glucose serum levels

endocrine/exocrine glands

renal/urinary system
• during terminal stages

behavior/neurological
• during terminal stages

digestive/alimentary system
N
• unlike mice null for Insr in beta cells mice, islet cell mass is not increased despite similar levels of elevated insulin levels

growth/size/body
• mice develop severe diabetes and loss 30% of their body weight in terminal stages




Genotype
MGI:3775309
cn7
Allelic
Composition
Insrtm1Khn/Insrtm1Khn
Tg(Ckmm-cre)5Khn/0
Genetic
Background
involves: 129S4/SvJae * C57BL/6 * FVB
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Insrtm1Khn mutation (1 available); any Insr mutation (94 available)
Tg(Ckmm-cre)5Khn mutation (4 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
cardiovascular system
N
• heart function and morphology is normal

growth/size/body
N
• mice exhibit normal growth rates

homeostasis/metabolism
N
• mice exhibit normal glucose homeostasis




Genotype
MGI:7327617
cn8
Allelic
Composition
Insrtm1Khn/Insrtm1Khn
Tg(Pdgfrb-cre)#Rha/0
Genetic
Background
involves: 129S4/SvJae * C57BL/6J
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Insrtm1Khn mutation (1 available); any Insr mutation (94 available)
Tg(Pdgfrb-cre)#Rha mutation (0 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
vision/eye
• mice show increased vascularity in the regions adjacent to retinal veins
• the number of sprouting tip cells is increased indicating increased angiogenic sporouting in superficial and deep layers
• however, pericyte coverage of capillaries is unaltered in perivenous and periartieral plexi and no differences in vascular regression or endothelial proliferation is seen
• mice show increased retinal vein diameter at P5 and P10
• mice show impaired venous pattering during developmental retinal angiogenesis with increased branching complexity in the peripheral half of the plexus at P5

cardiovascular system
• mice show increased vascularity in the regions adjacent to retinal veins
• the number of sprouting tip cells is increased indicating increased angiogenic sporouting in superficial and deep layers
• however, pericyte coverage of capillaries is unaltered in perivenous and periartieral plexi and no differences in vascular regression or endothelial proliferation is seen
• mice show increased retinal vein diameter at P5 and P10
• mice show impaired venous pattering during developmental retinal angiogenesis with increased branching complexity in the peripheral half of the plexus at P5
• mice exhibit abnormal retinal venous plexus development in early postnatal life, with increased branching complexity in the peripheral half of the plexus, increased venous plexus vascular density but not arterial plexus density, and increased vascular area of the deep vascular plexus indicating venous plexus hypervascularity
• perivenous hypervascularity diminishes with time, although it is still evident at P10

homeostasis/metabolism
• fasting serum insulin is increased in 8-week-old mice
• however, fasting serum glucose is normal

Mouse Models of Human Disease
DO ID OMIM ID(s) Ref(s)
diabetic retinopathy DOID:8947 J:327000




Genotype
MGI:3039262
cn9
Allelic
Composition
Insrtm1Khn/Insrtm1Khn
Speer6-ps1Tg(Alb-cre)21Mgn/Speer6-ps1+
Genetic
Background
involves: 129S4/SvJae * C57BL/6J * DBA/2 * FVB
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Insrtm1Khn mutation (1 available); any Insr mutation (94 available)
Speer6-ps1Tg(Alb-cre)21Mgn mutation (6 available); any Speer6-ps1 mutation (4 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
homeostasis/metabolism
• at 6 months, mutant mice show a 50% increase in alkaline phosphatase levels, suggesting biliary tract dysfunction
• at 6 months, mutant mice show a 2.6-fold increase in aspartate aminotransferase levels, suggesting hepatocellular damage
• in contrast, no significant changes in levels of lactate dehydrogenase or alanine aminotransferase are observed
• at 6 months, mutant mice display a 50% reduction in serum albumin levels
• at 2 months, male mutants fail to suppress hepatic glucose output in response to insulin administration, despite intact insulin signaling in adipose tissue and nearly normal insulin signaling in muscle
• at 6 months, female mutants show a have 5- to 6-fold increase in pancreatic insulin content measured in acid-ethanol extracts
• at 2 months, male mutants exhibit significantly elevated blood glucose in the fed state, despite a 20-fold increase in insulin levels relative to control mice; in contrast, glucagon levels remain unaffected
• at 2 months, male mutants also display a mild hyperglycemia in the fasted state, despite a 7-fold increase in serum insulin levels
• unexpectedly, as mutant mice age, the elevated fasting blood glucose levels progressively decline; by 6 months of age, mutants exhibit fasting hypoglycemia rather than hyperglycemia
• at 2 months, male mutants exhibit an extreme increase in serum insulin levels (20-fold in fed state; 7-fold in fasted state)
• at 2 months, fasted male mutants display pronounced glucose intolerance
• surprisingly, glucose intolerance is no longer apparent at 6 months
• in addition to increased insulin secretion, 11-week-old male mutants show a 75% reduction in the C-peptide:insulin ratio, indicating decreased insulin clearance
• at 2 months, random-fed male mutants are severely resistant to the blood glucose-lowering effects of exogenously administered insulin
• at 2 months, male mutants show a 40-50% reduction in the levels of serum free fatty acids
• at 2 months, male mutants show a 40-50% reduction in the levels of serum triglycerides
• mutant gallbladders show a notable increase in bile volume

