You are on page 1of 29

Received: 30 March 2020 Revised: 12 May 2020 Accepted: 28 May 2020

DOI: 10.1111/obr.13081

OBESITY COMORBIDITIES/ANIMAL MODELS

Diet-induced rodent models of obesity-related metabolic


disorders—A guide to a translational perspective

Inês Preguiça1,2,3 | André Alves1,2,3 | Sara Nunes1,2,3 |


1,2,3 1,2,3,4,5
Rosa Fernandes | Pedro Gomes | Sofia D. Viana1,2,3,6 |
Flávio Reis1,2,3

1
Institute of Pharmacology and Experimental
Therapeutics, and Coimbra Institute for Summary
Clinical and Biomedical Research (iCBR), Diet is a critical element determining human health and diseases, and unbalanced
Faculty of Medicine, University of Coimbra,
Coimbra, Portugal food habits are major risk factors for the development of obesity and related meta-
2
Center for Innovative Biomedicine and bolic disorders. Despite technological and pharmacological advances, as well as inten-
Biotechnology (CIBB), University of Coimbra,
sification of awareness campaigns, the prevalence of metabolic disorders worldwide
Coimbra, Portugal
3
Clinical Academic Center of Coimbra (CACC), is still increasing. Thus, novel therapeutic approaches with increased efficacy are
University of Coimbra, Coimbra, Portugal urgently required, which often depends on cellular and molecular investigations using
4
Department of Biomedicine, Faculty of
robust animal models. In the absence of perfect rodent models, those induced by
Medicine, University of Porto, Porto, Portugal
5
Center for Health Technology and Services
excessive consumption of fat and sugars better replicate the key aspects that are the
Research (CINTESIS), University of Porto, root causes of human metabolic diseases. However, the results obtained using these
Porto, Portugal
6
models cannot be directly compared, particularly because of the use of different
ESTESC-Coimbra Health School, Pharmacy,
Polytechnic Institute of Coimbra, Coimbra, dietary protocols, and animal species and strains, among other confounding factors.
Portugal This review article revisits diet-induced models of obesity and related metabolic
Correspondence disorders, namely, metabolic syndrome, prediabetes, diabetes and nonalcoholic fatty
Flávio Reis, PhD, Faculty of Medicine, University liver disease. A critical analysis focused on the main pathophysiological features
of Coimbra, 3000-548 Coimbra, Portugal.
Email: freis@fmed.uc.pt of rodent models, as opposed to the criteria defined for humans, is provided as a
practical guide with a translational perspective for the establishment of animal
Funding information
This work was supported by European models of obesity-related metabolic diseases.
Regional Development Fund (FEDER), through
Programa Operacional Factores de KEYWORDS
Competitividade COMPETE2020 (CENTRO-
diabetes, diet-induced rodent models, metabolic syndrome, nonalcoholic fatty liver disease,
01-0145-FEDER-000012-HealthyAging2020,
POCI-01-0145-FEDER-007440 and POCI- obesity, prediabetes
01-0145-FEDER-031712) and by National
funds via Portuguese Science and Technology
Foundation (FCT): Strategic Projects UID/
NEU/04539/2013, UID/NEU/04539/2019,
UIDB/04539/2020 and UIDP/04539/2020
(CIBB); PhD Fellowship SFRH/BD/109017/
2015; PTDC/SAU-NUT/31712/2017.

ABBREVIATIONS: ADA, American Diabetes Association; BMI, body mass index; BP, blood pressure; BW, body weight; CD, cafeteria diet; CDAA, choline-deficient L-amino acid; CHO,
carbohydrates; CVD, cardiovascular disease; DIO, diet-induced obesity; FBG, fasting blood glucose; FFA, free fatty acids; IFG, impaired fasting glucose; IGT, impaired glucose tolerance; ITT,
insulin tolerance test; HbA1c, glycated haemoglobin; HCC, hepatocellular carcinoma; HDL, high-density lipoprotein; HFD, high-fat diet; HFHSD, high-fat high-sugar diet; HSD, high-sugar diet;
HFHFD, high-fat and high-fructose diet; HOMA-IR, homeostatic model assessment of insulin resistance; MCD, methionine-choline-deficient; MetS, metabolic syndrome; NAFLD, nonalcoholic
fatty liver disease; NASH, nonalcoholic steatohepatitis; OGTT, oral glucose tolerance test; QUICKI, quantitative insulin check index of insulin sensitivity; STZ, streptozotocin; T2DM, type
2 diabetes mellitus; TGs, triglycerides; TWD, total Western diet; VLDL, very low-density lipoprotein; WAT, white adipose tissue; WC, waist circumference; WD, Western diets; WHO, World
Health Organization.

Inês Preguiça and André Alves contributed equally.

Obesity Reviews. 2020;1–29. wileyonlinelibrary.com/journal/obr © 2020 World Obesity Federation 1


2 PREGUIÇA ET AL.

1 | I N T RO D UC TI O N body weight (BW) changes, obesity strongly exacerbates common


pathophysiological traits involving insulin resistance, hyperglycaemia
1.1 | Overview of the problem and challenges and dyslipidaemia.22–24 For instance, excess circulating free fatty acids
(FFA) arising from enlarged adipose tissue may compete with glucose
Globally, obesity has reached epidemic proportions, affecting not as the substrate in peripheral tissues, fuelling insulin resistance. In
only adults but also an increasing number of children and adoles- addition to impaired glucose uptake by these tissues, increased circu-
cents.1
Furthermore, as obesity can progressively cause and/or lating FFA may also impair pancreatic β-cell function and insulin secre-
exacerbate a broad spectrum of chronic diseases, including metabolic tion.22,24,25 In fact, the transition from prediabetes to overt diabetes
syndrome (MetS), prediabetes, type 2 diabetes mellitus (T2DM), non- relies mostly on the loss of a long period of increased hyper-
alcoholic fatty liver disease (NAFLD), cardiovascular disease (CVD) insulinaemia to compensate for insulin resistance.24,26 At the end
2
and some types of cancer, it poses an increased risk for all-cause stage, β-cell failure and decreased insulin secretion further aggravate
mortality.3,4 abnormal hyperglycaemia.27 Similarly, a vicious cycle arising from
Although lifestyle changes, such as calorie restriction and regular glucotoxicity and lipotoxicity pathways underlies MetS and NAFLD
physical activity/exercise, are effective in reducing obesity and associ- pathogenesis. Overall, oxidative imbalance, low-grade inflammation,
5–7
ated complications, the majority of the population may not adhere plasmatic and/or hepatic dyslipidaemia, impaired incretin effect, gut
to these interventions. In addition, despite decades of extensive microbiota changes, endoplasmic reticulum stress and/or mitochon-
research, only few molecules progressed successfully into approved drial dysfunction are common cellular denominators of obesity-related
drugs for weight management in patients with obesity.8,9 Thus, metabolic diseases.28–31
approaches for overcoming obesity-related metabolic disorders and Similar to that in humans, chronic consumption of diets rich in fat
complications remain an open challenge. and sugar leads to deleterious metabolic effects in rodents, despite
Various animal models, mostly involving rodents, have been used differing responsiveness among species and strains. Thus, paradigms
for several decades as fundamental tools for studying obesity, T2DM of longstanding free access to palatable calorie-dense food are widely
10–14
and associated metabolic disorders. However, unlike human used in preclinical research to model the progression of obesity and
diseases, the diagnostic criteria have not been clearly established for related comorbidities.10,32 One approach for studying the develop-
animal models of human diseases, and the research community is ment of obesity in rodents involves the feeding of cafeteria diet (CD).
aware of their considerable drawbacks and limited predictability.15 This method, created to closely emulate human dietary patterns,
The absence of rigorous, standardized and clinically relevant criteria allows animals to freely select from a variety of tasty processed foods
continues to hamper the translation of preclinical metabolic research (e.g., cookies, cake and salami) mimicking the hedonic hyperphagia
findings into drug discovery programs regarding obesity and related frequently observed in humans with obesity.10,33 Although the cafete-
metabolic disturbances and preclude biological understanding of ria paradigm promotes pronounced weight gain and severe symptom-
the disease. atology of obesity-related metabolic diseases, the poor monitoring of
Therefore, in this review, we have highlighted the main fea- rodents' energy/nutrients/fibre intake in these regimens has hindered
tures of dietary rodent models, alone or in combination with its reliability.34
chemical or genetic manipulations, which can be used to experi- To ensure experimental reproducibility and improve translatabil-
mentally replicate human obesity and associated complications, ity, standardized laboratory animal diets formulated with purified raw
specifically MetS, prediabetes, T2DM and NAFLD. We further pro- materials and accurately reflecting the macronutrient and micronutri-
vide an in-depth comparison of the main variables for the diagnosis ent content are being used. The most frequently used are high-fat
of diseases in humans and characterization of animal models. Reso- diet (HFD), high-sugar diet (HSD) and the respective combinations of
lution of the prevailing discrepancies is expected to improve the high-fat high-sugar diet (HFHSD), collectively termed “Western diets”
reliability of preclinical studies and thereby improve their transla- (WD).35
tion to the clinic. Typically, HFD (45% and 60% calories from fat) consistently
induces obesity, insulin resistance, hypertriglyceridaemia, liver
steatosis, hypertension, adipocyte hypertrophy and impairment of the
1.2 | Diet as a driving force for the continuum of intestinal barrier/metabolic endotoxaemia; the observed variations
obesity-related metabolic disorders are largely due to the fat used (percentage and source: animal
vs. vegetable), the duration of the protocol and the rodent
Although genetic background plays a pivotal role in the establishment species.36–41 HSD are the most diverse, as they can be administered
of obesity, lifestyle features such as physical inactivity and chronic on their own or as a supplement to HFD. Owing to their palatability
hypercaloric dietary patterns are currently envisaged as the main and high energy density, sugar-based diets also promote positive
drivers behind the exponential increase in obesity-related metabolic energy balance.10 Sucrose, glucose and fructose are the most preva-
16–18
diseases. Several risk factors and pathophysiological pathways of lent sugar sources, although they differ considerably in terms of meta-
MetS, prediabetes, T2DM and NAFLD are overlapping.19–21 Apart bolic disruption.42 For instance, ad libitum access to fructose in
from few cases where the aforesaid disorders evolve without major beverages increases hepatic lipid accumulation and adiposity in mice
PREGUIÇA ET AL. 3

compared with a similar sucrose paradigm.43 Yet, HSD in rodents gen- 2 | D I E T - I N DU C E D R O D E N T M O D E LS


erally affects BW gain and adiposity lesser than HFD paradigms,
irrespective of their equally negative impact on glycaemic control.10 2.1 | Obesity
It is important to highlight that most of the available diets do not
closely recapitulate Western human dietary patterns, although they Obesity is defined by the World Health Organization (WHO) as
can replicate some of the metabolic diseases associated with this abnormal or excessive fat accumulation that may impair health and is
44
human dietary regimen in rodents. For instance, humans typically considered a chronic relapsing progressive disease process by the
consume 30% of their calories from fat, which is significantly lower World Obesity Federation.47,48 The worldwide prevalence of obesity
than the fat content observed in most of the commercially available has nearly tripled from 1975 to 2016, affecting 13% of the world's
standardized rodent HFD.10 Therefore, experimental protocols with adult population.49
45% calories from fat are more relevant for translational purposes. In humans, anthropometric measures, particularly body mass
However, important preclinical evidence has been obtained from diets index (BMI), calculated as weight divided by the square of the height
where 60% calories were obtained from fat, probably due to an accel- (kg/m2), and waist circumference (WC), have been used to define indi-
erated disease phenotype and inherent economic perks.35 viduals with overweight or obesity. In addition, body fat distribution,
Another important feature relies on the negligible resemblance in besides the total amount of body fat, is an important risk factor for
terms of micronutrients and additive content of human WD pattern obesity-related disorders.50 In particular, excess visceral adipose tis-
with the corresponding rodent's CD and WD regimens. To overcome sue, that is, the accumulation of adipose tissue within the omentum
these gaps, experimental diets aimed to better reproduce the basic and abdomen and around abdominal organs, is more strongly associ-
features of Western nutrition have been recently developed. The total ated with the development and progression of cardiometabolic dis-
Western diet (TWD), reflecting the median nutrient intake among the eases in humans than subcutaneous adiposity.51 Unfortunately, in
population of the United States, displays energy density similar to cer- clinical practice, as with the routine assessment of WC, body fat distri-
tain commercial diet-induced obesity (DIO) formulations. However, bution is rarely evaluated,52,53 irrespective of the methodologies
TWD was developed to achieve more diversity in terms of fat sources available,54–57 although the majority of the methods are laborious and
and balanced micronutrient composition, as well as increase in com- require expensive technology.
plex carbohydrate (CHO) and simple sugar content. Surprisingly, Unlike in humans, obesity has not been defined in experimental
TWD-fed mice did not show increase in energy intake, fat mass and animals. Obesity is usually considered as any significant increase in
BW and failed to acquire an MetS phenotype.45 Hence, dietary micro- the levels of a marker relative to that in control animals (Figure 1).
nutrients may play a major role in the hyperphagic response, increas- Several complementary markers have been used to assess obesity in
ing weight gain in mice.45 Another attempt to more realistically rodents, including body composition analysis, condition index (which
replicate the human WD pattern in rodents involves the use of a new can be evaluated from the BMI using the Quételet's index, for exam-
prototypic animal WD formulated with high amounts of simple CHO, ple), total BW gain, weight of adipose tissue depots and adipocyte
salt and fat (42.5% kcal from fat), along with a low-fibre percentage.46 size.10,58 Imaging techniques for assessing adipose tissue distribution
This novel dietary paradigm is additive free, nutritionally adequate and are also available for laboratory animals.10 The susceptibility of ani-
highly reproducible, as it overcomes food heterogeneity issues, unlike mals to obesity development is highly heterogeneous, being mostly
46
the CD regimens. Interestingly, this standardized WD approach dependent on strain, gender, age and diet composition. Therefore, we
effectively induced excess body fat accumulation as well as obesity- reasoned that the establishment of comprehensive and definitive
related features (e.g., hepatic steatosis and fasting hyperglycaemia), guidelines for obesity assessment in experimental animals will contrib-
which were independent of gut microbiota dysbiosis.46 Remarkably, ute to advancements in the field. Although the goal of this review is
WD-fed rats gained significantly more weight and showed higher adi- obviously not to determine the criteria for defining obesity in rodents,
posity index than rats fed two traditional obesogenic regimens, the data that demonstrate the diversity of parameters used in different
CD and the HFD, obtaining 42.5% and 60% kcal from fat, respec- studies, as well as the wide range of observed results, might be poten-
tively.46 Considering the same micronutrient base of the aforemen- tially useful. Above all, we intend to stimulate a more in-depth discus-
tioned dietary paradigms, this elegant work pinpoints the importance sion on this subject.
of diet choice when an obesogenic paradigm is to be selected. Several animal models, particularly rodents, have been used to
In sum, subtle changes in rodent dietary paradigms (e.g., nutrient replicate the phenotype and pathogenesis of human obesity.10,13,14,59
sources, diet and physical form) may considerably affect the extent of The most commonly used obesity models can be subdivided into
metabolic impairments. Hence, appropriate hypercaloric diets should those based on mutations or manipulations of one or a few individual
be carefully selected (and the corresponding control diets, as dis- genes (e.g., ob/ob mouse or Zucker fatty rat [fa/fa]), or those resulting
cussed later) to successfully achieve most of the desired experimental from exposure to obesogenic environments.10,59 Despite being a com-
outcomes. The following sections provide an in-depth analysis of the plex and multifactorial disorder, excess calorie intake and a sedentary
most common dietary paradigms used to experimentally replicate the lifestyle are recognized as the main drivers of obesity. Therefore, DIO
phenotypic features comprising obesity, MetS, prediabetes, T2DM models have gained increased relevance, as they mimic the process of
and NAFLD in rodents. obesity development in humans more closely than genetic models.
4 PREGUIÇA ET AL.

