Está en la página 1de 11

REVIEW ARTICLE doi: 10.1111/j.1463-1326.2004.00392.

Development and application of rodent models for type 2


diabetes

Desu Chen and Ming-Wei Wang


The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China

The increasing worldwide incidence of diabetes in adults constitutes a global public health burden. It is predicted that
by 2025, India, China and the United States will have the largest number of people with diabetes [1]. According to
the 2003 estimates of the International Diabetes Federation, the diabetes mellitus prevalence in the USA is 8.0%
and approximately 9095% of diabetic Americans have type 2 diabetes about 16 million people. Type 2 diabetes
is a complex, heterogeneous, polygenic disease characterized mainly by insulin resistance and pancreatic b-cell
dysfunction. Appropriate experimental models are essential tools for understanding the molecular basis, pathogenesis
of the vascular and neural lesions, actions of therapeutic agents and genetic or environmental influences that increase
the risks of type 2 diabetes. Among the animal models available, those developed in rodents have been studied most
thoroughly for reasons such as short generation time, inherited hyperglycaemia and/or obesity in certain strains and
economic considerations. In this article, we review the current status of most commonly used rodent diabetic models
developed spontaneously, through means of genetic engineering or artificial manipulation. In addition to these
models, the Psammomys obesus, rhesus monkeys and many other species are studied intensively and reviewed by
Shafrir [2], Bailey and Flatt [3,4] and Hansen [5].
Keywords: pharmacology, rodents, type 2 diabetes
Received 23 December 2003; returned for revision 5 April 2004; revised version accepted 20 April 2004

Spontaneously Diabetic Rodents of Long-Evans rats, which was purchased from Charles
River, Canada in 1982. A strain of rats developed from
The information described in this review was obtained
this rat by selective breeding has since been maintained
through a search of MEDLINE/PubMed for literatures
at the Otsuka Pharmaceutical and named OLETF [6].
published between 1993 and 2003. Search terms used
The characteristics of Otsuka Long-Evans Tokushima
include type 2 diabetes, OLETF rats, GK rats, db/db
Fatty (OLETF) rats include: (a) late onset of hypergly-
mice, ZDF rats and ob/ob mice. The analysis of our
caemia (after 18 weeks of age); (b) a chronic course of
search results is summarized in table 1, and the details
disease; (c) mild obesity; (d) clinical onset of diabetes
of each model are given in the order of its citation
mostly in males; (e) multiple recessive genes are
frequency and synopsized in table 2.
involved in the induction of diabetes, and the transmit-
tal of one of the diabetogenic genes, designated as odb-1,
is linked to the X-chromosome of OLETF rats; (f)
Otsuka Long-Evans Tokushima Fatty Rats
the changes of pancreatic islets can be classified into
A spontaneously diabetic rat with polyuria, polydipsia and three stages an early stage (less than 9 weeks of age)
mild obesity was discovered in 1984 in an outbred colony with mild lymphocyte infiltration, a hyperplastic stage

Correspondence:
Dr Ming-Wei Wang, The National Center for Drug Screening, 189, Guo Shou Jing Road, Shanghai 201203, PR China.
E-mail:
mwwang@siniwest.com

# 2004 Blackwell Publishing Ltd Diabetes, Obesity and Metabolism, 7, 2005, 307317 307
RA Rodent models for type 2 diabetes D. Chen and M.-W. Wang

Table 1 Citation frequency of spontaneously diabetic rodents increases in meal size, overall hyperphagia and obesity
[810]. Analyses of patterns of hypothalamic gene
Rodent Model Articles Frequency (%) expression in OLETF rats indicate the presence of a
Otsuka Long-Evans Tokushima Fatty rats 290 33.8 primary deficit in dorsomedial hypothalamus neuropep-
Goto-Kakizaki rats 170 19.8 tide Y signalling. Thus, the obesity in OLETF rats may be
db/db (C57BL/KsJ-db/db) mice 148 17.3 the outcome of two regulatory disruptions, one depending
Zucker Diabetic Fatty rats 135 15.8
upon a peripheral pathway such as meal satiety and the
ob/ob (C57BL/6J-ob/ob) mice 114 13.3
other relating to a central pathway critical to overall
energy balance [11].
(1040 weeks of age) with hyperplastic alterations In OLETF rats, hypertriglyceridaemia led to elevated
and fibrosis in or around islets and a final stage triglyceride (TG) storage in islets, which subsequently
(more than 40 weeks of age) showing atrophy of islets; inhibited glucose-induced insulin secretion, via reduced
and (g) diabetic nephropathy manifested by diffused glucokinase activity in the islet. Fat droplets in islets
glomerulosclerosis and nodular lesions (e.g. thickening may play an important role in accelerating the develop-
of basement membranes, mesangial proliferation ment of diabetes [12].
and fibrin cap). These clinical and pathological fea- The total content of glucose transporter 4 (GLUT4)
tures in OLETF rats resemble those of human type 2 protein significantly decreased while the plasma mem-
diabetes [7]. brane content of the GLUT4 protein in muscles of the
Expression of cholecystokinin A (CCK-A) receptor diabetic OLETF rats was increased in the basal state.
mRNA in the pancreas, small intestine and brain was Hyperinsulinaemic clamps increased GLUT4 levels in
not detected in OLETF rats by the reverse transcriptase the plasma membrane in muscles of normal rats but
polymerase chain reaction (RT-PCR). The lack of CCK-A failed to do so in diabetic OLETF rats. The distribution
receptors results in a reduced ability to process nutrient- of GLUT4 in OLETF rats is reminiscent of the character-
elicited gastrointestinal satiety signals, which leads to istics of human type 2 diabetes [13].

Table 2 Synopsis of pharmacological applications of spontaneously diabetic rodents

Species Pharmacologically related characteristics Studies of anti-diabetic agents

OLETF rats Lack of CCK-A receptors; deficit Beneficial effects of Ca2 antagonist (Cilnidipine),
in dorsomedial hypothalamus neuropeptide Y metformin and truncated GLP-1 on insulin-resistance;
signalling; elevated triglyceride storage in islets; PPARg (pioglitazone) agonist improves daily profiles
significantly decreased total content of GLUT4 in muscles of energy expenditure
GK rats Twofold higher activities of both basal and Long-term and favourable influences of GLP-1 or
insulin-stimulated PTP1B; GK-VMH rats as a exendin-4 on b-cell mass and glycemic control
useful model for non-obese type 2 diabetes
with microangiopathy and macroangiopathy
db/db mice Correlations between the glucagon/insulin ratio Long-lasting glycemic control by GLP-1 receptor
and the hepatic glucose-6-phosphatase/fructose- agonist in a dose-dependent manner; combination
1,6-diphosphatase activities; elevated renal therapy of PPARg and PPARa agonists improved
cortical activities of PI3 kinase, protein kinase B, body weight and glucose-stimulated insulin secretion;
extracellular signal-regulated protein kinase 1/2-type PTP1B inhibitors modulates insulin signalling in
mitogen-activated protein kinase the liver and fat tissues
ZDF rats 2555% reduction of GLUT4 in the adipose tissue, Nicotinamide or aminoguanidine halted inducible nitric
heart and skeletal muscle; loss of pancreatic oxide synthase expression in islets; metformin
duodenal homeobox gene expression; significantly reduced FFA levels; rosiglitazone
FFA and nitric oxide induced suppression maintained cell proliferation and blocked b-cell death;
of insulin output b-3-adrenergic receptor agonist improved glucose
tolerance and insulin responsiveness
ob/ob mice Overproduced neuropeptide Y in the hypothalamus; GLP-1 increased frequency of large, rapid spikes
45% higher hepatic microsomal TG transfer protein of [Ca2]i; exendin-4 treatment led to weight loss
mRNA level; b-adrenergic receptor mRNA levels and improved insulin sensitivity; PTP1B antisense
in white and brown adipose tissues reduced by oligonucleotides decreased PTP1B protein and
300-fold elevating other elements in insulin signalling pathway