liver/biliary system
• at 6- to 12-months of age, mutant gall bladders appear enlarged
• at 2 months, mutant livers appear a little darker in color, with areas of focal dysplasia and reduced glycogen storage in the parenchyma; no steatosis is observed
• at 6- to 12-months of age, mutant livers display multiple hyperplastic pale nodules scattered throughout all lobes
• by 12 months, the normal lobular structure is disrupted; hyperplastic nodules consist of highly vacuolated cells that compress the adjacent normal tissue, in the absence of fibrosis
• at 6 months, mutant hepatocytes exhibit a moderate increase in lipid accumulation; only a few electron-dense glycogen granules are observed
• at 2 months, mutant livers are 40% smaller than those of age- and gender-matched controls; liver size decreases significantly with increasing age
• insulin resistance in liver results in elevated expression of PEPCK, the rate-limiting enzyme for gluconeogenesis, and reduced expression of glucokinase and liver pyruvate kinase, two enzymes critical for glucose utilization
• the mutant liver appears to be chronically geared toward glucose production; early insulin signaling events, such as IRS-1 and IRS-2 phosphorylation, are lost

endocrine/exocrine glands
• at 6 months, increased pancreatic insulin content is associated with a 6-fold increase in pancreatic beta cell mass (J:63878)
• at 6 months, mutant mice show a ~4-fold increase in pancreatic beta cell mass, as a compensatory response to insulin resistance (J:92861)
• at 6 months, mutant mice show an adaptive islet hyperplastic response to insulin resistance
• at 6- to 12-months of age, mutant gall bladders appear enlarged
• at 6 months, female mutants show a have 5- to 6-fold increase in pancreatic insulin content measured in acid-ethanol extracts

cellular
• at 6 months, mutant hepatocytes exhibit giant mitochondria

growth/size/body
• at 6- to 12-months of age, mutant gall bladders appear enlarged
• newborn mutant mice appear normal, nurse successfully, and survive well upon weaning but display a mild growth deficit postweaning
• this moderate growth deficit appears to result from defects in the growth hormone-IGF axis (unpublished data)




Genotype
MGI:2389585
cn10
Allelic
Composition
Insrtm1Khn/Insrtm1Khn
Tg(Ckmm-cre)5Khn/0
Genetic
Background
involves: 129S4/SvJae * C57BL/6J * FVB
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Insrtm1Khn mutation (1 available); any Insr mutation (94 available)
Tg(Ckmm-cre)5Khn mutation (4 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
adipose tissue
• larger than normal fat depots in fat pads, including the perianal, subcutaneous and perigonadal fat pads

homeostasis/metabolism
N
• normal serum cholesterol levels
• normoglycemia, up to 11 months of age
• normal glucose tolerance
• normal concentrations of serum insulin
• 20% elevation in serum free fatty acids (FFAs)
• greater than70% elevated, observed from ages 4 to 11 months




Genotype
MGI:2389586
cn11
Allelic
Composition
Insrtm1Khn/Insrtm1Dac
Tg(Ckmm-cre)5Khn/0
Genetic
Background
involves: 129S4/SvJae * C57BL/6J * FVB
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Insrtm1Dac mutation (2 available); any Insr mutation (94 available)
Insrtm1Khn mutation (1 available); any Insr mutation (94 available)
Tg(Ckmm-cre)5Khn mutation (4 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
homeostasis/metabolism
• in the fed state, incomplete penetrance