F I G U R E 1 Comparison of human
diagnosis criteria for obesity,
metabolic syndrome, prediabetes,
diabetes and nonalcoholic fatty liver
disease with markers used in
preclinical research to define rodent
models of those conditions. ALT,
alanine aminotransferase; AST,
aspartate transaminase; BMI, body
mass index; FBG, fasting blood
glucose; GTT, glucose tolerance test;
HbA1c, glycated hemoglobin; HDL,
high-density lipoprotein; HOMA-IR,
homeostatic model assessment of
insulin resistance; IFG, impaired
fasting glucose; IGT, impaired glucose
tolerance; IR, insulin resistance; ITT,
insulin tolerance test; OGTT, oral
glucose tolerance test; SBP, systolic
blood pressure; TGs, triglycerides

The exposure of rodents to energy-dense diets enriched in fats such as mice. A study using a protocol of HFD for 8 weeks in C57BL6
often results in obesity and related complications, even with differ- was associated with 16% increase in BW,77 whereas another study
ences in terms of dietary composition and duration of intervention. performed using the same mouse strain but with 5 weeks of HFD
The development of obesity in this model is thought to result from induced 37% increase in BW.78 On the contrary, AKR/J and 129S1/
various interrelated factors, although excess calorie intake is a major SvlmJ mice under 10 and 20 weeks of HFD, respectively, showed
contributor. Some strains of rats and mice are prone to the develop- 35% and 37% BW gain, although information regarding adipose tissue
ment of DIO. For example, Sprague–Dawley and Wistar rats fed a diet alterations were lacking.79
containing 40–60% energy as fat normally take 8–12 weeks to Supported by strong epidemiological and experimental evidence
develop obesity.41,60,61 Similarly, several inbred mouse strains, notably associating increased sugar consumption to the obesity epidemic,80,81
C57BL/6J, FVB/N and DBA/2J, are also susceptible to HFD-induced diets with high sucrose or high fructose content have also been used
62–67
obesity. Interestingly, a small fraction of rodents are resistant to as parts of the nutritional strategy for obesity induction in rodents.
DIO, although the underlying mechanisms are currently not For example, male NMRI outbred mice with free access to fructose-
completely understood.68,69 However, critical information has been sweetened water for 2.5 months exhibited increased BW and adipos-
obtained using obesogenic diets for the selection of obesity-prone ity compared with control animals.43 Similarly, both male and female
(OP) or obesity-resistant (OR) or DIO/diet-resistant (DR) rodents Sprague–Dawley rats maintained for 6–7 months with access to fruc-
using outbred and/or inbred strains,68,70,71 as will be mentioned tose water gained significantly more weight than control animals.82
throughout the manuscript. However, compared with that observed on HFD, obesity marker
Regarding HFD-induced obesity, studies in Sprague–Dawley rats levels in male C57Bl/6 mice on HSD for 3 or 12 months are typically
with 4 weeks of feeding showed distinct percentage of BW gain (cal- less exacerbated and more variable, although with a similar negative
culated by discounting the change in BW of control animals) and adi- impact on glucose homeostasis.83,84 Several studies have also
72
pose tissue rise: 25% BW and 13% white adipose tissue (WAT) assessed the potential effect of the interaction between excess fat
versus 33% BW and 6.3% fat mass.73 Curiously, two other studies on and sugar in diets on metabolic risk factors. Indeed, compared with
74
Wistar rats showed highly divergent percentage of BW gain (22% HFD alone, feeding male C57Bl/6 mice and Wistar rats with HFHSD
and 50%75), without information regarding changes on adipose tissue for 4 and 8 weeks, respectively, resulted in rapid onset and more
depots. The former study, however, was performed with young ani- severe metabolic phenotype,85,86 including MetS, prediabetes, T2DM
mals, which might explain the differences. For longer protocols and NAFLD, which will be discussed in the following sections.
(10 weeks), another study showed major differences between Wistar Overall, these studies clearly demonstrated the diversity of out-
and Sprague–Dawley rats regarding both the percentage of BW gain comes in models of obesity induced by hypercaloric diets, strongly
(22% vs. 14%) and percentage of fat mass increase (43% vs. 21%).60 recommending a standardization of criteria, markers and cut-offs for
An even longer protocol of 25 weeks of HFD on Sprague–Dawley rats better comparison of results. This aspect should not be confused with
76
reported 32% BW gain, which is almost identical to that achieved the existence of diverse models that does not constitute a problem
(33%) by other studies with only 4 weeks under HFD.73 Disparity per se. On the contrary, it allows higher representation of heteroge-
among research findings was also observed in other rodent species, neous human diseases.
PREGUIÇA ET AL. 5

2.2 | Metabolic syndrome the duration of administration (from 4 weeks up to 1 year).102 How-
ever, the disease phenotypes induced by these species vary. In mice,
MetS, originally referred to as syndrome X, is roughly characterized HFD usually results in an MetS phenotype characterized by obesity,
by abdominal obesity, insulin resistance, hypertension and hyper- dyslipidaemia, hypertension and glucose intolerance, which could be
lipidaemia.87 MetS is not simply a disease, but a screening tool, due to species-specific susceptibility to dietary fat.79,80,103 Further-
where a complex cluster of metabolic abnormalities collectively pre- more, age has been shown to be an important factor modulating sus-
dict the risk of individuals to develop CVD, T2DM and NAFLD, as ceptibility to diet-induced MetS.104 In fact, compared with young
87,88
well as kidney and pancreatic disorders. In fact, the International mice, old C57BL/6 mice appear to be more susceptible to HFD,
Diabetes Federation (IDF) for Global Consensus Definition, in coop- resulting in an aggravated phenotype.105 In rats, different outcomes
eration with the American Heart Association/National Heart, Lung, were observed according to the age at which the animals were first
and Blood Institute, specified in 2009 that MetS is achieved when exposed to HFD. In fact, Sprague–Dawley rats exposed to HFD at
three out of the following five parameters are present89: (i) obesity, 3 weeks of age for 8 weeks developed obesity, impaired FBG,
with sex- and ethnicity/race-specific WC cut-offs; (ii) elevated levels dyslipidaemia and hypertension; however, when rats aged 8 weeks
of triglycerides (TGs): ≥150 mg/dl; (iii) reduced high-density lipopro- were fed HFD, they only developed obesity and hypertension.61
tein (HDL) cholesterol: <40 mg/dl in men and <50 mg/dl in women; Nonetheless, in Wistar rats, 8 weeks of HFD with 50% kcal from fat
(iv) elevated blood pressure (BP): systolic BP ≥ 130 mm Hg or dia- were sufficient to induce components of MetS, such as obesity,
stolic BP ≥ 85 mm Hg; (v) elevated fasting blood glucose (FBG): dyslipidaemia, impaired glucose tolerance (IGT) and hepatic
≥100 mg/dl (Figure 1). steatosis.106
The global prevalence of MetS is unknown; however, as it is Interestingly, HSD induces the same MetS phenotype in both
about three times more common than diabetes, it can be estimated to mice and rats, although most studies have been performed in the lat-
affect approximately one quarter of the world population.90,91 The ter.107 Obesity often does not develop under this condition, although
prevalence varies considerably between distinct world regions, with a different sources of sugar (sucrose vs. glucose vs. fructose) are added
92
variation of 29–40% across different states of the United States, for to the diet.42,108 However, the absence of differences in BW does not
92,93
example, and between sex. Despite the heterogeneity of epidemi- indicate identical body composition, as HSD-fed animals generally
ological studies with distinct populations, MetS prevalence appears to show a significantly higher percentage of epididymal fat weight than
consistently increase with age, and hypertension appears to be the the controls.109–111 Furthermore, under this diet, both species
90,91
most prevalent underlying feature of MetS across countries. showed clear dyslipidaemia (hypertriglyceridaemia and hyper-
Although some genetic differences may account for distinct preva- cholesterolaemia), accompanied by increased glucose intolerance and
lence of MetS among distinct communities/ethnicities,94,95 the insulin resistance, while presenting merely “mild” hypertension or
increased consumption of high-fat low-fibre meals and sedentary life- even the absence of BP elevation.112
96–98
style appear to be the key factors. Finally, the combination of high-fat and high-fructose diet
Considering these numbers, the establishment of animal models (HFHFD) is mandatory for achieving hyperglycaemia, which is another
of MetS for proper understanding of pathophysiological features and feature of MetS. In 10-week-old Wistar rats fed HFHFD, impaired
therapeutic opportunities is of utmost importance for global public fasting glucose (IFG) appears after 2 months of diet, whereas hyper-
health. Nevertheless, criteria for model validation have to be defined, glycaemia only manifested consistently after 6 months, and sustained
as otherwise, disparities in results that cannot be compared will be hyperinsulinaemia only after 8 months.74 In addition, in Wistar rats,
continuously generated. In the absence of an institutional definition fructose supplementation via HFD (22.5% kcal from fat) was key to
(by a consensus group, for example), Cheng et al. 61 suggested certain aggravation of hyperglycaemia.113 In mice, one study demonstrated
criteria to validate an experimental model of MetS, which included that the supplementation of sugar to HFD worsened the disease phe-
weight gain, increased visceral fat mass, impaired FBG, hypertension, notype via aggravation of the inflammatory state.114 Overall, HFHFD
hypertriglyceridaemia, elevated non-HDL cholesterol level and hepatic exacerbated the MetS phenotype in both species, characterized by
steatosis. Simultaneously, food and water consumption should be overt/sustained fasting hyperglycaemia, dyslipidaemia, hypertension
monitored rigorously to determine the precise caloric intake of each and obesity.
experimental group, a key factor for ensuring model validation and
scientific replication of data.
When inducing animal models of MetS, a panoply of protocols 2.3 | Prediabetes
has been used either solely via diet manipulation or in combination
with streptozotocin (STZ), a cytotoxic drug that elicits clear In conjunction with the epidemics of T2DM, the prevalence of
hyperglycaemia.99–101 Nonetheless, as previously mentioned, we have prediabetes has skyrocketed worldwide, mainly due to major changes
herein focused on diet-induced models of MetS (Table 1), which are in lifestyle associated with economic growth; in particular, altered
mainly based on high fat, high sugar or a combination of both. HFDs dietary patterns are promoters of obesity.115 Prediabetes is an
are commonly used for diet-induced metabolic abnormalities in both increasingly common asymptomatic condition associated with a high
rats and mice, inducing a wide range of fat content (30–60%) within risk for T2DM development.115,116 Prediabetes, or intermediate
6

TABLE 1 Main features of diet-induced rodent models of metabolic syndrome

Diet Duration
type Composition (%) (weeks) Specie/strain Main features Refs
High-fat based
HFD 60% kcal from fat, 20% kcal from protein, 20% kcal 13 Male C57BL/6 mouse Obesity: 74% Δ weight gain; fivefold increase in fat Roopchand et al.78
from CHO (6-week-old) mass
Increased serum TGs
Elevated FBG: 350 mg/ml
IGT
60% kcal from fat, 20% kcal from protein, 20% kcal 20 Male AKR/J mouse (6- to Obesity: 43% Δ weight gain Meng et al.79
from CHO 12-week-old) IGT
Elevated right ventricle systolic BP
49% kcal from fat, 14% kcal from proteins, 37% kcal 8 Male Wistar rats (10- to Obesity: 26% Δ weight gain; 1.5-fold increase in Lasker et al.106
from CHO 12-week-old) epididymal fat mass
Increased plasma TGs
Decreased plasma HDL
IGT
Hepatic steatosis and inflammation
60% kcal from fat, 20% kcal from protein, 20% kcal 8 Male Sprague–Dawley rats Obesity: 141% Δ weight gain; 1.5-fold increase in Cheng et al.61
from CHO (3-week-old) rWAT mass
Increased plasma TGs
Decreased plasma HDL
Fasting blood glucose: 117.5 mg/dl
Increased systolic blood pressure
Hepatic steatosis
High-sugar based
HSD 16.9% kcal in fat, 18.1% of kcal in protein 65% of kcal 12 Male C57BL/6 (8-week-old) Obesity: no BW changes, slight increase in Do et al.42
in CHO (85% from glucose or fructose and 15% epididymal WAT mass
from sucrose) Increased serum cholesterol and LDL fraction.
FBG: 175 mg/dl
IGT
Hepatic steatosis
10% kcal from fat, 18.6% kcal from protein, 60.6% 16 Male Wistar rats (6-week-old) Obesity: no BW changes Aguilera-Mendez et
kcal from CHO + 30% fructose (w/v) in water Increased serum triglycerides al.111

Decreased serum HDL


Increased systolic BP
IGT
PREGUIÇA ET AL.
PREGUIÇA ET AL.