CCK-A, cholecystokinin A; FFA, free fatty acid; GLUT4, glucose transporter protein 4; GLP-1, glucagons-like peptide 1; GK, Goto-Kakizaki;
OLETF, Otsuka Long-Evans Tokushima Fatty; PTP1B, protein tyrosine phosphatase 1B; TG, triglyceride.

308 Diabetes, Obesity and Metabolism, 7, 2005, 307317 # 2004 Blackwell Publishing Ltd
D. Chen and M.-W. Wang Rodent models for type 2 diabetes RA

Exercise training is effective at preventing the pathogenic factors: (a) three independent loci containing
development of diabetes mellitus in OLETF rats. The genes responsible for impaired insulin secretion;
cumulative incidences of diabetes mellitus in sedentary (b) heritable gestational metabolic (hyperglycaemic)
(in conventional cages) and trained-control rats (in impairment inducing deficiency in endocrine pancreas;
cages with a fixed rotary wheel) were 78 and 50%, and (c) secondary (acquired) loss of b-cell differentiation
respectively, while neither trained OLETF rats (in due to long-term exposure to hyperglycaemia (glucotoxi-
exercise wheel cages) nor non-diabetic Long-Evans city) [23].
rats became diabetic. The preventive effect of exercise Through metabolism, glucose generates adenosine
training against the development of type 2 diabetes triphosphate (ATP) needed for the closure of the
lasted for at least 3 months after the cessation of exercise ATP-sensitive K channels and membrane depolar-
[14,15]. ization. In the GK rat pancreatic islet, K induces a
Injection of insulin to OLETF rats was effective at delayed [Ca2]i response that probably results from a
preventing b-cell dysfunction and morphological defective metabolism of glucose in this tissue [24,25].
changes in the pancreas [16]. Cilnidipine, a long-acting Immunoprecipitation of protein tyrosine phosphatase
dihydropyridine Ca2 antagonist, had beneficial effects 1B (PTP1B) from skeletal muscle lysates, and analysis
on both insulin resistance and hypertension in OLETF of the enzyme activity indicate that both basal and
rats [17]. Metformin-induced improvement of insulin insulin-stimulated PTP1B activities were twofold higher
resistance in OLETF rats could lower blood pressure, in skeletal muscle of diabetic GK rats than those of
decrease sympathetic activity and reduce body weight normal rats. This alteration was associated with an
[18]. Acarbose prevented and reversed the metabolic increased expression of the enzyme protein. Elevated
derangement and histopathological changes of pancreas PTP1B activity would enhance tyrosine dephosphoryl-
in OLETF rats. Moreover, treatment with acarbose, even ation of insulin receptor and its substrates and thereby
for a short period, produced a marked delay in the devel- lead to impaired glucose tolerance and insulin resist-
opment of insulin insensitivity and obvious diabetes ance in GK rats [26].
[19]. Administration of pioglitazone, a peroxisome pro- The effects of GLP-1 and its long-acting analogue,
liferator-activated receptor g (PPARg) agonist, improved exendin-4 (Ex-4), were investigated during the predia-
daily profiles of energy expenditure through affecting betic period of GK rats. Treatment with GLP-1 or Ex-4
glucose and fat metabolism [20]. demonstrated long-term and favourable influences on
A long-term infusion of truncated glucagon-like b-cell mass, resulting from enhancement of differentia-
peptide 1 (GLP-1) increased the glucose infusion rate tion (neogenesis), proliferation and glycaemic control in
significantly during a euglycaemichyperinsulinaemic adulthood. Compared to untreated animals, basal
clamp experiment. This suggests that truncated GLP-1 plasma glucose levels in 2-month-old treated rats were
is capable of augmenting insulin action in peripheral significantly decreased [27].
tissues of diabetics, which can contribute, in part, to In GK rats, a marked prolongation in mean circulation
improve glucose intolerance in OLETF rats [21]. time and a significant reduction in segmental blood flow
appeared at an early stage of diabetes. This tendency
continued for a period of 5 months, and the endothe-
Goto-Kakizaki Rats
lial/pericyte ratio was found to be higher in 8-month-old
The Goto-Kakizaki (GK) rat is a non-obese rodent model animals as well as in animals greater than 2430 months
of mild type 2 diabetes with early hyperglycaemia, of age. These findings suggest that the GK rat appears to
hyperinsulinaemia and insulin resistance. This model be a suitable model for experimental studies on initial or
was obtained by selective breeding of individuals with latent-phase diabetic retinopathy [2830].
mild glucose intolerance from a non-diabetic Wistar rat Prolonged exposure to hyperglycaemia is one of Key
colony. factors in the induction of progressive diabetic nephrop-
In GK rats, the neonatal b-cell-mass deficit is con- athy in humans. The same phenomenon was observed
sidered to be the primary defect leading to basal hyper- in GK rats. Noticeable morphological changes in kid-
glycaemia, which is detectable around 3.5 weeks of age. neys, such as segmental glomerulosclerosis and tubu-
After 8 weeks, hyperglycaemia deteriorates and glu- lointerstitial fibrosis, were found only in 2-year-old
cose-stimulated insulin release by the islets is more animals. The renal alterations seen in GK rats at a later
severely impaired [22]. Heritability of defective b-cell stage were similar to those of progressive human diabetic
mass and its function in the GK model is thought nephropathy [31]. Experimental ventromedial hypo-
to reflect the complex interactions of the following thalamic (VMH) lesions induced marked hyperglycaemia