Genotype
MGI:3583775
cn12
Allelic
Composition
Insrtm1Khn/Insrtm1Khn
Tg(Nes-cre)1Kln/0
Genetic
Background
involves: 129S4/SvJae * C57BL/6J * SJL
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Insrtm1Khn mutation (1 available); any Insr mutation (94 available)
Tg(Nes-cre)1Kln mutation (4 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
adipose tissue
• in females, a two-fold increase in perigonadal adipose tissue is seen
• in males, a 1.5-fold increase in adipose tissue is seen

cellular
• 30% reduced in epididymal sperm content

behavior/neurological
• females exhibit a 20% increase in food intake per gram of body weight
• males did not exhibit any differences in food intake when compared to controls

endocrine/exocrine glands
• reduced number of antral follicles
• 20% of seminiferous tubules lacked a lumen and had few maturing spermatogonia
• normal seminal vesicles, prostate, and epididymis

growth/size/body
• males do not exhibit weight differences on a standard chow diet, but females exhibit a 10-15% increase over controls
• on a high-fat diet, male mice exhibit a 10% weight increase at 14 weeks of age and females exhibit a 20% increase over controls

homeostasis/metabolism
• 20% of seminiferous tubules lacked a lumen and had few maturing spermatogonia
• normal pituitary morphology and LH content; treatment with lupron indicated that LH is expressed, but not secreted from the pituitary in mutant mice
• at 4-6 months of age, mice exhibit a 1.5 fold increase in circulating insulin levels and females exhibit a 2 fold increase
• normal glucose tolerance
• 1.5 fold increased in males
• normal cholesterol concentrations
• 2.5 fold increased in females
• both males and females exhibit a 30% increase in circulating triglycerides
• females exhibit blunted response when injected with insulin and males performed similarly to controls

nervous system
N
• brain weights and general brain histology appeared similar to controls

reproductive system
• reduced number of antral follicles
• 20% of seminiferous tubules lacked a lumen and had few maturing spermatogonia
• normal seminal vesicles, prostate, and epididymis
• 30% reduced in epididymal sperm content




Genotype
MGI:3629045
cn13
Allelic
Composition
Insrtm1Khn/Insrtm1Khn
Tg(Nes-cre)1Kln/0
Genetic
Background
involves: 129S4/SvJae * C57BL/6 * SJL
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Insrtm1Khn mutation (1 available); any Insr mutation (94 available)
Tg(Nes-cre)1Kln mutation (4 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
homeostasis/metabolism
• insulin-induced suppression of hepatic glucose productionas assessed with a euglycemic hyperinsulinemic clamp is attenuated




Genotype
MGI:3620045
cn14
Allelic
Composition
Insrtm1Khn/Insrtm1Khn
Tg(Myhca-cre)1Abel/0
Genetic
Background
involves: 129S4/SvJae * FVB
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Insrtm1Khn mutation (1 available); any Insr mutation (94 available)
Tg(Myhca-cre)1Abel mutation (0 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
cardiovascular system
• hearts have 2.3 times the glycogen content of wild-type and insulin administration reduces levels to those seen in wild-type
• decrease in cardiomyocyte size, with myocyte length and width reduced by 6 and 9%, respectively
• exhibit a persistence of the fetal pattern of cardiac metabolism, indicating that the metabolic switch from predominant glucose metabolism, as seen in neonatal hearts, to fatty acid metabolism that characterizes the adult heart, is defective
• 20-30% reduction in size due to reduced cardiomyocyte size
• decrease in LV posterior wall thickness
• cardiac power, determined by cardiac output and developed pressure, is modestly reduced
• cardiac output is about 15% lower than in wild-type
• physiological concentrations of insulin do not cause an increase in myocyte glucose uptake as in wild-type
• decrease in basal glucose uptake in isolated cardiac myocytes
• 68% increase in the basal rate of cardiac glucose uptake in intact hearts in vivo and in isolated working hearts
• exhibit an increase in left ventricular systolic dimension without any change in diastolic dimension, a 29% reduction in fractional shortening, and 12% reduction in ejection fraction

homeostasis/metabolism
• basal rates of glycolysis in hearts are significantly higher than in wild-type, and do not increase further upon insulin treatment
• basal rates of glucose oxidation in isolated hearts are 30% lower than in wild-type, however insulin increases oxidation rates to a similar rate seen in insulin-treated wild-type hearts
• hearts have 2.3 times the glycogen content of wild-type and insulin administration reduces levels to those seen in wild-type
• basal rates of palmitate oxidation in isolated hearts are 34% lower than in wild-type and do not change further after insulin stimulation