TABLE 1 (Continued)

Diet Duration
type Composition (%) (weeks) Specie/strain Main features Refs
Hepatic steatosis

High-fat/high-sugar based
HSHFD 59% kcal from fat, 15% protein, 26% kcal from 8 Male C57BL/6 (12-week-old) Obesity: 62% Δ weight gain; threefold increase in Masi et al.114
CHO + condensed milk (23% kcal from fat, 9% kcal visceral adipose tissue
from protein, 68% kcal from CHO) Increased TGs
FBG: 214 mg/dl
Mild hepatic steatosis
60% kcal from fat, 20% from protein, 20% from 8 Male Sprague–Dawley rats Obesity: 26% Δ weight gain; 0.75-fold increase in Cheng et al.61
CHO + 30% sucrose (w/v) in water (8-week-old) rWAT mass
Elevated plasma TGs
Decreased plasma HDL
FBG: 99.7 mg/dl
Increased systolic BP
IGT
Early stage hepatic steatosis
22.5% kcal from fat, 17% kcal from protein, 42.3% 20 Male Wistar rat (9-week-old) Obese: 30% Δ weight gain; 1.84-fold increase in Moreno-Fernandez et
kcal from CHO + 25% glucose (w/v) in water epididymal mass al.113
Increased plasma TGs
Decreased HDL
FBG: 290 mg/dl
7
8 PREGUIÇA ET AL.

hyperglycaemia (the preferred terminology of WHO), defines a state and leptin levels, HOMA-IR index and glucose intolerance) were
in which the individuals have abnormal glucose homeostasis where detected earlier and were more pronounced in Wistar rats.60 A robust
blood glucose levels are above the normal but below the thresholds mouse model for IGT and early T2DM was established using female
established for T2DM diagnosis.115,117 C57BL/6J mice under HFD (58% calories from fat) for 12 months.131
The current gold standard measures used for prediabetes screen- One week after starting the diet protocol, the animals presented ele-
ing are FBG test and a 2-h plasma glucose analysis during an oral glu- vated baseline glucose and insulin levels and IGT, along with impaired
118
cose tolerance test (OGTT). Patients with prediabetes may have insulin response; over time, progressive worsening of insulin resis-
IFG and/or IGT, which are defined by the American Diabetes Associa- tance was observed in HFD-fed mice.131 Another study performed in
tion (ADA) as FBG levels of 100–125 mg/dl and 2-h glucose level of 12-week-old male C57BL/6J mice during a period of 10 weeks
140–199 mg/dl during an OGTT.119 Glycated haemoglobin (HbA1c) showed that diets rich in both saturated (60% calories from fat, con-
concentrations varying from 5.7% to 6.4% is further considered by taining lard) and monounsaturated (60% calories from fat, such as
ADA as an additional criterion for prediabetes diagnosis (Figure 1).119 from olive oil) fatty acids induced marked insulin resistance with
Whereas IFG is mainly related to hepatic insulin resistance, IGT typi- hyperinsulinaemia, accompanied by inflammation and altered distribu-
cally reflects peripheral insulin resistance and altered β-cell function. tion of adipose tissue.129
Both insulin resistance and impaired β-cell function are responsible Several experimental studies have shown the use of HSD (mainly,
for the increased risk of T2DM development in patients with high-sucrose and high-fructose) as an inexpensive nutritional strategy
prediabetes.120–122 to promote the development of insulin resistance and glucose intoler-
Unlike T2DM, options for prediabetes animal models are limited, ance in animal models; however, the results were inconsistent, appar-
and unified classification criteria for rodents are lacking, for whom the ently due to variations in the metabolic responses to diets based on
prediabetic phenotype has been defined/established according to the species/strain, duration of protocol, percentage of sugar and route of
following parameters: increased 2-h blood glucose values, glucose administration.77,132–135 In fact, a study suggested that C57BL/6 mice
intolerance, hyperinsulinaemia, hyperlipidaemia and moderate/mild are more resistant to sucrose treatment than Wistar rats.132 In that
75
hyperglycaemia (Figure 1). Some diet-induced animal models have study, male Wistar rats and C57BL/6 mice, both with 7 weeks of age,
been described to mimic the stages of prediabetes and/or insulin received 30% sucrose in drinking water during 20 weeks. The mice
resistance. Several experimental studies have shown that dietary mac- presented slight obesity and hyperlipidaemia without changes in glu-
ronutrient composition is an important environmental determinant of cose and insulin levels, whereas rats presented hypercholesterolaemia,
the quality of insulin action and a trigger for the prediabetes hyperglycaemia, hyperinsulinaemia and insulin resistance.132 Two
74,75,123
stage. Indeed, the modern WD and CD, rich in saturated fats studies using the same species and strain (male C57BL/6J mice), but
and in CHO (such as fructose and sucrose), are able to promote impor- distinct sucrose content and protocol duration, reported the absence
tant changes in CHO metabolism, causing prediabetes, although other of weight gain but diverse metabolic responses.77,84 In fact, 8 weeks
metabolic features resembling obesity and MetS may coexist. of HSD treatment (10% fat, 32% sucrose) of 3-month-old mice
Long-term HFD intake in several strains of rat and mouse can resulted in adipocyte hypertrophy, glucose intolerance, hyper-
cause hyperlipidaemia, peripheral insulin resistance, moderate hyper- insulinaemia, hyperlipidaemia, hepatic steatosis and increased secre-
glycaemia and IGT, together with increase in the expression of inflam- tion of inflammatory cytokines,77 whereas 4-week-old mice on
124,125
mation and oxidative stress markers. In rodents on HFD 55 weeks of solid food with high sucrose (50%, wt/wt) content did
feeding, insulin resistance is followed by insufficient β-cell compensa- not show changes in BW, but variable degrees of hyperglycaemia and
tion, resulting in hyperinsulinaemia and altered glucose metabo- hyperinsulinaemia.84 Furthermore, a study using male C57BL/6J mice
lism.126 The effects of HFD depend not only on the source and (8 weeks old) suggested that distinct modes of sucrose delivery differ-
percentage of calories but also on the animal species and strains entially affected body regulation and glucose homeostasis.134 The
used.127–129
A “humanized” model of prediabetes was suggested for authors showed that mice exposed to a liquid sucrose solution (50%
young male Wistar rats under HFD (45% calories from fat) for sucrose in the water plus 30% calories from sucrose in solid food)
6 months.75 The authors reported a progressive development of the presented higher adiposity, impaired glucose homeostasis, peripheral
phenotype typical of humans, starting with BW gain, gradual increase insulin sensitivity and insulin resistance than mice maintained under
of FBG and glucose intolerance, further evolving to hyperinsulinaemia equivalent levels of sucrose in solid diet (73% calories from
and insulin resistance, together with hypertrophy of pancreatic β-cells sucrose).134 Previously, we were able to reproduce a prediabetic rat
in the last month.75 Increased levels of FBG and insulin and aug- model using an oral regimen of 35% sucrose consumption for
mented homeostatic model assessment of insulin resistance (HOMA- 9 weeks.112,136 The animals were characterized to be prediabetic
IR), together with impaired β-cell function and OGTT, were also based on fasting normoglycaemia, IGT, hyperinsulinaemia and insulin
observed in Sprague–Dawley rats under HFD (46% calories from fat) resistance without obesity, which is consistent with the results of
for 12 weeks.130 As animal strains might present distinct responses to other studies;137 interestingly, the animals concomitantly displayed
HFD, the metabolic effects of HFD intake during 16 weeks were com- some early features of retinal and neuronal impairment.137–139
pared between Wistar and Sprague–Dawley rats; results showed that The use of fructose-rich diet is also a well-characterized approach
the majority of the metabolic changes (such as BW gain, fasting insulin for eliciting insulin resistance and IGT in rodents.140,141 Unlike
PREGUIÇA ET AL. 9

glucose-fed rodents, fructose-fed rodents displayed disruption of classification criteria for T2DM rodent models are lacking, probably
metabolic homeostasis, including IGT and dyslipidaemia.142,143 In one because normal and diabetic FBG differ considerably between rodent
study, low fructose (7%) content in drinking water was sufficient to species (Figure 1). It is generally assumed that the criteria used for rats
cause glucose intolerance with abnormal morphology and function of are closest to those used for humans, namely, FBG > 126 mg/dl
pancreatic islet cells in Wistar rats, but without changes in FBG, insu- (7.0 mmol/L).158,159 In contrast, the basal levels of FBG in mice are
lin and lipids levels.144 However, in another study, the same strain of higher than those in rats and humans (ranging between 140 and
rats fed with fructose-rich diet (10% w/v in drinking water) developed 220 mg/dl), indicating that according to the aforementioned criteria,
hypertriglyceridaemia and a state of insulin resistance (demonstrated all mice would develop diabetes. Hence, FBG values ≥250 mg/dl is
by hyperinsulinaemia with normoglycaemia, high insulin:glucose molar the recommended threshold for defining T2DM in mice.160,161 In addi-
ratio and HOMA-IR index) after 3 weeks of fructose tion, increased glycaemia following 2 h of OGTT frequently corrobo-
145
administration. rates the FBG values. Here, the large discrepancy in the currently
HFHSD is also commonly used to experimentally replicate predia- reported FBG cut-off values is possibly due to not only to the animal
betes.146,147 Different sources and amounts of CHO (mainly fructose strain used but also to the degree of disease severity/duration and
or sucrose) and fats have been used.148–150 In the HFHSD regimens the duration of the fasting period.162,163
administered to rodents, CHO content typically ranged between 10% Several different animal models of T2DM sharing many pheno-
and 60% (in the diet, drinking water or both), whereas fat content var- typic features with the human disease are currently available,38,154
ied between 20% and 60%.148,151 C57BL/6J mice fed HFHSD (45% which includes genetically manipulated (spontaneous or trans-
energy from fat and 20% by weight from sucrose) for 22 weeks genic/knockout), surgically treated (partial pancreatectomized animals)
showed increased BW, glucose intolerance, fasting hyperinsulinaemia and chemically induced (with STZ injection, for instance) ani-
and dyslipidaemia.146 Paradoxically, when the same diet was continu- mals.164,165 However, diet manipulations aimed at mimicking human
ously administered for additional 20 weeks, long-term (42 weeks) overnutrition and inadequate nutrient intake are often used (alone or
HSHFD consumption was able to change glucose tolerance and in combination with genetic/chemical approaches) to more closely
decrease plasma FFA levels, although the mice displayed elevated replicate the pathophysiological processes underlying T2DM progres-
FBG levels and dramatic hyperinsulinaemia.146 In Wistar rats, a predi- sion (Table 2). Owing to the large heterogeneity of experimental pro-
abetic stage was achieved by feeding the animals during 8 weeks with tocols aimed towards establishing an accurate diet-induced T2DM
WD (containing 45% kcal from fat and 35% kcal from CHO) and 20% animal model, careful choice of the most adequate model for a spe-
sucrose solution in drinking water. The animals developed obesity, cific purpose is required.
mild hyperglycaemia, hyperinsulinaemia, hyperlipidaemia and glucose The separate intake of HFD or HSD can promote important
intolerance.152 In other recent study, Vatandoust et al. 153
showed changes in metabolic pathways responsible for insulin resistance and
that consumption of a combination of solid HFD (30%) with fructose initial metabolic features underlying obesity, MetS or prediabetes, as
in drinking solution (30%) for 10 weeks effectively elicited a predia- mentioned above. However, the use of diets enriched with fat or sugar
betic state in Wistar rats, which was confirmed by increased FBG and to induce overt T2DM in rodents has yielded conflicting results.166–170
TG levels, together with insulin resistance and decreased insulin 170 For instance, C57BL/6J mice with a genetic predisposition for
secretion from pancreatic β-cells. developing features of T2DM have been widely used as diet-induced
models of T2DM and subsequent complications.171 In fact, although
some of the early diabetic features were noted immediately after
2.4 | Overt diabetes 4 weeks, a longer duration (16–25 weeks) of HFD (40–60% of calories
from fat) allowed the development of a well-established T2DM pheno-
Compared with what occurs in prediabetes and according to ADA and type, namely, higher hyperglycaemia, with FBG  240 mg/dl, IGT and
WHO, the current diagnostic criteria for diabetes are based on FBG insulin resistance, along with adipose tissue inflammation and obe-
or 2-h plasma glucose levels during OGTT and on HbA1c levels sity.172 This pattern of T2DM phenotype evolution in C57BL/6J mice
(Figure 1). A patient with diabetes is diagnosed when the FBG after has been described in several reports.171,173,174
8 h of fasting is ≥126 mg/dl (7.0 mmol/L) or 2-h plasma glucose level Unlike mice, the most commonly used rat strains (such as Wistar
is ≥200 mg/dl (11.1 mmol/L) during an OGTT (using a glucose load and Sprague–Dawley) are not genetically predisposed for T2DM phe-
containing the equivalent of 75-g glucose solution) or HbA1c ≥ 6.5% notype development.175 Nevertheless, there are noteworthy excep-
(48 mmol/L).119,154 tions, including the male Zucker diabetic fatty (ZDF) rat, a well-
Similar to the human condition, an animal model of T2DM relies established genetic model of obese T2DM derived from the selective
on the assessment of the key endpoints for glucose homeostasis, inbreeding of obese Zucker (leptin receptor-deficient) rat.176 The male
namely, blood glucose and insulin levels, GTT, insulin tolerance test ZDF rat displays a phenotype of progressive hyperglycaemia, reaching
(ITT), HOMA-IR, quantitative insulin sensitivity check index (QUICKI), a state of overt diabetes that can be accelerated or intensified with
32,155–157
HbA1c and whole-body insulin sensitivity. In most preclinical HFD intake.177,178 Apart from the expenses, the main drawback of
studies, FBG is the most common readout of a rodent's T2DM condi- this widespread model is the monogenic feature related to the muta-
tion. However, standardized guidelines and completely unified tion of the leptin receptor, which is uncommon in humans, thereby
Main features of diet-induced rodent models of prediabetes and overt diabetes
10

TABLE 2

Diet type Composition (%) Duration (weeks) Specie/strain Main features Refs
Prediabetes
High-fat based
HFD 46% kcal from fat, 24% kcal from 12 Male Sprague–Dawley rats Obesity: data not shown Abdel-Hamid and
CHO, 20.3% kcal from protein (±8-week-old) Increased FBG: ≈115 mg/ml Firgany130

Hyperinsulinaemia
IGT
45% kcal from fat, 35% kcal from 17 Male Sprague–Dawley rats Obesity: 32.2% Δ weight gain; Marques et al.60
CHO, 20% kcal from protein (7-week-old) 21.6% increase in fat mass
IGT
45% kcal from fat, 35% kcal from 17 Male Wistar rats Obesity: 66.9% Δ weight gain; 43% Marques et al.60
CHO, 20% kcal from protein (7-week-old) increase in fat mass
Fasting hyperinsulinaemia (at Week 9)
Increased HOMA-IR index (at Week 9)
IGT (at Weeks 9 and 17)
Hypertriglyceridaemia
59% kcal from fat, 20% kcal from 40 Male Wistar rats Obesity: 98% Δ weight gain; Chalkley et al.175
CHO (8-week-old) twofold increase in total fat mass
Mild elevated FBG: ≈135 mg/ml
Hyperinsulinaemia
Mild glucose intolerance
High-sugar based
HSD 35% sucrose in water standard 9 Male Wistar rats Obesity: no BW changes Nunes et al.112 and
diet (16-week-old) Fasting normoglycaemia: 102.90 mg/dl Burgeiro et al.136

Increased fed glucose: 162.90 mg/dl


Fasting hyperinsulinaemia
Increased HOMA-IR index
Decreased insulin sensitivity
IGT
Hypertriglyceridaemia
7% fructose (w/v) in water 12 Male Wistar rats Obesity: no BW changes Miranda et al.144
(6-week-old) Fasting normoglycaemia: 70.5 mg/dl
IGT
r-
PREGUIÇA ET AL.
TABLE 2 (Continued)

Diet type Composition (%) Duration (weeks) Specie/strain Main features Refs
Loss of β-cells
PREGUIÇA ET AL.