# 2004 Blackwell Publishing Ltd Diabetes, Obesity and Metabolism, 7, 2005, 307317 309
RA Rodent models for type 2 diabetes D. Chen and M.-W. Wang

and a distinct reduction in pancreatic insulin content in (GSIS) in db/db mice. Body-weight gain in db/db mice
male GK rats. Histological examinations of the kidneys was less with dual PPAR agonists treatment than with a
from these rats revealed that the glomerular basement single agonist such as pioglitazone or pioglitazone plus
membrane was thicker than that of controls. The bezafibrate. Plasma glucose, insulin, TG and free fatty
descending aorta in GK-VMH rats also showed acid (FFA) levels were significantly ameliorated and
morphologic changes in the intima, characteristic of an GSIS was increased [38]. Adipocyte complement-related
early stage of atherosclerosis. Thus, male GK-VMH rats may protein (30 kDa) (Acrp30, adiponectin or AdipoQ) is a
be a useful animal model for non-obese type 2 diabetes fat-derived secreted protein that circulates in blood.
with typical complications such as microangiopathy and Adipose tissue expression of Acrp30 is lower in insulin-
macroangiopathy [32,33]. resistant state, and it is implicated in the regulation
of in vivo insulin sensitivity. In obese diabetic db/db
mice, mean plasma Acrp30 protein levels increased
Db/db (C57BL/KsJ-db/db) Mice
after long-term treatment using PPARg agonists. Induc-
The diabetes db gene mutation occurred spontaneously tion of adipose tissue Acrp30 expression and conse-
in the leptin-receptor-deficient C57BL/KsJ strain of mice quent rises in circulating Acrp30 levels represent a
from Bar Harbor, ME, USA [34]. It is an autosomal reces- novel potential mechanism for PPARg-mediated
sive mutation on chromosome 4 with complete pene- enhancement of whole-body insulin sensitivity. Acrp30
trance. is likely to be a biomarker of in vivo PPARg activation
In diabetic mutant db/db mice, progressive impair- [39].
ment of insulin response to glucose is observable with In db/db mice, PTP1B antisense oligonucleotide treat-
increasing age. Blood glucose levels do not differ signifi- ment normalized plasma glucose levels, postprandial
cantly between 5-week-old db/db and / mice but glucose excursion and HbA1c. Hyperinsulinemia was
increase with age in the former until they are 16 weeks also reduced, with improved insulin sensitivity. PTP1B
old. Consistent with the idea of a protective effect of protein and mRNA were reduced in liver and fat tissues
oestrogen on the pancreatic b-cell, female diabetic db/ with no effect on skeletal muscle. Insulin signalling
db mice survive longer than males. proteins, insulin receptor substrate 2 and phosphatidy-
While plasma levels of insulin and glucagon in db/db linositol 3 (PI3) kinase regulatory subunit p50a were
mice, which peaked at 7 weeks of age, do not reflect the increased, and PI3-kinase p85a expression was
state of hyperglycaemia, the G/I (glucagon/insulin) ratio decreased in the liver and fat tissues. These changes
is roughly in parallel with the development of hypergly- in protein expression correlated with
caemia. Analysis of individual values revealed statistic- increased insulin-stimulated protein kinase B phosphor-
ally significant correlations between plasma glucose ylation. The expression of liver gluconeogenic enzymes,
levels and hepatic glucose-6-phosphatase or fructose- phosphoenolpyruvate carboxykinase and fructose-1, 6-
1,6-diphosphatase activities. There were also marked bisphosphatase, was also downregulated. The findings
correlations between the G/I ratio and activities of suggest that PTP1B modulates insulin signalling in the
these two hepatic gluconeogenic enzymes [35]. liver and fat tissues and that therapeutic modalities tar-
NN2211, a long-acting, metabolically stable GLP-1 geting PTP1B inhibition may have clinical benefits in
derivative, with enhanced stability due to attachment type 2 diabetes [40]. Indeed, a series of azolidinediones,
of the fatty acid moiety, dose dependently reduced gly- prepared as PTP1B inhibitors, with IC50 values in the
caemia levels in db/db mice, with an antihyperglycaemic range of 0.120.30 mM, normalized plasma glucose and
activity still evident 24 h after treatment. b-Cell insulin levels in db/db mice [41].
proliferation rate and mass size were also significantly Experimental data demonstrate that a variety of
increased [36]. Another novel GLP-1 receptor agonist, receptor signalling pathways are activated in the
ZP10A, decreased db/db mouse glycosylated haemo- renal cortex of db/db mice, pointing to a role of
globin A1c (HbA1c) from 8.4% to 6.2% in a dose- augmented insulin receptor activity in nephropathy
dependent manner. Fasting blood glucose and glucose of type 2 diabetes. In db/db mice, renal cortical
tolerance after an oral glucose tolerance test were activities of PI3 kinase, protein kinase B (PKB) and
notably improved. These effects lasted for 40 days after extracellular signal-regulated protein kinase (ERK)1/2-
cessation of treatment [37]. type mitogen-activated protein (MAP) kinase were all
Studies have shown that combination therapy of elevated considerably. The increased renal cortical
PPARg and PPARa agonists may have beneficial effects PI3 kinase activity was partially due to insulin recep-
on body weight and glucose-stimulated insulin secretion tor activation, as PI3 kinase activity associated with

310 Diabetes, Obesity and Metabolism, 7, 2005, 307317 # 2004 Blackwell Publishing Ltd
D. Chen and M.-W. Wang Rodent models for type 2 diabetes RA