muscle
• hearts have 2.3 times the glycogen content of wild-type and insulin administration reduces levels to those seen in wild-type
• decrease in cardiomyocyte size, with myocyte length and width reduced by 6 and 9%, respectively
• physiological concentrations of insulin do not cause an increase in myocyte glucose uptake as in wild-type
• decrease in basal glucose uptake in isolated cardiac myocytes
• 68% increase in the basal rate of cardiac glucose uptake in intact hearts in vivo and in isolated working hearts
• exhibit an increase in left ventricular systolic dimension without any change in diastolic dimension, a 29% reduction in fractional shortening, and 12% reduction in ejection fraction

cellular
• physiological concentrations of insulin do not cause an increase in myocyte glucose uptake as in wild-type
• decrease in basal glucose uptake in isolated cardiac myocytes
• 68% increase in the basal rate of cardiac glucose uptake in intact hearts in vivo and in isolated working hearts




Genotype
MGI:4941586
cn15
Allelic
Composition
Insrtm1Khn/Insrtm1Khn
Tg(Slc2a4-cre)546Dac/0
Genetic
Background
involves: 129S4/SvJaeSor * C57BL/6 * FVB
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Insrtm1Khn mutation (1 available); any Insr mutation (94 available)
Tg(Slc2a4-cre)546Dac mutation (0 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
growth/size/body
• diabetic mutants show reduced lean body mass but normal locomotion
• 16-week-old males exhibit a 19% decrease in body weight relative to controls; reduction affects lean and fat mass equally, and plasma leptin levels are normal
• diabetic mutants have significantly lower body weights than controls or non-diabetic mutants
• body weight of mutants is normal as weanlings, but mice show persistent growth retardation from that time point onward

adipose tissue
• tissue shows striking disruption of multilocular structure in mutants; this is more pronounced in diabetic mutants
• size is increased in mutants; this is more pronounced in diabetic mutants
• epididymal white adipose tissue has a heterogeneous appearance compared to controls

endocrine/exocrine glands
• euglycemic mutants display beta cell hyperplasia, but otherwise islet structure is normal
• euglycemic mutants display beta cell hyperplasia, but otherwise islet structure is normal
• diabetic mutants show spotted pattern of loss of insulin immunoreactivity in pancreatic islets

liver/biliary system
• glycogen depletion in liver is observed only in diabetic mutant animals; non-diabetic mutants have normal-appearing livers
• this is observed only in diabetic mutant animals; non-diabetic mutants have normal-appearing livers

homeostasis/metabolism
N
• fasting plasma glucagon and beta-hydroxyl butyrate levels are similar to controls
• clamp hyperinsulinemia suppresses free fatty acid levels by 43% but has no effect on GIRKO (Glut4-cre-driven InsR knockout) mutants
• diabetic mutants show increased oxygen consumption when normalized to body weight compared to controls
• during hyperinsulinemic phase of (euglycemic-hyperglycemic) clamp experiments on 13-15 week-old mice, rate of glucose infusion to maintain euglycemia is increased 52% relative to controls (suggesting impaired glucose clearance)
• basal hepatic glucose production is increased by 32% and hyperinsulinemia cannot suppress it (suggesting hepatic insulin resistance)
• glucose disappearance is reduced by 28% and glycolysis is decreased by 33% compared to controls, while glycogen synthesis shows a downward trend
• insulin-dependent glucose uptake is reduced by 34% in muscle
• under basal conditions, mice show slightly elevated blood glucose levels (119 vs 87 mg/dl)
• at 5 weeks, a subset of mice (male and female) show hyperglycemia in the fed state
• prevalence of diabetes (glycemia > +2 standard deviations) in males increases from 20% at 5 weeks to 25% at 12 weeks and 46% at 24 weeks; in females, diabetes prevalence remains around 10% in 5-12 week-old mice
• subset of mice exhibit hyperinsulinemia in fed and fasted state at 12 weeks
• at 12-13 weeks, males show mild glucose intolerance
• glycogen depletion in liver is observed only in diabetic mutant animals; non-diabetic mutants have normal-appearing livers
• 12-13 week old females show mild insulin resistance

behavior/neurological
• non-diabetic mutants show reduced food consumption during the light phase compared to controls (26% vs 34% in controls)