Increased β-cell mass


10% fructose (w/v) in drinking water 3 Male Wistar rats Obesity: no BW changes Fracini et al.145
(±8-week-old) Fasting normoglycaemia: 90 mg/dl
Fasting hyperinsulinaemia
Hypertriglyceridaemia
High-fat/high-sugar based
HSHFD 42% kcal from fat %, 37% kcal 22 Male C57BL/6 mice Obesity: ≈80% Δ weight gain; Kowalski et al.146
from CHO, 21% kcal from protein, (8-week-old) increased fat mass
Fasting normoglycaemia: 189 mg/dl
Fasting hyperinsulinaemia
Impaired glucose tolerance
42 Obesity: ≈60% Δ weight gain;
increased fat mass
Fasting normoglycaemia: 160 mg/dl
Fasting hyperinsulinaemia
45% kcal from fat, 35% kcal from 8 Male Wistar rats Obesity: 26% Δ weight gain; almost van den Brom et al.152
CHO; 21% kcal from protein (4-week-old) equal to twofold increase for both
epididymal and perirenal fat weight
Elevated FBG: 192 mg/dl
Decreased insulin sensitivity
IGT
Hypertriglyceridaemia
30% kcal from fat and 70% kcal from 10 Male Wistar rats Obesity: 23% Δ weight gain Vatandoust et al.153
CHO + 30% (w/v) of (6-week-old) Elevated fasting glucose: 105 mg/dl
fructose solution
Impaired glucose tolerance
Hypertriglyceridaemia
Overt diabetes
HSHFD 24% kcal from fat + 25% (w/v) 16 Male Wistar rats Obesity: 23% Δ weight gain Van der Werf et al.159
fructose in water (7-week-old) Elevated FBG: 135 mg/dl
Increased HOMA index
Fasting hyperinsulinaemia
Insulin resistance
Dyslipidaemia
11

(Continues)
12

TABLE 2 (Continued)

Diet type Composition (%) Duration (weeks) Specie/strain Main features Refs
HFD 60% kcal from fat, 20% kcal from 21 Male C57BL/6J mice Obesity: 85% Δ weight gain; 20% Guo et al.172
CHO, 20% kcal from protein, (4-week-old) increased body fat mass
Elevated FBG: 237.9 mg/dl
Fasting hyperinsulinaemia
Elevated HbA1c
Increased HOMA indexes
IGT
Insulin resistance
HSHF + 30% kcal from fat and 70% kcal 12 Male Wistar rats Obesity: 23% Δ weight gain Vatandoust et al.153
STZ from CHO + 30% (w/v) for fructose (6-week-old) Elevated FBG: 170–198 mg/dl
solution + STZ (30 mg/kg, twice)
Elevated fed glucose: ≥340 mg/dl
IGT
25.7-wt% lard and 46.5-wt% f 56 Male Wistar rats Obesity: No Δ weight gain Barriere et al.191
ructose + STZ (25 mg/kg, thrice) (10-week-old) Elevated fasting glucose: 270 mg/dl
(at 42 weeks)
IGT (starting at 10 weeks)
Dyslipidaemia
Pancreatic β-cell dysfunction with
subsequent insulinopenia
HFD + High fat diet (undisclosed 12 Male C57BL/6 mice Obesity: 20% Δ weight gain Goodzari et al.188
STZ composition) + STZ (45 mg/kg) (8-week-old) Elevated FBG: 195 mg/dl
IGT
Hyperinsulinaemia
Dyslipidaemia
Insulin resistance
PREGUIÇA ET AL.
PREGUIÇA ET AL. 13

educing the translational relevance.179 Although some studies have cardiovascular injury, these animals developed microvascular
shown typical features of overt diabetes in rat models of diet-induced angiopathies, namely, nephropathy, retinopathy and behavioural signs
T2DM, particularly with long-term protocols, the issue remains con- of peripheral neuropathy.191 In C57BL/6 mice, T2DM was success-
troversial, as diverse results have been reported. Upon dietary supple- fully induced by HFD plus STZ (45 mg/kg) for 12 weeks.188 The ani-
mentation of fructose (25% in drinking water) within an HFD regimen mals presented increase in BW, and serum TG and total cholesterol
of 4 months, Wistar rats displayed T2DM features comprising sys- levels, as well as increased FBG (>150 mg/dl) and OGTT (>300 mg/dl)
temic hyperglycaemia (FBG > 126 mg/dl), insulin resistance (HOMA- responses, together with hyperinsulinaemia, IGT and insulin resis-
IR > 2.4), glucose intolerance, dyslipidaemia, oxidative stress associ- tance.188 In another study, the C57BL/6J mouse model of T2DM was
159
ated with endothelial dysfunction and hepatic steatosis. Similarly, developed using HFD for 14 weeks with an STZ injection (100 mg/kg,
Kalaki-Jouybari et al.180 also reported a diabetic rat model in Wistar i.p.).192 Once again, these animals weighed significantly more than the
rats after supplementation with HFHSD (30% fat and 20% fructose) control mice and presented hyperglycaemia (FBG > 400 mg/dl) with-
for 5 months. These authors confirmed the presence of T2DM fea- out changes in TG levels.192 These observations have been corrobo-
tures, such as hyperglycaemia (FBG > 200 mg/dl), insulin resistance rated by other reports.184,186,187
(elevated HOMA-IR), dyslipidaemia and hepatic steatosis.180 How- Finally, it should be emphasized that diet-induced regimens com-
ever, both studies confirmed the diabetic phenotype in animals dis- bined with STZ injection should closely replicate the pathophysiologi-
playing FBG > 126 mg/dl, without discriminating between the time cal mechanisms underlying T2DM progression. As high doses of STZ
period of fasting. On the contrary, several studies reported that rats (>50 mg/kg) rapidly lead to destruction of pancreatic β-cells with
do not become overtly diabetic, even with extended periods of HFD acute insulin deficiency (a feature of typical type 1 diabetes), low
132,175 175
consumption. Accordingly, Chalkley et al. showed that the doses of STZ should be used as a more suitable approach for experi-
long-term (10 months) intake of an HFD (59% of the calories as fat mental T2DM and associated complications.
plus 20% of the calories as carbohydrate) by Wistar rats induced obe-
sity with large amounts of central abdominal fat and insulin resistance.
Nevertheless, this protocol failed to replicate overt fasting hyper- 2.5 | Nonalcoholic fatty liver disease
glycaemia.74,175 These observations were corroborated by other
authors who were unable to establish an overt T2DM phenotype after NAFLD is currently the most common chronic liver disease, with an
chronic HFHSD administration in rats.123,181 estimated global prevalence of 25%.193 NAFLD encompasses a spec-
As diet-induced paradigms for establishing overt T2DM are long- trum of diseases that develop in the absence of excessive alcohol con-
lasting and highly demanding, STZ administration is widely used as a sumption, ranging from simple steatosis, the hallmark feature of the
tool for inducing partial β-cell destruction and impairment of insulin disease, to nonalcoholic steatohepatitis (NASH), a more severe form of
secretion.164,182–184 This toxin is usually administered intraperitone- NAFLD characterized by inflammation and fibrosis. NAFLD and NASH
ally (i.p.) via single or multiple injections of varying dosages, ranging may progress to cirrhosis and ultimately hepatocellular carcinoma
from 25 to 45 mg/kg BW in rats and 40 to 100 mg/kg BW in (HCC).194 The development and progression of NAFLD is linked to vis-
185–189 183
mice. As an example, Mansor et al. showed that low doses ceral adiposity, insulin resistance, dyslipidaemia and T2DM, which in
(15–25 mg/kg BW) of STZ in Wistar rats, following 3 weeks of HFD turn are driven by several risk factors, including high intake of satu-
feeding (60% calories from fat), induced a T2DM phenotype, whereas rated fats and/or sugar, sedentary lifestyle and genetic susceptibility.
30 mg/kg BW of STZ more closely recapitulated type 1 diabetes. The lack of approved pharmacological therapies for
Identical results were recently reported by Guo et al.184 Conversely, NAFLD/NASH has stimulated intense drug discovery efforts in this
Srinivasan et al.190 reported that a single injection of 35 mg/kg BW of field. Several animal models of NAFLD have been developed, mostly
STZ is the optimal dose for T2DM phenotype development in in mice and rats, as platforms for increasing our understanding of the
Sprague–Dawley rats following 2 weeks of HFD (58% calories as fat), disease pathogenesis and for the evaluation of therapeutic interven-
with frank hyperglycaemia (FBG > 400 mg/dl) and hyper- tions. However, the currently available models have translational limi-
triglyceridaemia. Another report mentioned that HFHSD (30% and tations and do not completely recapitulate the entire spectrum of
70% of total calories from fat and carbohydrate, respectively) treat- human disease progression. Animal models of NAFLD use dietary,
ment for 10 weeks, followed by injections of low doses of STZ chemical, genetic or a combination of multiple approaches to recreate
(30 mg/kg, in two steps with 1-week interval), led to T2DM in Wistar specific pathogenic features. The assessment of NAFLD/NASH in
rats.153 Furthermore, administration of HFHFD (containing 46.5-wt% rodents is commonly based on liver histological analysis, liver triglyc-
fructose and 25.7-wt% lard) combined with low-doses of STZ eride content and plasma biomarkers (Figure 1).
(25 mg/kg, multiple injection) in Wistar rats for 56 weeks was used to As the primary trigger of NAFLD is overnutrition leading to obe-
mimic the evolutive nature of T2DM and the underlying advanced sity, rodent models based on nutritional manipulation are thought to
stage complications.191 At the end of the experimental protocol, the best reflect the human pathology. The majority of animal models focus
animals displayed frank hyperglycaemia (>300 mg/dl), marked on providing dietary components individually or in combination to
dyslipidaemia, hepatic fibrosis and pancreatic β-cell dysfunction with induce simple steatosis and steatohepatitis (Table 3). Furthermore,
subsequent insulinopenia.191 Interestingly, in addition to signs of even in genetic animal models of NASH, diet acts as a secondary
14 PREGUIÇA ET AL.

trigger for disease progression. Dietary regimens with high fat content to the atherogenic diet (80% kcal from fat) promoted hepatic insulin
(45–75% energy from fat) are widely used to induce NAFLD in resistance and further accelerated the progression to NASH.206
195
rodents. For example, an early study showed that consumption of Finally, male and female C57Bl/6 mice fed a diet with high fat (40%
HFD for approximately 10 weeks led to obesity, insulin resistance, kcal from fat), cholesterol (2%) and fructose (42 g/L) levels in the
hyperinsulinaemia, glucose intolerance and hepatic steatosis, whereas drinking water for 25 weeks exhibited obesity, steatosis with hepato-
increase in alanine aminotransferase (ALT) and aspartate aminotrans- cyte ballooning and progressive fibrosis.207
ferase (AST) levels, indicative of steatohepatitis, was observed only In rodents, the addition of fructose to drinking water results in
after 34–36 weeks in male C57BL/6J mice.196 Another study reported steatosis without features of NASH after 8 weeks and induces a sig-
steatosis in Sprague–Dawley rats within 1–2 weeks after HFD nificant increase in BW, and plasma TG and glucose levels.208,209
onset,197 whereas only minimal fibrosis was observed after extended Recently, Asgharpour et al.210 developed a dietary mouse model
196,198
exposure (36–50 weeks). However, the results regarding the based on an inbred isogenic strain of C57BL6/J and S129S1/svlmJ
onset and degree of steatosis, inflammation and fibrosis varied, which mice fed HFD with ad libitum consumption of glucose and fructose.
appeared to depend on species, gender, dietary fat content and com- These mice sequentially developed steatosis (4–8 weeks),
position and duration of HFD feeding.195 Nevertheless, HFD more steatohepatitis (16–24 weeks), progressive fibrosis (16 weeks
closely resembles the metabolic parameters of human NAFLD, onwards) and HCC.210
although the pathological and molecular alterations are not as severe.
The methionine–choline-deficient (MCD) diet is a well-
established nutritional model of advanced NASH. The complete defi- 3 | DISCUSSION
ciency of the essential nutrients, methionine and choline, combined
with high sucrose (40% of energy) and moderate fat (10–20%) con- 3.1 | Open challenges for diet-induced animal
tent, results in impaired formation and secretion of very low-density models
lipoprotein (VLDL) and impaired hepatic β-oxidation. This model is
characterized by macrovesicular steatosis, hepatocellular death, lobu- In strict contrast to the expectations of the medical and scientific
lar inflammation, oxidative stress (after 3–5 weeks) and early onset of community, research on animal models has not been translated into
liver fibrosis (after 8–10 weeks) in C57BL/6J mice.199 A limitation of significant therapeutic advancements, particularly for more complex
the MCD diet model is that its metabolic profile is opposite of that of and multifactorial diseases, such as the obesity-related metabolic dis-
human NASH, as this model shows significant weight loss (up to 40% orders. One reason for this failure may be the inability to translate the
in 10 weeks) and no peripheral insulin resistance, as well as low serum enormous preclinical research into effective therapies in humans,
insulin, fasting glucose and triglyceride levels,200 which prevents long- which may be strongly related to the complexity of these diseases per
term experimental fibrogenesis. These limitations can be overcome by se, and also to the poor representativeness of animal models with
long-term feeding of a choline-deficient HFD (CD-HFD), which respect to human diseases.
induces the key features of MetS (obesity and insulin resistance), Actually, in most of cases, the way the disease is induced, in par-
steatosis, fibrosis and HCC development.201 ticular, the main features of its evolution, do not reliably represent the
The choline-deficient, L-amino acid-defined (CDAA) dietary model complex pathophysiology associated with the development of obesity,
is similar to the MCD model regarding choline deficiency, although its evolution to MetS and prediabetes and further progression to
the dietary proteins are replaced by an equivalent and corresponding T2DM, with complications and associated consequences, including
mixture of L-amino acids. The CDAA model recapitulates features of NAFLD. Hence, a large number of studies using animal models are
human NASH in rodents by sequentially inducing steatohepatitis, liver induced by hypercaloric diets, which in some ways can more closely
202,203
fibrosis and HCC without any loss of BW. Feeding the CDAA mimic the evolution of diseases in humans. In fact, unhealthy eating,
diet to rats results in rapid progression of steatosis to fibrosis, typically hypercaloric diets, based on high sugar and fat contents and
followed by a rise in ALT level.203 In mice, the diet caused little or low amounts of micronutrients, is one of the main risk factors (and
negligible increase in ALT,204 and long-term feeding (20 weeks) was direct contributors) for the appearance and evolution of these condi-
202
required before liver fibrosis was observed. Finally, combining the tions. Furthermore, diet-induced models are usually associated with
CDAA diet with HFD accelerated the development of NASH with slow evolution, thus imitating human disease progression and opening
fibrosis (6–9 weeks) in mice.205 up the possibility of testing the efficacy of preventive or curative ther-
Western (atherogenic) diets typically contain cholesterol apeutic or nutraceutical strategies.
(1–1.25%) and cholate (0.5%), which promote atherosclerosis, Notwithstanding these advantages, the use of animal models of
resulting in liver damage and fibrosis in experimental animals. Male obesity and related metabolic diseases induced by hypercaloric diets
C57Bl/6J mice fed WD (30% kcal from fat) exhibited steatosis, and is not free from limitations. On the contrary, they are usually associ-
increased ALT and total cholesterol levels after 6 weeks, and hepato- ated with variables that strongly affect the type of model that can be
206
cellular ballooning and fibrosis after 24 weeks. However, obesity obtained, as discussed below. Another important issue involves the
was not observed in this model, and glucose tolerance and insulin sen- precise definition and application of criteria that enable reliable
sitivity appeared to improve.206 The addition of a high fat component cataloguing of a model. Although criteria for human diseases are
TABLE 3 Main features of diet-induced rodent models of NAFLD

Diet type Composition (%) Duration (weeks) Specie/strain Main features Refs
High-fat based
PREGUIÇA ET AL.