insulin receptor b-chain was increased nearly with reduced islet insulin secretion [47]. Chronic and
fourfold. Additionally, the kinase activity of immuno- progressive hyperglycaemia in diabetic ZDF rats is
precipitated b-chain in the diabetic renal cortex and related to the loss of insulin and pancreatic duodenal
tyrosine phosphorylation of insulin receptor were homeobox (PDX-1) mRNAs and lack of glucose-
both augmented [42]. stimulated insulin secretion. Prevention of hyperglycae-
mia could block the associated defects in insulin and
PDX-1 gene expression and improve insulin secretion
Zucker Diabetic Fatty Rats
[48].
The diabetic trait in this model originated from a colony The lipoapoptosis of b-cells observed in fat-laden
of outbred Zucker rats in the laboratory of Dr Walter islets of ZDF rats results from the overproduction of
Shaw at Eli Lilly Research Laboratories in Indianapolis, ceramide, an initiator of the apoptotic cascade which is
IN, USA, during 19741975. Several groups of animals induced by long-chain fatty acids (FA). The onset of type
with diabetic lineage were identified and redefined at 2 diabetes is preceded by a striking increase in the
the beginning of 1981. The inbred line of Zucker diabetic plasma levels of FFA and by a sixfold rise in TG content
fatty (ZDF) rats was established in 1985 and developed in the pancreatic islets. FFA could be the signal from
to a genetic model in 1991. adipocytes that elicits b-cell compensation sufficient to
Male Zucker rats homozygous for non-functional lep- prevent diabetes [49,50]. Studies show that FFA-
tin receptors (fa/fa) develop obesity, hyperlipidemia and induced suppression of insulin output in prediabetic
hyperglycaemia, but rats with homozygous dominant ZDF rats is mediated by nitric oxide (NO). In cultured
(/) and heterozygous (fa/) genotypes remain lean prediabetic ZDF islets, FFA induced a fourfold rise in
and normoglycaemic. Insulin resistance occurs in NO, upregulated mRNA of inducible nitric oxide
young fa/fa rats followed by evolution of an insulin- synthase (iNOS) and reduced insulin output. Both nico-
secretory defect that triggers hyperglycaemia [43]. tinamide and aminoguanidine, which lower NO, pre-
Thus, the ZDF male rat has become an experimental vented the FFA-mediated increase in iNOS mRNA,
model for type 2 diabetes, with a predictable progression reduced NO and minimized the loss of insulin secretion.
from prediabetic to diabetic state. Hyperglycaemia is In vivo nicotinamide or aminoguanidine treatment of
initially manifested at about 7 weeks of age, and all prediabetic ZDF rats stopped iNOS expression in islets
obese male rats are fully diabetic by 12 weeks. Between and mitigated b-cell dysfunction through blocking b-cell
7 and 10 weeks, serum insulin levels are high but subse- destruction and hyperglycaemia [51].
quently drop as pancreatic b-cells cease to respond to In ZDF rats, hyperphagia leads to hyperinsulinemia,
glucose stimulus [44]. In this diabetic model, GLUT4 in which upregulates transcription factors that stimulate
the adipose tissue, heart and skeletal muscle is reduced lipogenesis. This causes ectopic deposition of triacylgly-
by 2555% [45]. cerol in non-adipocytes, thereby providing FA substrate
The ZDF rat has both insulin resistance (as a result of a for pathological non-oxidative metabolism, such as cer-
mutant leptin receptor that causes obesity) and inad- amide synthesis. In b-cells and myocardium, the resulting
equate b-cell compensation. Studies demonstrate that functional impairment and apoptosis lead to diabetes
the ZDF rat carries a genetic defect in b-cell transcription and cardiomyopathy. Interventions that lower ectopic
that is inherited independently of the leptin receptor lipid accumulation or block FA non-oxidative metabo-
mutation and insulin resistance. The genetic reduction lism and ceramide formation could completely prevent
in b-cell gene transcription in homozygous animals these complications [52].
probably contributes to the development of diabetes in Metformin prevented hyperglycaemia in diabetic ZDF
the background of insulin resistance [46]. In prediabetic rats aged between 6 and 12 weeks and significantly
ZDF rats, there is no change in insulin mRNA levels. reduced TG and FFA levels. It delays the onset of dia-
However, significant reductions (3070%) of many other betes in the ZDF rat, which is correlated with improve-
islet mRNA levels, such as glucokinase, mitochondrial ment in b-cell function, consistent with the lipotoxicity
glycerol-3-phosphate dehydrogenase, voltage-dependent hypothesis for adipogenic diabetes [53]. Treatment with
Ca2 and K channels, CA2-ATPase and transcription rosiglitazone protected obese ZDF rats against loss of
factor islet-1, could be detected. In contrast, glucose-6- b-cell mass through maintaining cell proliferation and
phosphatase and 12-lipoxygenase mRNA levels are blocking increased b-cell death [54].
increased by 4050%. Gene-expression patterns in the Insulin release was defective in ZDF obese rats and
islets of diabetic ZDF rats show marked alterations, could be partially restored by GLP-1 therapy. In ZDF islets,
including a decrease in insulin mRNA levels associated the action of GLP-1 is mediated via Ca2-independent

# 2004 Blackwell Publishing Ltd Diabetes, Obesity and Metabolism, 7, 2005, 307317 311
RA Rodent models for type 2 diabetes D. Chen and M.-W. Wang

signalling pathway. Infusion of GLP-1 (737) in ob/ mice. These observations indicate that obesity-
hyperinsulinemic and hyperglycaemic ZDF rats induced type 2 diabetes in mice causes increases in
produced a transitory increase in plasma insulin hepatic MTP expression and increases the secretion of
concentration and normalized blood glucose level TG-rich lipoproteins [63].
[5557]. Studies on dipeptidyl peptidase-IV (DPP-IV) All b-adrenergic receptor (b-AR) mRNA levels in
inhibitors in ZDF rats indicated their therapeutic white and brown adipose tissues were dramatically
value of delaying progression from impaired glucose reduced (by approximately 300-fold) in 12-week-old
tolerance to type 2 diabetes [58]. obese mice. The reduction in the expression of b-1AR
Long-term b-3-adrenergic receptor agonist treatment and b-3AR impaired b-AR agonist-stimulated, adenylyl-
in obese ZDF rats improved glucose tolerance and insu- cyclase response, over a broad concentration range, by
lin responsiveness through a mechanism similar to that greatly lowering the maximum stimulation and shifting
induced by chronic cold exposure, i.e. by stimulating the adrenergic sensitivity at low concentrations from a
facultative thermogenesis, mitochondriogenesis and glu- mixed b-1AR/b-2AR response to predominantly b-2AR
cose utilization in brown and white adipose tissues. The [64].
reduction in plasma FFA levels may enhance glucose The effects of GLP-1 (736 amide, GLP-1a) on [Ca2]i
uptake in skeletal muscles (a tissue that does not express were determined using Fura-2 fluorescence ratio
typical b-3-adrenergic receptors) via the glucosefatty imaging on cultured ob/ob mouse pancreatic b-
acid cycle [59]. cells. GLP-1a increases the frequency of sustained, stable
plateau responses to elevated glucose and the frequency
of large, rapid spikes of increased [Ca2]i associated with
Ob/ob (C57BL/6J-ob/ob) Mice
either plateaus or oscillations [65]. A series of novel
The C57BL/6J-ob/ob mouse originated from the Jackson GLP-1 analogues displays resistance to plasma DPP-IV
Laboratory in Bar Harbor, ME, USA [60]. The ob gene degradation and enhances insulin-releasing and antihy-
was transferred from the stock of origin onto the B/6 perglycaemic activities in 20- to 25-week-old obese dia-
genomic background and is located on chromosome 6. betic ob/ob mice [66]. Administration of Ex-4 leads to an
The obesity syndrome, which is prominent in ob/ob anti-diabetic effect associated with weight loss and
mice, results from the lack of leptin, a hormone released improved insulin sensitivity (up to 32% and 49% respect-
by fat cells and which acts on brain to suppress feeding ively) in ob/ob mice [67]. Studies on the effects of the
and stimulate metabolism. The ob/ob mice are less GIP receptor antagonist and exendin (939) amide on
hyperglycaemic than the db/db mice. GIP- and GLP-1-induced cyclic AMP generation, insulin
Neuropeptide Y (NPY) is a neuromodulator impli- secretion and postprandial insulin release in ob/ob mice
cated in the control of energy balance and is overpro- provide evidence that GIP is the major physiological
duced in the hypothalamus of ob/ob mice. The same incretin of the enteroinsular axis. Administration of
strain of mice deficient in NPY is less obese because of (Pro3)GIP, exendin (939) amide or a combination of
reduced food intake and increased energy expenditure. both peptides to fasted ob/ob mice decreased the plasma
As a result, these animals are less affected by diabetes, insulin responses by 42%, 54% or 49% respectively.
sterility and somatotropic defects. This suggests that The hyperinsulinaemia of non-fasted ob/ob mice was
NPY is a central effector of leptin deficiency [61]. decreased by 19%, 27% or 18%, respectively, following
Chronically elevated NPY levels in the hypothalamus, injection of (Pro3)GIP, exendin (939) amide or com-
as seen in genetically obese ob/ob mice, are associated bined peptides [68].
with obesity, a typical symptom of type 2 diabetes, Semiquantitative RT-PCR experiments show that
and infertility. Crossing the Y2-receptor knockout PPARg2 mRNA was significantly upregulated in ob/ob
mouse [Y2(/)] onto the ob/ob background attenuates mouse liver in comparison with that of wild-type mice.
increased adiposity, hyperinsulinaemia, hyperglycaemia In addition, insulin resistance index was positively asso-
and increased hypothalamicpituitaryadrenal axis ciated with liver PPARg2 mRNA expression. The find-
activity of ob/ob mice [62]. ings suggest a possible compensatory response through
Recent studies demonstrate that microsomal TG trans- which type 2 diabetic and obese animals strive to main-
fer protein (MTP) is a rate-limiting factor for the assem- tain insulin sensitivity in liver [69]. Experimental obes-
bly and secretion of apoB-containing lipoproteins. ity in rodents is related to severely defective resistin
Obese diabetic ob/ob mice have 45% higher hepatic expression. In response to several different classes of
MTP mRNA level, 54% higher microsomal TG transfer PPARg agonists, resistin expression in ob/ob mouse adi-
activity and 70% higher TG concentration compared to pose tissue was increased [70].