Genotype
MGI:3775310
cn16
Allelic
Composition
Igf1rtm1.1Mhz/Igf1r+
Insrtm1Khn/Insrtm1Khn
Tg(Ckmm-cre)5Khn/0
Genetic
Background
involves: 129/Sv * 129S4/SvJae * C57BL/6 * FVB
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Igf1rtm1.1Mhz mutation (0 available); any Igf1r mutation (86 available)
Insrtm1Khn mutation (1 available); any Insr mutation (94 available)
Tg(Ckmm-cre)5Khn mutation (4 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
mortality/aging
• 3 of 13 mice die by 6 months of age

cardiovascular system
• at P17, left ventricular fractional shortening is reduced 23% compared to Igf1rtm1.1Mhz Insrtm1Khn homozygote controls

respiratory system
• beginning at 3 months of age, 1 of 13 mice appear to gasp for air

behavior/neurological
• beginning at 3 months of age, 1 of 13 mice exhibit less activity than wild-type mice

muscle
• at P17, left ventricular fractional shortening is reduced 23% compared to Igf1rtm1.1Mhz Insrtm1Khn homozygote controls

homeostasis/metabolism
N
• mice exhibit normal glucose homeostasis




Genotype
MGI:3775311
cn17
Allelic
Composition
Igf1rtm1.1Mhz/Igf1rtm1.1Mhz
Insrtm1Khn/Insrtm1Khn
Tg(Ckmm-cre)5Khn/0
Genetic
Background
involves: 129/Sv * 129S4/SvJae * C57BL/6 * FVB
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Igf1rtm1.1Mhz mutation (0 available); any Igf1r mutation (86 available)
Insrtm1Khn mutation (1 available); any Insr mutation (94 available)
Tg(Ckmm-cre)5Khn mutation (4 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
mortality/aging
• mice die within the first month usually after birth 2 days after beginning to gasp for air at P16
• death is either spontaneous or precipitated by the stress of routine handling

cardiovascular system
• mice exhibit irregular and disrupted sarcomeric Z and M lines and increased mitochondria with central crowding compared to wild-type hearts
• at P8 and P20, heart weights reduced 15% and 25%, respectively, compared to in wild-type mice
• mice exhibit decreased heart weight to body weight
• at P17, mice exhibit an increase in left ventricle diameter of 6.7% in diastolic and 57.1% in systolic states
• at P17, left ventricular fractional shortening is reduced 33% compared to Igf1rtm1.1Mhz Insrtm1Khn homozygote controls

behavior/neurological
• at P16, mice become less active than wild-type mice

respiratory system
• at P16, mice gasp for air

growth/size/body
• at P20, mice weigh 15% to 20% less than wild-type mice
• despite normal growth rates during the first two weeks after birth, mice exhibit reduced growth rates during week 3

muscle
• mice exhibit irregular and disrupted sarcomeric Z and M lines and increased mitochondria with central crowding compared to wild-type hearts
• at P17, left ventricular fractional shortening is reduced 33% compared to Igf1rtm1.1Mhz Insrtm1Khn homozygote controls

homeostasis/metabolism
N
• mice exhibit normal glucose homeostasis

cellular




Genotype
MGI:3775313
cn18
Allelic
Composition
Igf1rtm1.1Mhz/Igf1rtm1.1Mhz
Insrtm1Khn/Insr+
Tg(Ckmm-cre)5Khn/0
Genetic
Background
involves: 129/Sv * 129S4/SvJae * C57BL/6 * FVB
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Igf1rtm1.1Mhz mutation (0 available); any Igf1r mutation (86 available)
Insrtm1Khn mutation (1 available); any Insr mutation (94 available)
Tg(Ckmm-cre)5Khn mutation (4 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
cardiovascular system
N
• heart function and morphology is normal

growth/size/body
N
• mice exhibit normal growth rates

homeostasis/metabolism
N
• mice exhibit normal glucose homeostasis




Genotype
MGI:3775314
cx19
Allelic
Composition
Igf1rtm1.1Mhz/Igf1rtm1.1Mhz
Insrtm1Khn/Insrtm1Khn
Genetic
Background
involves: 129/Sv * 129S4/SvJae * C57BL/6 * FVB
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Igf1rtm1.1Mhz mutation (0 available); any Igf1r mutation (86 available)
Insrtm1Khn mutation (1 available); any Insr mutation (94 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
cardiovascular system
N
• heart function and morphology is normal

growth/size/body
N
• mice exhibit normal growth rates

homeostasis/metabolism
N
• mice exhibit normal glucose homeostasis





Contributing Projects:
Mouse Genome Database (MGD), Gene Expression Database (GXD), Mouse Models of Human Cancer database (MMHCdb) (formerly Mouse Tumor Biology (MTB)), Gene Ontology (GO)
Citing These Resources
Funding Information
Warranty Disclaimer, Privacy Notice, Licensing, & Copyright
Send questions and comments to User Support.
last database update
04/16/2024
MGI 6.23
The Jackson Laboratory