HFD 60% kcal from fat (lard, soybean oil), 50 Male C57BL/6J mice (8-week-old) Hepatic steatosis without inflammation Ito et al.196
20% kcal CHO, 20% kcal from protein (10 weeks)
Steatohepatitis with inflammatory
markers (40 weeks)
Obesity: 83% Δ weight gain
Glucose intolerance
Insulin resistance
Hyperinsulinaemia (10 weeks)
42% kcal from fat (lard, corn oil), 16 Female Sprague–Dawley rat Hepatic steatosis Gauthier et al.197
36% kcal from CHO, 22% kcal protein (6-week-old) Obesity: 26% Δ weight gain; 1.4-fold
increase in fat mass
Elevated plasma glucose: 153 mg/dl
Hyperglycaemia (12 weeks)
Hyperinsulinaemia
Hypertriglyceridaemia
45% kcal from fat (lard, soybean oil), 43 Male Sprague–Dawley rat Hepatic steatosis with inflammation Omagari et al.198
35% kcal from CHO, 20% kcal (3-week-old) and mild fibrosis
from protein Obesity: 188% Δ weight gain
Elevated FBG: 174 mg/dl
Hyperinsulinaemia
Methionine and choline deficient diets
MCD Methionine-choline deficiency, 8 Male C57BL/6J mice Hepatic steatosis (3 weeks), McCuskey et al.199
sucrose (40%) and fat (10%) (8-week-old) necroinflammation (5 weeks) and
perisinusoidal fibrosis (8 weeks)
CD-HFD Choline deficiency, and high fat (45%) 52 Male C57BL/6J mice Mild hepatic steatosis (12 weeks), Wolf et al.201
(4- to 5-week-old) marked steatosis (24 weeks),
hepatocyte ballooning and fibrosis
(52 weeks)
Obesity: 75% Δ weight gain; more than
threefold increase in fat mass
Dyslipidaemia
IGT (24 weeks)
CDAA Choline deficiency, L-amino 16 Male Fischer 344 rat Hepatic steatosis and inflammation Uto et al.203
acid-defined (7-week-old) (2 weeks)
Hepatic fibrosis and cirrhosis
(16 weeks)
15

(Continues)
(Continued)
16

TABLE 3

Diet type Composition (%) Duration (weeks) Specie/strain Main features Refs
Elevated blood glucose: ≈140 mg/dl

Hyperinsulinaemia
CDAA Choline deficiency, L-amino acid-defined 22 Male C57BL/6J mice (8-week-old) Hepatic steatosis with inflammatory Miura et al.202
(14% kcal from fat, 68.5% kcal from CHO, cell infiltration, hepatocyte
17.4% kcal from protein) ballooning and death, and hepatic
fibrosis
Obesity: 45.6% Δ weight gain; more
than threefold increase in epididymal
fat mass
Increased HOMA-IR index
Insulin resistance
Hypertriglyceridaemia
CDAA-HFD Choline deficiency, L-amino acid-defined, 14 Male C57BL/6J mice (6-week-old) Hepatic steatosis with lobular Matsumoto et al.205
high fat, methionine (0.1%) (62% kcal from inflammation and hepatocyte
fat, 21% kcal from CHO, 18% kcal from ballooning (3 weeks), hepatic
protein) fibrosis (6 weeks)
Obesity: 30% Δ weight gain
Hypertriglyceridaemia
Western (atherogenic) diet
WD Cholesterol (1.25%), cholate (0.5%) 24 Male C57BL/6J mice (8-week-old) Hepatic steatosis and inflammation Matsuzawa et al.206
(6 weeks), hepatocellular ballooning
and fibrosis (24 weeks)
Obesity: −4% Δ weight gain in relation
to control
FBG: 85 mg/dl
Hyperinsulinaemia
Increased cholesterol
WD+HF Cholesterol (1.25%), cholate (0.5%), 24 Male C57BL/6J mice (8-week-old) Exacerbated hepatic steatosis and Matsuzawa et al.206
high fat (60%) inflammation (6 weeks)
Hepatocellular ballooning and fibrosis
(24 weeks)
Obesity: −12% Δ weight gain in
relation to control
FBG: 93 mg/dl
Hyperinsulinaemia
Increased cholesterol
High-sugar based
PREGUIÇA ET AL.
PREGUIÇA ET AL.

TABLE 3 (Continued)

Diet type Composition (%) Duration (weeks) Specie/strain Main features Refs
HSD 20% (w/v) fructose in drinking water 8 Male Wistar rats (8-week-old) Hepatic steatosis Mamikutty et al.208
Obesity: 6.9% Δ weight gain
FBG: 145.8 mg/dl
Hypertriglyceridaemia
Elevated systolic blood pressure
30% (w/v) fructose in drinking water 8 C3H/HeOuJ mice (6- to 8-week-old) Hepatic steatosis and increased Spruss et al.209
inflammatory markers
Obesity: 24.2% Δ weight gain
Combinations
HSHFD 40% kcal from fat and 2% 25 Male and female C57BL/6J mice Hepatic steatosis, hepatocyte Charlton et al.207
cholesterol + 42%, (w/v) of ballooning, fibrosis and lobular
fructose inflammation
Obesity: 68% Δ weight gain
Elevated glucose levels: 243 mg/dl
Hyperinsulinaemia
Hypercholesterolaemia
42% kcal from fat, 0.1% kcal from 52 Male B6/129 mice (8- to 12-week-old) Hepatic steatosis (4 weeks), hepatocyte Asgharpour et al.210
cholesterol + solution of 23% ballooning (8 weeks), steatohepatitis,
fructose and 18% glucose lobular inflammation and progressive
fibrosis (16 weeks), spontaneous
hepatocellular cancer (52 weeks)
Obesity: 50% Δ weight gain
Insulin resistance
Hypertriglyceridaemia
17
18 PREGUIÇA ET AL.

defined by reputable international medical–scientific organizations, model, as summarized in Figure 2. The choice of the model will
the same is not always true for animal models. An exemplary case is depend on the study purpose and the pathophysiological relevance of
that of obesity. In humans, waist circumference is recognized as a the model. Animal models are used for various purposes, including
good predictor of obesity, which has been consistently shown to pro- understanding the mechanism of disease pathogenesis, identification
vide independent and better information than BMI, despite limited of new biomarkers and/or evaluation of therapeutic options, among
implementation in routine clinical practice.53 In addition, information others. Although the majority of genetic or diet-induced models of
regarding the amount and distribution of adipose tissue is of potential metabolic diseases are obese, the presence or absence of obesity
interest; however, it remains to be accurately assessed.52 In animal depends on the study purpose. Furthermore, some diet-induced
models of obesity, weight gain values and/or quantitative or qualita- models of obesity and related disorders evolve with other conditions,
tive changes in the various adipose tissue deposits that can be used to including dyslipidaemia and vascular complications,12 which may be
universally define cut-offs for rodents (rat and mouse) are lacking. The essential for the study, or confounders, depending on the study
absence of well-established criteria conflicts with the scientific rigour objectives.
of these experiments, rendering the translation of preclinical results The duration of the protocol is a strong determinant of the meta-
difficult. As mentioned above in the article, a similar absence of clear bolic outcome, as mentioned throughout this article. In general, con-
criteria applies to animal models of diet-induced diabetes. sidering the above noted variations between different species and
strains, obesity and MetS in rodents can be achieved by submitting
the animals to few (up to 10) weeks of HFD, although the percentage
3.2 | Issues to consider when choosing a diet- of BW increase and the changes in fat composition and distribution
induced animal model may vary substantially between studies, as described above. Overall,
HSD cannot induce robust models of obesity but can be used to
When a research group proposes to work with diet-induced animal develop MetS and prediabetes.112,136 Reduction of protocols might
models, it must consider all the variables that may interfere with the be possible, for example, when combining HFD with HSD, which

F I G U R E 2 Major variables to be considered


when choosing a diet-induced rodent model of
obesity and related metabolic disorders, with
focus on animal-related factors and protocol-
related factors. HFD, high-fat diet; HSD high-
sugar diet; MetS, metabolic syndrome; NAFLD,
non-alcoholic fatty liver disease; Ob, obesity; STZ,
streptozotocin; T2DM, type 2 diabetes mellitus
PREGUIÇA ET AL. 19

usually causes slight impairment of both glucose and lipid metabolism, larger adipocytes and higher visceral adipose inflammation than their
with obesity, increased TG and glucose intolerance as possible out- OR counterparts.219 Gender differences were observed when studies
comes. If longer protocols of HFD or HSD (but especially when com- showed that male mice are more likely to become obese than female
bined) are chosen, more advanced metabolic stages, such as NAFLD, mice and that the protection against obesity in female mice is obliter-
can be achieved in rodents. Overt diabetes, in particular, is easily (and ated by ovariectomy.220 Other relevant female-specific physiological
rapidly) achieved when STZ is used to accelerate β-cell failure. How- conditions, such as menopause and breast cancer, have been revealed
ever, the associated toxicity introduces an additional variable. A sum- via modelling of diet-induced obesity with OP/OR rats.221 This strat-
mary of the most representative protocols for each condition is egy may also be useful for studying the mechanisms underlying the
shown in Tables 1, 2 and 3. development of obesity-related disorders, such as MetS, diabetes and
Regarding the animal variables, species and strain differences are NAFLD.
critical points when selecting a model, as the susceptibilities to DIO The role of sexual dimorphism in the development of several types
and related metabolic disorders vary significantly in both rat and of diseases, including obesity-related metabolic disorders, is an emerg-
mouse,62,63,211 as discussed earlier in the article. Hence, whenever ing issue that has triggered enormous interest in the scientific commu-
possible, more than one species and strain should be used. In fact, nity, and which cannot be overlooked when selecting animal models
numerous studies based on single strains have shown that a subset of of metabolic diseases.211,221,222 Sex must be considered a biological
the most commonly used rodent strains, namely, the inbred C57Bl/6 variable as important, if not more, than other variables, such as age or
mice and the outbred Sprague–Dawley rats, do not respond to HFD strain. For many years, experimental animal research has been per-
feeding.68,212 Importantly, studies evaluating a large collection of formed on males, with the argument that the female hormonal cycle
inbred strains not only provide better understanding of this variation induces higher variability and that the metabolic disease phenotype is
but also mimic the diversity observed in human populations. One such more pronounced in males, However, obesity, T2DM and other asso-
study compared the metabolic responses to HFD in males and females ciated metabolic diseases occur in both men and women, despite
of 43 inbred mouse strains.213 That study reported broad phenotypic some important sex-based differences in susceptibility to DIO and
213
diversity among strains and gender for most traits measured. It is diet-induced insulin resistance.223,224 Although obesity is more preva-
noteworthy that specific rat and mouse strains from different sup- lent among women,225 they are usually less likely to suffer from diet-
pliers differ significantly, which introduces an additional degree of induced consequences of T2DM,226–228 which is in line with its lesser
complexity to the issue. global prevalence in women than in men.229 The effect of gonadal
These issues cannot be separated from genetic and epigenetic hormones on fat distribution might explain, at least partially, the sex-
diversity, as it is well known that obesity and associated disorders are ual dimorphism observed in both women and female rodents. In fact,
caused by a combination of environmental factors (such as diet) and compared with their male counterparts, females have less visceral and
polygenic predisposition. Some inbred strains are maintained to spon- more subcutaneous fat, which negatively affects cardiometabolic
taneously develop a DIO phenotype, which are crucial for determining risk.230,231 In female rats and mice, the masculinization of fat distribu-
the precise underlying genetic mechanisms. Unfortunately, inbred tion due to ovariectomy, which ablates the production of endogenous
strains that are prone to DIO development represent only a small frac- oestradiol and progesterone, augments the susceptibly to diet-
tion of human metabolic phenotypes.59,214 Recently, researchers have induced insulin resistance.232 Evidences suggest that sex chromo-
focused on outbred strains, which represent the polygenic variability somes, independent of gonadal sex, are also involved in distinct meta-
of the human population more reliably.215 The administration of HFD bolic pathways, which may be linked with susceptibility to DIO and
to an outbred strain (e.g., Wistar or Sprague–Dawley) will differently diet-induced insulin resistance, such as feeding behaviour, glucose
impact the animal's metabolism; whereas some will show rapid homeostasis, adiposity and fatty liver.233,234
increase in weight (OP), others will not show any changes in BW Nowadays, improved information on sex differences in rodent
(OR) despite the environmental pressure.70 Thus, the first step in species and strains are available, some of which have been obtained
these studies involves identification of the animals that present a from elegant studies using OP/OR or DIO/DR rodents selected from
polygenic predisposition or resistance to obesity. The differences outbred and/or inbred strains; these are indubitably valuable tools for
between these phenotypes are the keys for understanding metabo- studying obesity and the associated metabolic complications, includ-
lism and the mechanism via which genetics and the environment ing sex-specific differences.221,235,236 In general, progression of DIO is
interact and modulate each other. In fact, there are several character- comparable between male and female rats,68,237 whereas male mice
ized differences, namely, gastrointestinal fat absorption, appetite, are more susceptible than females, developing obesity earlier and
216
physical activity, fat storage and/or consumption. These changes more prominently.220,232 Sexual dimorphism regarding diet-induced
appear to indicate that these animals have contrasting metabolic flexi- insulin resistance and glucose intolerance is also observed in both
bility, a readout of their capacity to adjust fuel oxidation to fuel avail- mice and rats, with males being the most affected.237,238 The same
ability.217 In fact, studies have shown that different mice strains with pattern is observed in most rodent models of T2DM.222 As described
distinct genetic backgrounds may display significant differences in glu- above, the ZDF rat is a clear and interesting example, as males
cose pathways, detected as early as from the time of weaning.218 develop severe T2DM under normal rodent chow diet, unlike the
Regarding fat storage, OP rats are characterized by the presence of inability of females to develop hyperglycaemia, despite developing
20 PREGUIÇA ET AL.