312 Diabetes, Obesity and Metabolism, 7, 2005, 307317 # 2004 Blackwell Publishing Ltd
D. Chen and M.-W. Wang Rodent models for type 2 diabetes RA

Recent studies indicate that a reduction in PTP1B Artificially Induced Diabetic Rodents
activity is sufficient to increase insulin-dependent
metabolic signalling and to improve insulin sensitivity Neonatal Streptozotocin-induced Diabetes
in ob/ob mice. In PTP1B-antisense-oligonucleotide-
Intraperitoneal administration of streptozotocin (STZ) to
treated mice, in which PTP1B protein was decreased
neonatal rats (2 days postpartum) produces a type 2
by 60% in the liver, there was elevated tyrosine
diabetes mellitus model in adulthood. At the fourth
phosphorylation of insulin receptor and its substrates
week, fed serum glucose concentration is normal or
(IRS-1 and IRS-2). A threefold increase in IRS-2-
mildly elevated due to b-cell regeneration. However,
associated PI3 kinase activity was accompanied by
hyperglycaemia develops progressively with age, and
augmentations of PKB serine phosphorylation in the
by the twelfth week, marked impairment of glucose-
liver upon insulin stimulation, phosphorylation of PKB
stimulated insulin release could be observed [7375].
substrates and glycogen synthase kinases (3a and 3b)
[71].
STZ-Spontaneously Hypertensive Rat (SHR)

Two-day-old SHRs intraperitoneally injected with STZ


Genetically Engineered Diabetic Mice develop a hyperglycaemic syndrome, associated with
In recent years, genetically engineered mouse models, other biochemical, morphological and haemodynamic
including transgenic and knockout mice, have been properties that, to some extent, confer insulin resistance
developed for the study of the pathophysiological con- combined with hypertension. Plasma glucose levels in
sequences of defined alterations in a single gene or in a STZ-SHRs increase in a dose-dependent manner at the
set of candidate diabetogenes, especially the expression twelfth week. During long-term observations, hyper-
of key actors in insulin signalling, action or secretion glycaemia persisted until the twenty-eighth week but later
(table 3 reproduced with permission of EMBO Reports) gradually ameliorated, accompanied by continued hyper-
[72]. In this review, besides the phenotypic alterations of tension as measured at the fifty-second week [76,77].
the knockout models, the specific roles of individual
genes in the control of glucose homeostasis are also
Fat-fed/STZ-induced Diabetic Rodents
comprehensively addressed.
Genetically engineered mice open the door for inves- Fat-fed/STZ diabetic rodents, developed by combination
tigating the role of genegene or geneenvironment of diet-induced insulin resistance and relatively-low-dose
interactions in the development of type 2 diabetes. STZ, provide a novel animal model for type 2 diabetes. It

Table 3 Monogenic and polygenic diabetic mice

General or tissue-specific knockout mice Phenotypic alterations

GK/ or b-cell GK/ Impaired insulin secretion, mild diabetes


GLUT2/ Moderate insulin resistance, glucose intolerance, decreased adipose mass
GLUT4/ Insulin resistance, diabetic hypertension
GLUT4/ Diabetic ketoacidosis, early postnatal death
IR/ Growth retardation, mild insulin resistance, b-cell hyperplasia, hyperinsulinaemia
IRS-1/ Severe insulin resistance, b-cell hyperplasia, diabetes in 50% of adult mice
IRS-2/ Severe insulin resistance in liver, limited b-cell hyperplasia, diabetes in adult mice
IR/IRS-1/ Sever insulin resistance, early onset of diabetes, marked b-cell hyperplasia
IR/IRS-1/IRS-2/ Insulin resistance, b-cell hyperplasia, diabetes in adult mice
IRS-1/GK/ Lack of glucose-induced first phase insulin release, glucose intolerance with ageing
bIRKO Sever insulin resistance, fasting hyperglycaemia, b-cell hyperplasia
LIRKO Mild hyperglycaemia
Liver GK/ Normal glucose homeostasis, dyslipidaemia, increased adiposity
MIRKO Insulin resistance, glucose intolerance
MG4KO Diabetic ketoacidosis, early postnatal death
PG4KO Insulin resistance in muscle and liver, glucose intolerance, hyperinsulinaemia

GK, Goto-Kakizaki; GLUT, glucose transporter protein; IR, insulin receptor; IRS, insulin receptor substrate.

# 2004 Blackwell Publishing Ltd Diabetes, Obesity and Metabolism, 7, 2005, 307317 313
RA Rodent models for type 2 diabetes D. Chen and M.-W. Wang

simulates the human syndrome and is suitable for tegies aimed at improving insulin action or b-cell func-
expedient in vivo evaluation of anti-diabetic agents. tion. The newly developed fat-fed/STZ-induced diabetic
In C57BL/6J mice, insulin resistance could be induced model offers significant advantages in replicating the
by diets enriched in either fructose or fat, and hypergly- natural history and metabolic characteristics of human
caemia is introduced by injecting a dose of STZ that does conditions and is also cost-effective compared to the
not cause diabetes in chow-fed mice. Insulin concentra- genetic models currently available. Because it is a com-
tions initially increase in response to the fructose- or fat- plex, heterogeneous, multifactorial syndrome resulting
rich diets and then decrease to levels still higher than from both genetic susceptibility and environmental risk
those in chow-fed mice following STZ injection. Accom- factors, more promising animal models that closely
panied by this decrease in insulin levels after STZ injec- simulate human type 2 diabetes regarding inheritance
tion, fat- or fructose-fed C57BL/6J mice become traits, environmental risks, pathogeneses and complica-
significantly hyperglycaemic [78]. tions have yet to be developed in the future.
Another rodent model employs 7-week-old, non-
obese, outbred male SpragueDawley rats. They are fed
with a high-fat diet (40% of calories as fat) for 2 weeks Acknowledgement
and then injected with STZ (50 mg/kg intravenously).
We are indebted to Dr D. E. Mais for his valuable comments
Before STZ injection, fat-fed rats have glucose con-
and suggestions.
centrations similar to chow-fed counterparts. Plasma
insulin levels in response to oral glucose are increased
twofold by fat feeding, and adipocyte glucose clearance References
under maximal insulin stimulation is significantly
reduced, suggesting that fat feeding induces insulin 1 King H, Aubert RE, Herman WH. Global burden of dia-
resistance. STZ injection elevates glucose, insulin, FFA betes, 19952025: prevalence, numerical estimates, and
projections. Diabetes Care 1998; 21: 14141431.
and TG concentrations. Fat-fed/STZ rats are not insulin
2 Shafrir E. Animal models of non-insulin-dependent dia-
deficient compared to normal chow-fed rats but display
betes. Diabetes Metab Rev 1992; 8: 179208.
hyperglycaemia associated with reduced insulin- 3 Bailey CJ, Flatt PR. Islet defects and insulin resistance in
stimulated adipocyte glucose clearance [79]. models of obese non-insulin-dependent diabetes. Dia-
betes Metab Rev 1993; 9 (Suppl. 1): 43S50S.
4 Bailey CJ, Flatt PR. Animal syndromes resembling type 2
Summary
diabetes. In: Pickup JC, Williams G eds. Textbook of
With worldwide rises of metabolic disease incidences, Diabetes, 3rd edn, Vol. 1. Oxford: Blackwell Science,
development of novel investigational methods and tech- 2003; 25.125.30.
nologies becomes one of the top priorities in combating 5 Hansen BC. Pathophysiology of obesity-associated type II
diabetes (NIDDM): implications from longitudinal studies
this health crisis. Newly emerged and available rodent
of non-human primates. Nutrition 1989; 5: 4850.
models for the study of obesity, diabetes, hyperlipid-
6 Kawano K, Hirashima T, Mori S, Saitoh Y, Kurosumi M,
aemia and other metabolic complications would
Natori T. Spontaneous long-term hyperglycemic rat with
accelerate the ongoing process of finding a cure. diabetic complications. Otsuka Long-Evans Tokushima
Spontaneously diabetic rodent models such as OLETF Fatty (OLETF) Strain. Diabetes 1992; 41: 14221428.
rats, GK rats, db/db mice, ZDF rats and ob/ob mice are 7 Kawano K, Hirashima T, Mori S, Natori T. OLETF (Otsuka
most commonly used in drug discovery. OLETF rats Long-Evans Tokushima Fatty) rat: a new NIDDM rat strain.
closely simulate the metabolic abnormalities of the Diabetes Res Clin Pract 1994; 24 (Suppl.): S317S320.
human syndrome, especially the diabetic nephropathy. 8 Funakoshi A, Miyasaka K, Shinozaki H, Arita Y, Nakano I,
While the GK rat appears to be a suitable model for non- Nawata H. Regulation of pancreatic exocrine function in
obese diabetes, ZDF rats are generally applied to studies Otsuka Long-Evans Tokushima Fatty (OLETF) rats with-
of diabetes with obesity and cardiovascular complica- out gene expression of cholecystokinin-A receptor. Intern
Med 1996; 35: 249256.
tions due to the dyslipidaemia background. Obesity is
9 Moran TH, Katz LF, Plata-Salaman CR, Schwartz GJ.
necessary but not sufficient alone for the development of
Disordered food intake and obesity in rats lacking chole-
type 2 diabetes. Inheritance is the more important factor. cystokinin A receptors. Am J Physiol 1998; 274: 618625.
Genetically engineered diabetic mice represent new 10 Schwartz GJ, Whitney A, Skoglund C, Castonguay TW,
tools that provide us with invaluable insights into the Moran TH. Decreased responsiveness to dietary fat in
pathogenesis, and further studies may identify potential Otsuka Long-Evans Tokushima fatty rats lacking CCK-A
targets for the development of novel therapeutic stra- receptors. Am J Physiol 1999; 277: 11441151.

314 Diabetes, Obesity and Metabolism, 7, 2005, 307317 # 2004 Blackwell Publishing Ltd
D. Chen and M.-W. Wang Rodent models for type 2 diabetes RA