obesity to an extent similar to that observed in the males.177,239 The rates vary with fatty acids, contributing to different responses regard-
overall conclusion is that the reliability of the results and their poten- ing fat accumulation, weight gain, insulin resistance and
244,250–253
tial for translation into human disease-related research can signifi- inflammation. Whereas monounsaturated fatty acids posi-
cantly increase if researchers consider sexual dimorphism when tively affect weight control via increase in postprandial fat oxidation
selecting the animal model, as has been strongly suggested in previous and diet-induced thermogenesis, the intake of saturated fatty
studies.222,240,241 acids251,252 or n-3 polyunsaturated fatty acids (PUFAs; flaxseed oil
Furthermore, animal age should be carefully considered during rich in linoleic acid) produces opposite effects, promoting body fat
the experiment because of its effect on the disease, in particular, the accumulation.250,253 Furthermore, the micronutrient components of
age at the beginning of a dietary intervention. In humans, increasing TWD have been used in rodent studies to evaluate their effect on
age is a risk factor for the development of chronic metabolic diseases. weight gain and biomarkers of metabolic function.45 This report
BW increases with age, accompanied by redistribution of fat mass, showed that mice fed HFD containing vitamins and oil minerals, which
namely, to the muscle and hepatic tissues, which might explain the reflects the Western dietary pattern, inhibited hyperphagia and
increased susceptibility to glucose intolerance and insulin resistance, weight gain associated with the higher fat content of TWD. These
as reviewed by Kleinert et al.10 Similar metabolic changes in BW and observations reinforce the importance and necessity of considering
242,243
fat composition and redistribution occur in rodents with age. the contribution of micronutrients intake in preclinical models of obe-
However, glucose intolerance in rats is affected more by age than in sity and associated metabolic disorders.45 It is noteworthy that some
10
mice. The age at the beginning of a dietary protocol should also be transgenic or knockout models may not show an overt diabetic phe-
considered, as metabolism changes substantially from preadult age to notype under normal conditions and have to be fed hypercaloric diet
old age, and different effects of dietary interventions should be to completely express the effect of genetic background on the
expected if young, adult, or old animals are used at the beginning of expected phenotype.254–256 In addition to diet composition, the tex-
61,211
the protocol. ture and palatability of the diet pellets are important factors affecting
The type of diet and its specific composition are also particularly consumption, considering the high sensitivity of animals to sensorial
important. The specific composition of diets, such as the percentages stimuli; this will ensure that diet consumption remains unchanged due
of CHO, fats and proteins (as well as fibre and water), considerably to these parallel factors.
affects the appearance and progression of obesity and associated con- Importantly, in studies on diet-induced rodent models of meta-
ditions and complications.244,245 Furthermore, researchers should also bolic disorders, the amount of food consumed should be accurately
consider the effect of the micronutrient composition on the study monitored, as rodents fed more energy-dense diets (such as DIO 45
outcome, as they are responsible for several key metabolic functions or 60) will compensate by consuming less food. However, despite
in distinct organs and tissues.246–248 In fact, intake of vitamins and the lower intake, they will consume more energy than animals fed
mineral plays a key role in moderating the hyperphagic behaviour of lower energy feed (such as AIN-93). In addition, both total and
44
C57BL/6J mice fed HFD. The percentage of fat is a key aspect that fractionated (from CHO, fat and protein) caloric intake should be
should be considered. Diets containing 45% and 60% fat by calories calculated, as they may be potentially relevant for the study. In
are considered HFD for rodents, as both induce obesity. As mice on this context, the inclusion of accurate control groups fed diets of
60% fat diet become more obese in less time than mice on 45% fat composition that can be properly compared with those of the
35
diet, the 60% rodent diet appears to be better at first. However, diseased animals should be considered when planning an experi-
compared with the typical American or European human diet (con- mental protocol.
taining about 30–40% fat by energy), experimental use of diets with Regarding the choice of proper control diets, most metabolic stud-
high levels of fat do not adequately mimic the levels of fat in the ies have frequently used refined diet (e.g., refined HFD or rHFD), in
human diet. A detailed analysis of the resultant obesity and metabo- which each nutrient is supplied by a specific and purified ingredient,
lism of mice fed 45% and 60% fat diets revealed that both the extent compared with a plant-based, unrefined control chow diet.32,257
of obesity and dysregulation of glucose homeostasis increased with However, refined and chow diets exhibit marked differences in
the fat percentage.103 Comparison of the metabolite profiles in the micronutrient and macronutrient content. Therefore, when comparing
lungs of mice fed low fat (10%) versus HFD (45% fat) versus very the effects of a chow diet with those of a refined HFD, the effects of
HFD (60% fat) showed that the profile of 80 metabolites had changed the dietary fat will be confounded with the effects of other compo-
in the HFD-fed mice relative to that fed the control diet.249 Alter- nents in the diets. The two major differences between chow and
ations in the metabolites of the tricarboxylic acid cycle, antioxidants refined diets are the phytoestrogen content, which is high in chow
and nucleotide pathways, as well as significant changes in neutral diets but is absent from refined diets, and sucrose, which is used as a
lipids, fatty acids, phospholipids and sphingolipids, suggested that diet carbohydrate source in refined diets but is absent from chow. In addi-
composition and obesity induced lung metabolome changes, which tion, chow diets are not standardized, as their composition varies
may contribute to disease progression.249 A caveat of this type of diet with the supplier and batch. Considering these differences, a refined
is the absence of other components, such as fatty acids, which are low-fat diet (rLFD), formulated using the same purified ingredients as
known to influence the level of obesity, as its presence in rodent diets rHFD, is a more appropriate control diet than chow,258,259 which is
250
may better mimic human diet. In fact, the oxidation and deposition particularly clear when focusing on the effects of gut microbiota,260
PREGUIÇA ET AL. 21

as will be discussed later. However, recent studies have reported human diseases and might also yield important insights regarding our
conflicting results regarding the metabolic effects of LFD and chow understanding of mammalian physiology.
diets. In one study, mice fed LFD did not show changes in BW or fat The gut microbiota plays major roles in host homeostasis due to
mass,261 which disagrees with the results of other studies in which several metabolic functions associated with the regulation of glucose,
mice fed LFD showed increased BW and adiposity compared with lipids and energy.269 Several lines of evidence suggest that impair-
mice fed chow diet.258,259 Furthermore, the effects of rHFD on meta- ment of the gut microbiota (dysbiosis) contributes to the development
bolic outcomes, such as adiposity, inflammation and insulin sensitiv- of obesity, diabetes and associated complications.270 At the same
ity, depend on the control LFD used, which further reinforces the time, the microbial composition is highly shaped by dietary substrates,
notion that comparisons of studies using chow as the control diet are and HFD supply has been associated with changes in gut microbial
prone to unintended but significant metabolic disturbances, strongly composition and diversity, often referred to as obese microbiota.271
suggesting that adequate rLFD should be used as the control for Conversely, other studies reported body fat accumulation, hepatic
rHFD.258,259 steatosis and fasting hyperglycaemia in animals fed standardized WD,
The manner in which determinations are performed (such as independent of gut microbiota dysbiosis,46 thereby indicating an
fasted or fed conditions and tests, among other relevant details) and effect of specific diet composition on the gut microbiome. Fibres, in
the researchers' experience in performing these procedures should particular, are chief dietary substrates shaping gut microbiota compo-
also be considered. In this complex system, even the origin (supplier) sition.272,273 Gut microbiota modulation using dietary components is
of the animals and their housing conditions (e.g., temperature) can important in the context of both hypercaloric and control diets used
affect the results, for example, by interfering with gut microbial com- in animal models of obesity and associated metabolic disorders. In
munity of rodents, as discussed below. 262
fact, as mentioned above, preclinical studies often use “chow diet” as
In fact, housing temperature is another variable that profoundly the control for refined HFDs, which may result in significant bias, as
affects the outcome and interpretation of metabolic experiments. An refined diets contain purified ingredients, whereas chow diets contain
increasing body of evidence shows that the cold stress to which dietary fibre obtained from unrefined cereals and legumes, such as in
rodents are commonly subjected significantly affects physiology and low fat grain-based diet.260 The absence of fermentable fibres can
may compromise the ability to model some human diseases, namely explain, at least partially, why mice fed refined low-fat diet (rLFD), for-
obesity and related disorders.263,264 Most studies are conducted at an mulated using the same purified ingredients as rHFD, presented more
ambient temperature of 20–22 C, which is below the thermoneutral adiposity than chow-fed mice.259,260 These experiments emphasized
 265
zone for mice and rats (29–31 C). Housing of rodents below their that chow diet and rLFD are indeed not nutritionally equivalent and
thermoneutral zone results in the activation of adaptive thermogene- that the former should be used as the control for rHFD.258,259
sis for maintaining their core temperature. Indeed, at 22 C, rodents Complementing these observations, Dalby et al.261 showed that
expend twice as much energy and consume more food than those at microbiome composition was more dependent on whether the experi-
 265
ambient temperatures of 30 C, which results is substantial differ- mental diet was refined or complex, independent of an obese pheno-
ences in their metabolic, cardiovascular and immune phenotypes. type, suggesting more careful characterization of the association
Thus, the constant thermal stress induced by the animal housing con- between gut microbiota and obesity.
ditions may significantly affect the outcomes of previous metabolic In addition, evidence shows that the impact of diet on gut micro-
studies. Certain phenotypes observed under conditions of thermal biota may vary substantially between distinct species, strains and sex.
stress disappear when mice are housed at thermoneutrality, whereas A comparative study between 7-week-old male Wistar and Sprague–
others that are more consistent with human physiology emerge. For Dawley rats under HFD (45% energy from lipids and 17% from
example, a recent study showed that male C57BL/6J mice housed at sucrose) for 17 weeks suggested that the differences observed in the
thermoneutrality showed accelerated onset of HFD-induced inflam- gut microbial ecology may contribute to the worsened metabolic sce-
mation in WAT and vasculature, which promotes atherosclerosis but nario observed in the Wistar rat.60 In another study, 4 weeks of
266
not insulin resistance. Another study using C57BL/6 mice investi- methionine-restricted diet (0.15%) was able to promote significant
gated the role of thermoneutral housing as opposed to standard hous- sex-specific changes in the intestinal microbiome of 20-week-old male
267
ing in HFD-induced NAFLD. The authors reported that 24 weeks and female C57BL/6 mice, compared with mice on control diet con-
of thermoneutral housing lowered stress-driven production of cortico- taining 0.65% methionine.274 Finally, even rodents from different pro-
sterone, increased proinflammatory immune responses and exacer- duction facilities possess distinct gut microbiota, as reported in a
bated HFD-induced NAFLD pathogenesis without significant changes study using obesity/diabetes-prone C57Bl/6J and obesity/diabetes-
267
in BW gain. In particular, female mice housed at thermoneutrality, resistant 129S1/SvImJ mice from Jackson Laboratory and obesity-
which are typically resistant to HFD-induced obesity and NAFLD, also prone/diabetes-resistant 129S6/SvEvTac mice from Taconic,
developed severe obesity and hepatocellular damage.267 However, suggesting a significant contribution to differences in susceptibility to
others have challenged this notion and suggested that the optimal DIO.262 Collectively, the available evidence emphasized the impor-

ambient temperature is 23–25 C for single-housed mice and tance of careful choice of animals and diets when using models of
20–22 C for group-housed mice.268 Nevertheless, future research in obesity and related metabolic conditions, particularly for studies
thermoneutral rodents might enable more predictive modelling of aimed at evaluating the effects on gut microbiota.
22 PREGUIÇA ET AL.

4 | CO NC LUSIO NS FUNDING INF ORMATI ON


This work was also supported by European Regional Development
Owing to the dearth of perfect animal models of obesity and Fund (FEDER), through Programa Operacional Factores de Com-
related metabolic diseases, selection of models that can be path- petitividade COMPETE2020 (CENTRO-01-0145-FEDER-000012-
ologically relevant for the scientific aims of a study is important. HealthyAging2020) and by National funds via Portuguese Science and
Despite the underlying limitations and critical aspects, diet modi- Technology Foundation (FCT): Strategic Projects UID/NEU/04539/
fication can be an appealing strategy, as unbalanced eating habit 2013, UID/NEU/04539/2019, UIDB/04539/2020 and UIDP/04539/
is one of the main risk factors for the development of these 2020 (CIBB); PhD Fellowship SFRH/BD/109017/2015; PTDC/SAU-
pathologies in humans. In addition, the slow and progressive evo- NUT/31712/2017, as well as by COMPETE-FEDER funds (POCI-01-
lution of the disease and the continuum of associated pheno- 0145-FEDER-007440 and POCI-01-0145-FEDER-031712).
types (glucose intolerance, hyperglycaemia, insulin resistance,
hyperlipidaemia, glucotoxicity and lipotoxicity, oxidative stress and OR CID
inflammation and β-cell failure) better mimic human metabolic Inês Preguiça https://orcid.org/0000-0002-7371-1079
diseases and allow evaluation of the potential of therapeutic or André Alves https://orcid.org/0000-0002-8161-5239
nutraceutical interventions. Sara Nunes https://orcid.org/0000-0002-0240-9059
HFD and HSD, alone or in combination, are the most widely Rosa Fernandes https://orcid.org/0000-0001-7828-2296
used diets for inducing metabolic diseases associated with obesity Pedro Gomes https://orcid.org/0000-0003-1766-9133
in rodent models. The percentage and type of each of the main Sofia D. Viana https://orcid.org/0000-0002-1316-1319
macromolecules (especially sugars and fats), the specific composition Flávio Reis https://orcid.org/0000-0003-3401-9554
of micronutrients, as well as the duration of exposure, are the key
factors determining the development of a desired model. In the case RE FE RE NCE S
of overt diabetes, the use of chemical inducers/accelerators of pan- 1. Ng M, Fleming T, Robinson M, et al. Global, regional, and national
creatic β-cell damage (mainly STZ) should be additionally considered. prevalence of overweight and obesity in children and adults during
However, researchers should consider the innumerable variables 1980–2013: a systematic analysis for the Global Burden of Disease
affecting the correct choice of an animal model of obesity and dis- Study 2013. Lancet. 2014;384(9945):766-781.
2. Pi-Sunyer X. The medical risks of obesity. Postgrad Med. 2009;
orders associated with obesity (MetS, prediabetes, T2DM and
121(6):21-33.
NAFLD). In fact, besides diet type and composition, control diet and 3. Global BMIMC, Di Angelantonio E, Bhupathiraju Sh N, et al. Body-
protocol duration, the rodent species and strain used, as well as mass index and all-cause mortality: individual-participant-data meta-
their age and gender, can also significantly affect the results. In analysis of 239 prospective studies in four continents. Lancet. 2016;
388(10046):776-786.
addition, factors such as the origin of the animals and the way they
4. Collaborators GBDO, Afshin A, Forouzanfar MH, et al. Health effects
are housed and maintained can also bias the expected outcome. of overweight and obesity in 195 countries over 25 years. N Engl J
Furthermore, as the main goal of animal models is to recapitulate Med. 2017;377(1):13-27.
the various forms of human diseases, leveraging of gen- 5. Webb VL, Wadden TA. Intensive lifestyle intervention for obesity:
principles, practices, and results. Gastroenterology. 2017;152(7):
etic/epigenetic diversity to maximize a specific phenotype of inter-
1752-1764.
est is clearly a compelling and powerful approach that warrants 6. Foright RM, Presby DM, Sherk VD, et al. Is regular exercise an effec-
further investigation. tive strategy for weight loss maintenance? Physiol Behav. 2018;188:
All these conditions and variables demand careful evaluation, 86-93.
7. Teixeira-Lemos E, Nunes S, Teixeira F, Reis F. Regular physical exer-
such that the best model can be selected to achieve scientific results
cise training assists in preventing type 2 diabetes development:
with higher potential for translation into human disease-related focus on its antioxidant and anti-inflammatory properties. Cardiovasc
research. In addition to the use of efficient and overlapping methods, Diabetol. 2011;10:12.
definition of clear and universally accepted criteria for these diseases 8. Kim GW, Lin JE, Blomain ES, Waldman SA. Antiobesity pharmaco-
therapy: new drugs and emerging targets. Clin Pharmacol Ther. 2014;
in rodents, which can be reliably compared with those existing for dis-
95(1):53-66.
ease diagnosis in humans, will be a key step towards improving pre- 9. Bray GA, Fruhbeck G, Ryan DH, Wilding JP. Management of obesity.
clinical research and its ability to generate better tools for use in Lancet. 2016;387(10031):1947-1956.
humans. 10. Kleinert M, Clemmensen C, Hofmann SM, et al. Animal models of
obesity and diabetes mellitus. Nat Rev Endocrinol. 2018;14(3):
140-162.
CONF LICT OF IN TE RE ST
11. King AJ. The use of animal models in diabetes research. Br J
The authors declare no conflict of interest. Pharmacol. 2012;166(3):877-894.
12. Preguica I, Alves A, Nunes S, et al. Diet-induced rodent models of
AUTHOR CONTRIBUTIONS diabetic peripheral neuropathy, retinopathy and nephropathy. Nutri-
ents. 2020;12(1):250.
All the authors have substantially contributed to the final version,
13. Bray GA, York DA, Fisler JS. Experimental obesity: a homeostatic
which was revised and approved by all. I.P., S.N. and P.G. performed failure due to defective nutrient stimulation of the sympathetic ner-
the tables. A.A. and F.R. performed the first draft of figures. vous system. Vitam Horm. 1989;45:1-125.
PREGUIÇA ET AL. 23