11 Bi S, Moran TH. Actions of CCK in the controls of food 23 Portha B. Transmitted beta-cell dysfunction as a cause
intake and body weight: lessons from the CCK-A receptor for type 2-diabetes. Med Sci (Paris) 2003; 19: 847853.
deficient OLETF rat. Neuropeptides 2002; 36: 171181. 24 Tsuura Y, Ishida H, Okamoto Y et al. Reduced sensitivity
12 Man ZW, Zhu M, Noma Y et al. Impaired beta-cell func- of dihydroxyacetone on ATP-sensitive K channels of
tion and deposition of fat droplets in the pancreas as a pancreatic beta cells in GK rats. Diabetologia 1994; 37:
consequence of hypertriglyceridemia in OLETF rat, a 10821087.
model of spontaneous NIDDM. Diabetes 1997; 46: 25 Zaitsev S, Efanova I, Ostenson CG, Efendic S, Berggren
17181724. PO. Delayed Ca2 response to glucose in diabetic GK rat.
13 Sato T, Man ZW, Toide K, Asahi Y. Plasma membrane Biochem Biophys Res Commun 1997; 239: 129133.
content of insulin-regulated glucose transporter in ske- 26 Dadke SS, Li HC, Kusari AB, Begum N, Kusari J. Elevated
letal muscle of the male Otsuka Long-Evans Tokushima expression and activity of protein-tyrosine phosphatase
Fatty rat, a model of non-insulin-dependent diabetes 1B in skeletal muscle of insulin-resistant type II diabetic
mellitus. FEBS Lett 1997; 407: 329332. Goto-Kakizaki rats. Biochem Biophys Res Commun
14 Shima K, Shi K, Sano T, Iwami T, Mizuno A, Noma Y. Is 2000; 274: 583589.
exercise training effective in preventing diabetes mellitus 27 Tourrel C, Bailbe D, Lacorne M, Meile MJ, Kergoat M,
in the Otsuka-Long-Evans-Tokushima fatty rat, a model Portha B. Persistent improvement of type 2 diabetes in
of spontaneous non-insulin-dependent diabetes mellitus? the Goto-Kakizaki rat model by expansion of the beta-cell
Metabolism 1993; 42: 971977. mass during the prediabetic period with glucagon-like
15 Shima K, Shi K, Mizuno A, Sano T, Ishida K, Noma Y. peptide-1 or exendin-4. Diabetes 2002; 51: 14431452.
Exercise training has a long-lasting effect on prevention 28 Miyamoto K, Ogura Y, Nishiwaki H et al. Evaluation of
of non-insulin-dependent diabetes mellitus in Otsuka- retinal microcirculatory alterations in the Goto-Kakizaki
Long-Evans-Tokushima Fatty rats. Metabolism 1996; 45: rat. A spontaneous model of non-insulin-dependent dia-
475480. betes. Invest Ophthalmol Vis Sci 1996; 37: 898905.
16 Ishida K, Mizuno A, Sano T, Noma Y, Shima K. Effect of 29 Agardh CD, Agardh E, Zhang H, Ostenson CG. Altered
timely insulin administration on pancreatic B-cells of endothelial/pericyte ratio in Goto-Kakizaki rat retina.
Otsuka-Long-Evans-Tokushima-Fatty (OLETF) strain J Diabetes Complications 1997; 11: 158162.
rats. An animal model of non-insulin dependent dia- 30 Sone H, Kawakami Y, Okuda Y et al. Ocular vascular
betes mellitus (NIDDM). Horm Metab Res 1995; 27: endothelial growth factor levels in diabetic rats are ele-
398402. vated before observable retinal proliferative changes.
17 Harada N, Ohnaka M, Sakamoto S, Niwa Y, Nakaya Y. Diabetologia 1997; 40: 726730.
Cilnidipine improves insulin sensitivity in the Otsuka 31 Sato N, Komatsu K, Kurumatani H. Late onset of diabetic
Long-Evans Tokushima fatty rat, a model of spontaneous nephropathy in spontaneously diabetic GK rats. Am
NIDDM. Drugs Ther 1999; 13: 519523. J Nephrol 2003; 23: 334342.
18 Kosegawa I, Katayama S, Kikuchi C et al. Metformin 32 Yoshida S, Yamashita S, Tokunaga K et al. Visceral fat
decreases blood pressure and obesity in OLETF rats via accumulation and vascular complications associated
improvement of insulin resistance. Hypertens Res 1996; with VMH lesioning of spontaneously non-insulin-
19: 3741. dependent diabetic GK rat. Int J Obes Relat Metab Disord
19 Yamamoto M, Jia DM, Fukumitsu KI et al. Metabolic 1996; 20: 909916.
abnormalities in the genetically obese and diabetic Otsuka 33 Nishida M, Miyagawa JI, Tokunaga K et al. Early morpho-
Long-Evans Tokushima Fatty rat can be prevented and logic changes of atherosclerosis induced by ventromedial
reversed by alpha-glucosidase inhibitor. Metabolism hypothalamic lesion in the spontaneously diabetic Goto-
1999; 48: 347354. Kakizaki rat. J Lab Clin Med 1997; 129: 200207.
20 Yoshida Y, Ichikawa M, Ohta M et al. A peroxisome 34 Coleman DL. Lessons from studies with genetic forms of
proliferator-activated receptor gamma agonist influ- diabetes in the mouse. Metabolism 1980; 32: 162164.
enced daily profile of energy expenditure in genetically 35 Kodama H, Fujita M, Yamazaki M, Yamaguchi I. The pos-
obese diabetic rats. Jpn J Pharmacol 2002; 88: 279284. sible role of age-related increase in the plasma glucagon/
21 Mizuno A, Kuwajima M, Ishida K et al. Extrapancreatic insulin ratio in the enhanced hepatic gluconeogenesis and
action of truncated glucagon-like peptide-I in Otsuka hyperglycemia in genetically diabetic (C57BL/KsJ-db/db)
Long-Evans Tokushima Fatty rats, an animal model for mice. Jpn J Pharmacol 1994; 66: 281287.
non-insulin-dependent diabetes mellitus. Metabolism 36 Rolin B, Larsen MO, Gotfredsen CF et al. The long-acting
1997; 46: 745749. GLP-1 derivative NN2211 ameliorates glycemia and
22 Suzuki N, Aizawa T, Asanuma N et al. An early insulin increases beta-cell mass in diabetic mice. Am J Physiol
intervention accelerates pancreatic beta-cell dysfunction Endocrinol Metab 2002; 283: E745E752.
in young Goto-Kakizaki rats, a model of naturally occur- 37 Thorkildsen C, Neve S, Larsen BD, Meier E, Petersen JS.
ring noninsulin-dependent diabetes. Endocrinology Glucagon-like peptide 1 receptor agonist ZP10A
1997; 138: 11061110. increases insulin mRNA expression and prevents

# 2004 Blackwell Publishing Ltd Diabetes, Obesity and Metabolism, 7, 2005, 307317 315
RA Rodent models for type 2 diabetes D. Chen and M.-W. Wang