14. York DA. Lessons from animal models of obesity. Endocrinol Metab 35. Speakman JR. Use of high-fat diets to study rodent obesity as a
Clin North Am. 1996;25(4):781-800. model of human obesity. Int J Obes (Lond). 2019;43(8):1491-1492.
15. van der Worp HB, Howells DW, Sena ES, et al. Can animal models 36. Aguila MB, Pinheiro Ada R, Parente LB, Mandarim-de-Lacerda CA.
of disease reliably inform human studies? PLoS Med. 2010;7(3): Dietary effect of different high-fat diet on rat liver stereology. Liver
e1000245. Int. 2003;23(5):363-370.
16. Beigrezaei S, Ghiasvand R, Feizi A, Iraj B. Relationship between die- 37. Flanagan AM, Brown JL, Santiago CA, Aad PY, Spicer LJ, Spicer MT.
tary patterns and incidence of type 2 diabetes. Int J Prev Med. 2019; High-fat diets promote insulin resistance through cytokine gene
10:122. expression in growing female rats. J Nutr Biochem. 2008;19(8):
17. Rice Bradley BH. Dietary fat and risk for type 2 diabetes: a review of 505-513.
recent research. Curr Nutr Rep. 2018;7(4):214-226. 38. Taylor LE, Gillis EE, Musall JB, Baban B, Sullivan JC. High-fat diet-
18. Forouhi NG, Misra A, Mohan V, Taylor R, Yancy W. Dietary and induced hypertension is associated with a proinflammatory T cell
nutritional approaches for prevention and management of type profile in male and female Dahl salt-sensitive rats. Am J Physiol Heart
2 diabetes. BMJ. 2018;361:k2234. Circ Physiol. 2018;315(6):H1713-H1723.
19. Rodriguez-Monforte M, Sanchez E, Barrio F, Costa B, Flores- 39. Bae CR, Hino J, Hosoda H, et al. Adipocyte-specific expression of C-
Mateo G. Metabolic syndrome and dietary patterns: a systematic type natriuretic peptide suppresses lipid metabolism and adipocyte
review and meta-analysis of observational studies. Eur J Nutr. 2017; hypertrophy in adipose tissues in mice fed high-fat diet. Sci Rep.
56(3):925-947. 2018;8(1):2093.
20. Huang D, Refaat M, Mohammedi K, Jayyousi A, Al Suwaidi J, 40. Han L, Li T, Du M, Chang R, Zhan B, Mao X. Beneficial Effects of
Abi KC. Macrovascular complications in patients with diabetes and Potentilla discolor Bunge Water Extract on Inflammatory Cytokines
prediabetes. Biomed Res Int. 2017;2017:7839101. Release and Gut Microbiota in High-Fat Diet and Streptozotocin-
21. Riazi K, Raman M, Taylor L, Swain MG, Shaheen AA. Dietary Induced Type 2 Diabetic Mice. Nutrients. 2019;11 (3):670. https://
patterns and components in nonalcoholic fatty liver disease doi.org/10.3390/nu11030670
(NAFLD): what key messages can health care providers offer? Nutri- 41. Buettner R, Parhofer KG, Woenckhaus M, et al. Defining high-fat-
ents. 2019;11(12). diet rat models: metabolic and molecular effects of different fat
22. Del Prato S. Role of glucotoxicity and lipotoxicity in the pathophysi- types. J Mol Endocrinol. 2006;36(3):485-501.
ology of type 2 diabetes mellitus and emerging treatment strategies. 42. Do M, Lee E, Oh M-J, Kim Y, Park H-Y. High-Glucose or -Fructose
Diabet Med. 2009;26(12):1185-1192. Diet Cause Changes of the Gut Microbiota and Metabolic Disorders
23. Akhtar DH, Iqbal U, Vazquez-Montesino LM, Dennis BB, Ahmed A. in Mice without Body Weight Change. Nutrients. 2018;10 (6):761.
Pathogenesis of insulin resistance and atherogenic dyslipidemia in https://doi.org/10.3390/nu10060761
nonalcoholic fatty liver disease. J Clin Transl Hepatol. 2019;7(4): 43. Jurgens H, Haass W, Castaneda TR, et al. Consuming fructose-
362-370. sweetened beverages increases body adiposity in mice. Obes Res.
24. Bergman M. Pathophysiology of prediabetes and treatment implica- 2005;13(7):1146-1156.
tions for the prevention of type 2 diabetes mellitus. Endocrine. 2013; 44. Hintze KJ, Benninghoff AD, Cho CE, Ward RE. Modeling the West-
43(3):504-513. ern diet for preclinical investigations. Adv Nutr. 2018;9(3):263-271.
25. Savage DB, Petersen KF, Shulman GI. Disordered lipid metabolism 45. Monsanto SP, Hintze KJ, Ward RE, Larson DP, Lefevre M,
and the pathogenesis of insulin resistance. Physiol Rev. 2007;87(2): Benninghoff AD. The new total Western diet for rodents does not
507-520. induce an overweight phenotype or alter parameters of metabolic
26. DeFronzo RA. Pathogenesis of type 2 diabetes mellitus. Med Clin syndrome in mice. Nutr Res. 2016;36(9):1031-1044.
North Am. 2004;88(4):787-835. ix 46. Bortolin RC, Vargas AR, Gasparotto J, et al. A new animal diet based
27. Zheng Y, Ley SH, Hu FB. Global aetiology and epidemiology of type on human Western diet is a robust diet-induced obesity model: com-
2 diabetes mellitus and its complications. Nat Rev Endocrinol. 2018; parison to high-fat and cafeteria diets in term of metabolic and gut
14(2):88-98. microbiota disruption. Int J Obes (Lond). 2018;42(3):525-534.
28. Li Y, Xu W, Liao Z, et al. Induction of long-term glycemic control in 47. Organization WH. Obesity and overweight. February 16, 2018;
newly diagnosed type 2 diabetic patients is associated with improve- www.who.int/news-room/fact-sheets/detail/obesity-and-
ment of β-cell function. Diabetes Care. 2004;27(11):2597-2602. overweight
29. Lebeaupin C, Vallee D, Hazari Y, Hetz C, Chevet E, Bailly-Maitre B. 48. Bray GA, Kim KK, Wilding JPH, World OF. Obesity: a chronic
Endoplasmic reticulum stress signalling and the pathogenesis of relapsing progressive disease process. A position statement of the
non-alcoholic fatty liver disease. J Hepatol. 2018;69(4):927-947. World Obesity Federation. Obes Rev. 2017;18(7):715-723.
30. Quesada-Vázquez S, Aragonès G, Del Bas JM, Escoté X. Diet, Gut 49. Collaboration NCDRF. Worldwide trends in body-mass index,
Microbiota and Non-Alcoholic Fatty Liver Disease: Three Parts of underweight, overweight, and obesity from 1975 to 2016: a pooled
the Same Axis. Cells. 2020;9 (1):176. https://doi.org/10.3390/ analysis of 2416 population-based measurement studies in 128.9
cells9010176 million children, adolescents, and adults. Lancet. 2017;390(10113):
31. Mohan S, Brown L, Ayyappan P. Endoplasmic reticulum stress: a 2627-2642.
master regulator of metabolic syndrome. Eur J Pharmacol. 2019;860: 50. Despres JP. Body fat distribution and risk of cardiovascular disease:
172553. an update. Circulation. 2012;126(10):1301-1313.
32. Lai M, Chandrasekera PC, Barnard ND. You are what you eat, or are 51. Neeland IJ, Turer AT, Ayers CR, et al. Dysfunctionlal adiposity and
you? The challenges of translating high-fat-fed rodents to human the risk of prediabetes and type 2 diabetes in obese adults. JAMA.
obesity and diabetes. Nutr Diabetes. 2014;4:e135. 2012;308(11):1150-1159.
33. Gasparin FRS, Carreno FO, Mewes JM, et al. Sex differences in 52. Nuttall FQ. Body mass index: obesity, BMI, and health: a critical
the development of hepatic steatosis in cafeteria diet-induced obe- review. Nutr Today. 2015;50(3):117-128.
sity in young mice. Biochim Biophys Acta Mol Basis Dis. 2018; 53. Ross R, Neeland IJ, Yamashita S, et al. Waist circumference as a vital
1864(7):2495-2509. sign in clinical practice: a Consensus Statement from the IAS and
34. Moore BJ. The cafeteria diet—an inappropriate tool for studies of ICCR Working Group on Visceral Obesity. Nat Rev Endocrinol. 2020;
thermogenesis. J Nutr. 1987;117(2):227-231. 16(3):177-189.
24 PREGUIÇA ET AL.

54. Wagner DR, Castaneda F, Bohman B, Sterr W. Comparison of a 2D 74. Lozano I, Van der Werf R, Bietiger W, et al. High-fructose and high-
iPad application and 3D body scanner to air displacement plethys- fat diet-induced disorders in rats: impact on diabetes risk, hepatic
mography for measurement of body fat percentage. J Hum Nutr Diet. and vascular complications. Nutr Metab (Lond). 2016;13:15.
2019;32(6):781-788. 75. Hafizur RM, Raza SA, Chishti S, Shaukat S, Ahmed A. A ‘humanized’
55. Gibby JT, Njeru DK, Cvetko ST, Heiny EL, Creer AR, Gibby WA. rat model of pre-diabetes by high fat diet-feeding to weaning wistar
Whole-body computed tomography-based body mass and body fat rats. Integr Obes Diabetes. 2015;1(2):44-48.
quantification: a comparison to hydrostatic weighing and air dis- 76. Sugano M, Yamato H, Hayashi T, et al. High-fat diet in low-dose-
placement plethysmography. J Comput Assist Tomogr. 2017;41(2): streptozotocin-treated heminephrectomized rats induces all features
302-308. of human type 2 diabetic nephropathy: a new rat model of diabetic
56. Nickerson BS, Fedewa MV, Cicone Z, Esco MR. The relative accu- nephropathy. Nutr Metab Cardiovasc Dis. 2006;16(7):477-484.
racy of skinfolds compared to four-compartment estimates of body 77. Oliveira LS, Santos DA, Barbosa-da-Silva S, Mandarim-de-
composition. Clin Nutr. 2019;39(4):1112-1116. Lacerda CA, Aguila MB. The inflammatory profile and liver damage
57. Shen W, Wang Z, Punyanita M, et al. Adipose tissue quantification of a sucrose-rich diet in mice. J Nutr Biochem. 2014;25(2):193-200.
by imaging methods: a proposed classification. Obes Res. 2003;11(1): 78. Roopchand DE, Carmody RN, Kuhn P, et al. Dietary polyphenols
5-16. promote growth of the gut bacterium Akkermansia muciniphila and
58. Peig J, Green AJ. The paradigm of body condition: a critical attenuate high-fat diet-induced metabolic syndrome. Diabetes.
reappraisal of current methods based on mass and length. Functional 2015;64(8):2847-2858.
Ecology. 2010;24(6):1323-1332. 79. Meng Q, Lai YC, Kelly NJ, et al. Development of a mouse model of
59. Lutz TA, Woods SC. Overview of animal models of obesity. Curr metabolic syndrome, pulmonary hypertension, and heart failure with
Protoc Pharmacol. 2012;58(1):5-61. Chapter 5:Unit5 61 preserved ejection fraction. Am J Respir Cell Mol Biol. 2017;56(4):
60. Marques C, Meireles M, Norberto S, et al. High-fat diet-induced 497-505.
obesity rat model: a comparison between Wistar and Sprague– 80. Malik VS, Popkin BM, Bray GA, Despres JP, Hu FB. Sugar-
Dawley Rat. Adipocyte. 2016;5(1):11-21. sweetened beverages, obesity, type 2 diabetes mellitus, and cardio-
61. Cheng HS, Ton SH, Phang SCW, Tan JBL, Abdul KK. Increased vascular disease risk. Circulation. 2010;121(11):1356-1364.
susceptibility of post-weaning rats on high-fat diet to metabolic 81. Tappy L, Le KA. Metabolic effects of fructose and the worldwide
syndrome. J Adv Res. 2017;8(6):743-752. increase in obesity. Physiol Rev. 2010;90(1):23-46.
62. Alexander J, Chang GQ, Dourmashkin JT, Leibowitz SF. Distinct 82. Bocarsly ME, Powell ES, Avena NM, Hoebel BG. High-fructose corn
phenotypes of obesity-prone AKR/J, DBA2J and C57BL/6J mice syrup causes characteristics of obesity in rats: increased body
compared to control strains. Int J Obes (Lond). 2006;30(1):50-59. weight, body fat and triglyceride levels. Pharmacol Biochem Behav.
63. Lee YS, Li P, Huh JY, et al. Inflammation is necessary for long-term 2010;97(1):101-106.
but not short-term high-fat diet-induced insulin resistance. Diabetes. 83. Tillman EJ, Morgan DA, Rahmouni K, Swoap SJ. Three months of
2011;60(10):2474-2483. high-fructose feeding fails to induce excessive weight gain or leptin
64. Turner N, Kowalski GM, Leslie SJ, et al. Distinct patterns of tissue- resistance in mice. PLoS ONE. 2014;9(9):e107206.
specific lipid accumulation during the induction of insulin resistance 84. Sumiyoshi M, Sakanaka M, Kimura Y. Chronic intake of high-fat and
in mice by high-fat feeding. Diabetologia. 2013;56(7):1638-1648. high-sucrose diets differentially affects glucose intolerance in mice.
65. Montgomery MK, Hallahan NL, Brown SH, et al. Mouse strain- J Nutr. 2006;136(3):582-587.
dependent variation in obesity and glucose homeostasis in response 85. Yang ZH, Miyahara H, Takeo J, Katayama M. Diet high in fat and
to high-fat feeding. Diabetologia. 2013;56(5):1129-1139. sucrose induces rapid onset of obesity-related metabolic syndrome
66. Martin-Cordero L, Galvez I, Hinchado MD, Ortega E. β2 adrenergic partly through rapid response of genes involved in lipogenesis,
regulation of the phagocytic and microbicide capacity of macro- insulin signalling and inflammation in mice. Diabetol Metab Syndr.
phages from obese and lean mice: effects of exercise. Nutrients. 2012;4(1):32.
2019;11(11):2721. 86. Zhang L, Song H, Ge Y, Ji G, Yao Z. Temporal relationship between
67. Galvez I, Martin-Cordero L, Hinchado MD, Alvarez-Barrientos A, diet-induced steatosis and onset of insulin/leptin resistance in male
Ortega E. Anti-inflammatory effect of β2 adrenergic stimulation on Wistar rats. PLoS ONE. 2015;10(2):e0117008.
circulating monocytes with a pro-inflammatory state in high-fat diet- 87. Roberts CK, Hevener AL, Barnard RJ. Metabolic syndrome and
induced obesity. Brain Behav Immun. 2019;80:564-572. insulin resistance: underlying causes and modification by exercise
68. Levin BE, Dunn-Meynell AA, Balkan B, Keesey RE. Selective breed- training. Compr Physiol. 2013;3(1):1-58.
ing for diet-induced obesity and resistance in Sprague–Dawley rats. 88. Kassi E, Pervanidou P, Kaltsas G, Chrousos G. Metabolic syndrome:
Am J Physiol. 1997;273(2 Pt 2):R725-R730. definitions and controversies. BMC Med. 2011;9:48.
69. Souza GF, Solon C, Nascimento LF, et al. Defective regulation of 89. Alberti KG, Eckel RH, Grundy SM, et al. Harmonizing the metabolic
POMC precedes hypothalamic inflammation in diet-induced obesity. syndrome: a joint interim statement of the International Diabetes
Sci Rep. 2016;6:29290. Federation Task Force on Epidemiology and Prevention; National
70. Chang S, Graham B, Yakubu F, Lin D, Peters JC, Hill JO. Metabolic Heart, Lung, and Blood Institute; American Heart Association; World
differences between obesity-prone and obesity-resistant rats. Heart Federation; International Atherosclerosis Society; and Interna-
Am J Physiol. 1990;259(6 Pt 2):R1103-R1110. tional Association for the Study of Obesity. Circulation. 2009;
71. MacLean PS, Higgins JA, Johnson GC, et al. Enhanced metabolic effi- 120(16):1640-1645.
ciency contributes to weight regain after weight loss in obesity- 90. Nolan PB, Carrick-Ranson G, Stinear JW, Reading SA, Dalleck LC.
prone rats. Am J Physiol Regul Integr Comp Physiol. 2004;287(6): Prevalence of metabolic syndrome and metabolic syndrome compo-
R1306-R1315. nents in young adults: a pooled analysis. Prev Med Rep. 2017;7:
72. Aziz AA, Kenney LS, Goulet B, Abdel-Aal el S. Dietary starch type 211-215.
affects body weight and glycemic control in freely fed but not 91. Saklayen MG. The global epidemic of the metabolic syndrome. Curr
energy-restricted obese rats. J Nutr. 2009;139(10):1881-1889. Hypertens Rep. 2018;20(2):12.
73. Belobrajdic DP, King RA, Christophersen CT, Bird AR. Dietary resis- 92. Gurka MJ, Filipp SL, DeBoer MD. Geographical variation in the
tant starch dose-dependently reduces adiposity in obesity-prone prevalence of obesity, metabolic syndrome, and diabetes among US
and obesity-resistant male rats. Nutr Metab (Lond). 2012;9(1):93. adults. Nutr Diabetes. 2018;8(1):14.
PREGUIÇA ET AL. 25