diabetic progression in db/db mice. J Pharmacol Exp 52 Unger RH, Orci L. Diseases of liporegulation: new per-
Ther 2003; 307: 490496. spective on obesity and related disorders. FASEB J 2001;
38 Yajima K, Hirose H, Fujita H et al. Combination therapy 15: 312321.
with PPARgamma and PPARalpha agonists increases 53 Sreenan S, Sturis J, Pugh W, Burant CF, Polonsky KS.
glucose-stimulated insulin secretion in db/db mice. Am Prevention of hyperglycemia in the Zucker diabetic fatty
J Physiol Endocrinol Metab 2003; 284: E966E971. rat by treatment with metformin or troglitazone. Am J
39 Combs TP, Wagner JA, Berger J et al. Induction of Physiol 1996; 271: E742E747.
adipocyte complement-related protein of 30 kilodaltons 54 Finegood DT, McArthur MD, Kojwang D et al. Beta-cell
by PPARgamma agonists: a potential mechanism mass dynamics in Zucker diabetic fatty rats. Rosiglita-
of insulin sensitization. Endocrinology 2002; 143: zone prevents the rise in net cell death. Diabetes 2001;
9981007. 50: 10211029.
40 Zinker BA, Rondinone CM, Trevillyan JM et al. PTP1B 55 Shen HQ, Roth MD, Peterson RG. The effect of glucose
antisense oligonucleotide lowers PTP1B protein, nor- and glucagon-like peptide-1 stimulation on insulin
malizes blood glucose, and improves insulin sensitivity release in the perfused pancreas in a non-insulin-
in diabetic mice. Proc Natl Acad Sci USA 2002; 99: dependent diabetes mellitus animal model. Metabolism
1135711362. 1998; 47: 10421047.
41 Malamas MS, Sredy J, Gunawan I et al. New azolidine- 56 Sreenan SK, Mittal AA, Dralyuk F, Pugh WL, Polonsky KS,
diones as inhibitors of protein tyrosine phosphatase 1B Roe MW. Glucagon-like peptide-1 stimulates insulin
with antihyperglycemic properties. J Med Chem 2000; secretion by a Ca2-independent mechanism in Zucker
43: 9951010. diabetic fatty rat islets of Langerhans. Metabolism 2000;
42 Feliers D, Duraisamy S, Faulkner JL et al. Activation of 49: 15791587.
renal signaling pathways in db/db mice with type 2 57 Hargrove DM, Nardone NA, Persson LM, Parker JC,
diabetes. Kidney Int 2001; 60 (2): 495504. Stevenson RW. Glucose-dependent action of glucagon-
43 Etgen GJ, Oldham BA. Profiling of Zucker diabetic fatty like peptide-1(737) in vivo during short- or long-term
rats in their progression to the overt diabetic state. administration. Metabolism 1995; 44: 12311237.
Metabolism 2000; 49: 684688. 58 Sudre B, Broqua P, White RB et al. Chronic inhibition of
44 Peterson RG, Neel MA, Little LA, Kincaid JC, Eichberg J. circulating dipeptidyl peptidase IV by FE 999011 delays
Zucker diabetic fatty as a model for non-insulin-dependent the occurrence of diabetes in male zucker diabetic fatty
diabetes mellitus. ILAR News 1990; 32: 1619. rats. Diabetes 2002; 51: 14611469.
45 Slieker LJ, Sundell KL, Heath WF et al. Glucose trans- 59 Liu X, Perusse F, Bukowiecki LJ. Mechanisms of
porter levels in tissues of spontaneously diabetic Zucker the antidiabetic effects of the beta 3-adrenergic agonist
fa/fa rat (ZDF/drt) and viable yellow mouse (Avy/a). CL-316243 in obese Zucker-ZDF rats. Am J Physiol 1998;
Diabetes 1992; 41: 187193. 274: R1212R1219.
46 Griffen SC, Wang J, German MS. A genetic defect in beta- 60 Coleman DL. Obese and diabetes: two mutant genes
cell gene expression segregates independently from the cauding diabetesobesity syndromes in mice. Diabetes
fa locus in the ZDF rat. Diabetes 2001; 50: 6368. 1982; 31 (Suppl. 1): 16.
47 Tokuyama Y, Sturis J, DePaoli AM et al. Evolution of 61 Erickson JC, Hollopeter G, Palmiter RD. Attenuation of
beta-cell dysfunction in the male Zucker diabetic fatty the obesity syndrome of ob/ob mice by the loss of neuro-
rat. Diabetes 1995; 44: 14471457. peptide Y. Science 1996; 274: 17041707.
48 Harmon JS, Gleason CE, Tanaka Y, Oseid EA, Hunter- 62 Sainsbury A, Schwarzer C, Couzens M, Herzog H. Y2
Berger KK, Robertson RP. In vivo prevention of receptor deletion attenuates the type 2 diabetic syn-
hyperglycemia also prevents glucotoxic effects on drome of ob/ob mice. Diabetes 2002; 51: 34203427.
PDX-1 and insulin gene expression. Diabetes 1999; 48: 63 Bartels ED, Lauritsen M, Nielsen LB. Hepatic expression
19952000. of microsomal triglyceride transfer protein and in vivo
49 Lee Y, Hirose H, Zhou YT, Esser V, McGarry JD, Unger RH. secretion of triglyceride-rich lipoproteins are increased
Increased lipogenic capacity of the islets of obese rats: a in obese diabetic mice. Diabetes 2002; 51: 12331239.
role in the pathogenesis of NIDDM. Diabetes 1997; 46: 64 Collins S, Daniel KW, Rohlfs EM, Ramkumar V, Taylor IL,
408413. Gettys TW. Impaired expression and functional activity of
50 Hirose H, Lee YH, Inman LR, Nagasawa Y, Johnson JH, the beta 3- and beta 1-adrenergic receptors in adipose
Unger RH. Defective fatty acid-mediated beta-cell com- tissue of congenitally obese (C57BL/6J ob/ob) mice. Mol
pensation in Zucker diabetic fatty rats. Pathogenic Endocrinol 1994; 8: 518527.
implications for obesity-dependent diabetes. J Biol Chem 65 Cullinan CA, Brady EJ, Saperstein R, Leibowitz MD.
1996; 271: 56335637. Glucose-dependent alterations of intracellular free
51 Shimabukuro M, Ohneda M, Lee Y, Unger RH. Role of calcium by glucagon-like peptide-1(736) amide in indi-
nitric oxide in obesity-induced beta cell disease. J Clin vidual ob/ob mouse beta-cells. Cell Calcium 1994; 15:
Invest 1997; 100: 290295. 391400.

316 Diabetes, Obesity and Metabolism, 7, 2005, 307317 # 2004 Blackwell Publishing Ltd
D. Chen and M.-W. Wang Rodent models for type 2 diabetes RA

66 OHarte FP, Mooney MH, Kelly CM, McKillop 72 Baudry A, Leroux L, Jackerott M, Joshi RL. Genetic
AM, Flatt PR. Degradation and glycemic effects of His manipulation of insulin signaling, action and secretion
(7)-glucitol glucagon-like peptide-1(736) amide in ob- in mice. EMBO Reports 2002; 4: 323328.
ese diabetic ob/ob mice. Regul Pept 2001; 96: 95104. 73 Permutt MA, Kakita K, Malinas P et al. An in vivo
67 Young AA, Gedulin BR, Bhavsar S et al. Glucose- analysis of pancreatic protein and insulin biosynthesis
lowering and insulin-sensitizing actions of exendin-4: in a rat model for non-insulin-dependent diabetes. J Clin
studies in obese diabetic (ob/ob, db/db) mice, diabetic Invest 1984; 73: 13441350.
fatty Zucker rats, and diabetic rhesus monkeys (Macaca 74 Giddings SJ, Orland MJ, Weir GC, Bonner-Weir S,
mulatta) Diabetes 1999; 48: 10261034. Permutt MA. Impaired insulin biosynthetic capacity in
68 Gault VA, OHarte FP, Harriott P et al. Effects of the a rat model for non-insulin-dependent diabetes. Studies
novel (Pro3) GIP antagonist and with dexamethasone. Diabetes 1985; 34: 235240.
exendin (939) amide on GIP- and GLP-1-induced cyclic 75 Portha B, Blondel O, Serradas P et al. The rat models of
AMP generation, insulin secretion and postprandial in- non-insulin dependent diabetes induced by neonatal
sulin release in obese diabetic (ob/ob) mice: evidence streptozotocin. Diabetes Metab 1989; 15: 6175.
that GIP is the major physiological incretin. Diabetologia 76 Iwase M. A new animal model of non-insulin-dependent
2003; 46: 222230. diabetes mellitus with hypertension: neonatal strepto-
69 Rahimian R, Masih-Khan E, Lo M, van Breemen C, zotocin treatment in spontaneously hypertensive rats.
McManus BM, Dube GP. Hepatic over-expression of Fukuoka Igaku Zasshi 1991; 82: 415427.
peroxisome proliferator activated receptor gamma2 in 77 Van Zwieten PA, Kam KL, Pijl AJ, Hendriks
the ob/ob mouse model of non-insulin dependent MG, Beenen OH, Pfaffendorf M. Hypertensive diabetic
diabetes mellitus. Mol Cell Biochem 2001; 224: 2937. rats in pharmacological studies. Pharmacol Res 1996;
70 Way JM, Gorgun CZ, Tong Q et al. Adipose tissue resistin 33: 95105.
expression is severely suppressed in obesity and stimu- 78 Luo J, Quan J, Tsai J et al. Nongenetic mouse models of
lated by peroxisome proliferator-activated receptor non-insulin-dependent diabetes mellitus. Metabolism
gamma agonists. J Biol Chem 2001; 276: 2565125653. 1998; 47: 663668.
71 Gum RJ, Gaede LL, Koterski SL et al. Reduction of protein 79 Reed MJ, Meszaros K, Entes LJ et al. A new rat model of
tyrosine phosphatase 1B increases insulin-dependent type 2 diabetes: the fat-fed, streptozotocin-treated rat.
signaling in ob/ob mice. Diabetes 2003; 52: 2128. Metabolism 2000; 49: 13901394.

# 2004 Blackwell Publishing Ltd Diabetes, Obesity and Metabolism, 7, 2005, 307317 317

También podría gustarte