93. Sigit FS, Tahapary DL, Trompet S, et al. The prevalence of metabolic 113. Moreno-Fernandez S, Garces-Rimon M, Vera G, Astier J,
syndrome and its association with body fat distribution in middle- Landrier JF, Miguel M. High fat/high glucose diet induces metabolic
aged individuals from Indonesia and the Netherlands: a cross- syndrome in an experimental rat model. Nutrients. 2018;10(10):
sectional analysis of two population-based studies. Diabetol Metab 1502.
Syndr. 2020;12:2. 114. Masi LN, Martins AR, Crisma AR, et al. Combination of a high-fat
94. Lind L. Genome-wide association study of the metabolic syndrome diet with sweetened condensed milk exacerbates inflammation and
in UK biobank. Metab Syndr Relat Disord. 2019;17(10):505-511. insulin resistance induced by each separately in mice. Sci Rep. 2017;
95. Prasad G, Bandesh K, Giri AK, et al. Genome-wide association study 7(1):3937.
of metabolic syndrome reveals primary genetic variants at CETP 115. Hostalek U. Global epidemiology of prediabetes—present and future
locus in Indians. Biomolecules. 2019;9(8):321. perspectives. Clin Diabetes Endocrinol. 2019;5:5.
96. Wei ZY, Liu JJ, Zhan XM, Feng HM, Zhang YY. Dietary patterns and 116. Tabak AG, Herder C, Rathmann W, Brunner EJ, Kivimaki M. Predia-
the risk of metabolic syndrome in Chinese adults: a population- betes: a high-risk state for diabetes development. Lancet. 2012;
based cross-sectional study. Public Health Nutr. 2018;21(13):2409- 379(9833):2279-2290.
2416. 117. Wilson ML. Prediabetes: beyond the borderline. Nurs Clin North Am.
97. Wang LX, Gurka MJ, Deboer MD. Metabolic syndrome severity and 2017;52(4):665-677.
lifestyle factors among adolescents. Minerva Pediatr. 2018;70(5): 118. Faerch K, Borch-Johnsen K, Holst JJ, Vaag A. Pathophysiology and
467-475. aetiology of impaired fasting glycaemia and impaired glucose toler-
98. Di Daniele N, Noce A, Vidiri MF, et al. Impact of Mediterranean diet ance: does it matter for prevention and treatment of type 2 diabe-
on metabolic syndrome, cancer and longevity. Oncotarget. 2017; tes? Diabetologia. 2009;52(9):1714-1723.
8(5):8947-8979. 119. American Diabetes A. 2. Classification and diagnosis of diabetes:
99. Wong SK, Chin KY, Suhaimi FH, Fairus A, Ima-Nirwana S. Animal standards of medical care in diabetes-2019. Diabetes Care. 2019;
models of metabolic syndrome: a review. Nutr Metab (Lond). 2016; 42(Suppl 1):S13-S28.
13:65. 120. Ausk KJ, Boyko EJ, Ioannou GN. Insulin resistance predicts mortality
100. Wanrooy BJ, Kumar KP, Wen SW, Qin CX, Ritchie RH, Wong CHY. in nondiabetic individuals in the U.S. Diabetes Care. 2010;33(6):
Distinct contributions of hyperglycemia and high-fat feeding in met- 1179-1185.
abolic syndrome-induced neuroinflammation. J Neuroinflammation. 121. Hedblad B, Nilsson P, Engstrom G, Berglund G, Janzon L. Insulin
2018;15(1):293. resistance in non-diabetic subjects is associated with increased inci-
101. Mohammad Saifur Rohman ML, Nugroho DA, Nashi W, dence of myocardial infarction and death. Diabet Med. 2002;19(6):
Nugraheini NIP, Sardjono TW. Development of an experimental 470-475.
model of metabolic syndrome in Sprague Dawley rat. Res J Life Sci. 122. Abdul-Ghani MA, DeFronzo RA. Pathophysiology of prediabetes.
2017;4:76-86. Curr Diab Rep. 2009;9(3):193-199.
102. Heydemann A. An overview of murine high fat diet as a model for 123. Saravanan N, Patil MA, Kumar PU, Suryanarayana P, Reddy GB. Die-
type 2 diabetes mellitus. J Diabetes Res. 2016;2016:2902351. tary ginger improves glucose dysregulation in a long-term high-fat
103. Hu S, Wang L, Yang D, et al. Dietary fat, but not protein or carbohy- high-fructose fed prediabetic rat model. Indian J Exp Biol. 2017;
drate, regulates energy intake and causes adiposity in mice. Cell 55(3):142-150.
Metab. 2018;28(3):415-431. e414 124. Tanajak P, Sa-Nguanmoo P, Apaijai N, et al. Comparisons of cardi-
104. Ghezzi AC, Cambri LT, Botezelli JD, Ribeiro C, Dalia RA, de oprotective efficacy between fibroblast growth factor 21 and
Mello MA. Metabolic syndrome markers in wistar rats of different dipeptidyl peptidase-4 inhibitor in prediabetic rats. Cardiovasc Ther.
ages. Diabetol Metab Syndr. 2012;4(1):16. 2017;35(4):e12263.
105. Nunes-Souza V, Cesar-Gomes CJ, Da Fonseca LJ, Guedes Gda S, 125. Sweazea KL, Lekic M, Walker BR. Comparison of mechanisms
Smaniotto S, Rabelo LA. Aging increases susceptibility to high fat involved in impaired vascular reactivity between high sucrose and
diet-induced metabolic syndrome in C57BL/6 mice: improvement in high fat diets in rats. Nutr Metab (Lond). 2010;7:48.
glycemic and lipid profile after antioxidant therapy. Oxid Med Cell 126. Mosser RE, Maulis MF, Moulle VS, et al. High-fat diet-induced β-cell
Longev. 2016;2016:1987960. proliferation occurs prior to insulin resistance in C57Bl/6J male
106. Lasker S, Rahman MM, Parvez F, et al. High-fat diet-induced meta- mice. Am J Physiol Endocrinol Metab. 2015;308(7):E573-E582.
bolic syndrome and oxidative stress in obese rats are ameliorated by 127. Small L, Brandon AE, Turner N, Cooney GJ. Modeling insulin resis-
yogurt supplementation. Sci Rep. 2019;9(1):20026. tance in rodents by alterations in diet: what have high-fat and high-
107. Oron-Herman M, Kamari Y, Grossman E, et al. Metabolic syndrome: calorie diets revealed? Am J Physiol Endocrinol Metab. 2018;314(3):
comparison of the two commonly used animal models. E251-E265.
Am J Hypertens. 2008;21(9):1018-1022. 128. Yang T, Xu WJ, York H, Liang NC. Diet choice patterns in rodents
108. Ritze Y, Bardos G, D'Haese JG, et al. Effect of high sugar intake on depend on novelty of the diet, exercise, species, and sex. Physiol
glucose transporter and weight regulating hormones in mice and Behav. 2017;176:149-158.
humans. PLoS ONE. 2014;9(7):e101702. 129. Catta-Preta M, Martins MA, Cunha Brunini TM, Mendes-
109. Montgomery MK, Fiveash CE, Braude JP, et al. Disparate metabolic Ribeiro AC, Mandarim-de-Lacerda CA, Aguila MB. Modulation
response to fructose feeding between different mouse strains. Sci of cytokines, resistin, and distribution of adipose tissue in
Rep. 2015;5:18474. C57BL/6 mice by different high-fat diets. Nutrition. 2012;28(2):
110. Aslam M, Madhu SV. Development of metabolic syndrome in high- 212-219.
sucrose diet fed rats is not associated with decrease in adiponectin 130. Abdel-Hamid AAM, Firgany AEL. Correlation between pancreatic
levels. Endocrine. 2017;58(1):59-65. mast cells and the low grade inflammation in adipose tissue of
111. Aguilera-Mendez A, Hernandez-Equihua MG, Rueda-Rocha AC, et al. experimental prediabetes. Acta Histochem. 2019;121(1):35-42.
Protective effect of supplementation with biotin against high-fruc- 131. Winzell MS, Ahren B. The high-fat diet-fed mouse: a model for
tose-induced metabolic syndrome in rats. Nutr Res. 2018;57:86-96. studying mechanisms and treatment of impaired glucose tolerance
112. Nunes S, Soares E, Fernandes J, et al. Early cardiac changes in a rat and type 2 diabetes. Diabetes. 2004;53(Suppl 3):S215-S219.
model of prediabetes: brain natriuretic peptide overexpression 132. Chen GC, Huang CY, Chang MY, et al. Two unhealthy dietary habits
seems to be the best marker. Cardiovasc Diabetol. 2013;12:44. featuring a high fat content and a sucrose-containing beverage
26 PREGUIÇA ET AL.

intake, alone or in combination, on inducing metabolic syndrome in 152. van den Brom CE, Boly CA, Bulte CSE, van den Akker RFP,
Wistar rats and C57BL/6J mice. Metabolism. 2011;60(2):155-164. Kwekkeboom RFJ, Loer SA, Boer C, Bouwman RA. Myocardial Per-
133. Thresher JS, Podolin DA, Wei Y, Mazzeo RS, Pagliassotti MJ. Com- fusion and Function Are Distinctly Altered by Sevoflurane Anesthe-
parison of the effects of sucrose and fructose on insulin action and sia in Diet-Induced Prediabetic Rats. Journal of Diabetes Research.
glucose tolerance. Am J Physiol Regul Integr Comp Physiol. 2000; 2016;2016:1–9. https://doi.org/10.1155/2016/5205631
279(4):R1334-R1340. 153. Vatandoust N, Rami F, Salehi AR, et al. Novel high-fat diet formula-
134. Togo J, Hu S, Li M, Niu C, Speakman JR. Impact of dietary sucrose tion and streptozotocin treatment for induction of prediabetes and
on adiposity and glucose homeostasis in C57BL/6J mice depends on type 2 diabetes in rats. Adv Biomed Res. 2018;7:107.
mode of ingestion: liquid or solid. Mol Metab. 2019;27:22-32. 154. Saeedi P, Petersohn I, Salpea P, Malanda B, Karuranga S, Unwin N,
135. Surwit RS, Feinglos MN, Rodin J, et al. Differential effects of fat and Colagiuri S, Guariguata L, Motala AA, Ogurtsova K, Shaw JE,
sucrose on the development of obesity and diabetes in C57BL/6J Bright D, Williams R. Global and regional diabetes prevalence esti-
and A/J mice. Metabolism. 1995;44(5):645-651. mates for 2019 and projections for 2030 and 2045: Results from
136. Burgeiro A, Cerqueira MG, Varela-Rodriguez BM, et al. Glucose and the International Diabetes Federation Diabetes Atlas, 9th edition.
lipid dysmetabolism in a rat model of prediabetes induced by a high- Diabetes Research and Clinical Practice. 2019;157:107843. https://
sucrose diet. Nutrients. 2017;9(6):638. doi.org/10.1016/j.diabres.2019.107843
137. Kanazawa M, Xue CY, Kageyama H, et al. Effects of a high-sucrose 155. Bowe JE, Franklin ZJ, Hauge-Evans AC, King AJ, Persaud SJ,
diet on body weight, plasma triglycerides, and stress tolerance. Nutr Jones PM. Metabolic phenotyping guidelines: assessing glucose
Rev. 2003;61(5 Pt 2):S27-S33. homeostasis in rodent models. J Endocrinol. 2014;222(3):G13-G25.
138. Alves MRP, Boia R, Campos EJ, et al. Subtle thinning of retinal layers 156. Andrikopoulos S, Blair AR, Deluca N, Fam BC, Proietto J. Evaluating
without overt vascular and inflammatory alterations in a rat model the glucose tolerance test in mice. Am J Physiol Endocrinol Metab.
of prediabetes. Mol Vis. 2018;24:353-366. 2008;295(6):E1323-E1332.
139. Soares E, Prediger RD, Nunes S, et al. Spatial memory impairments 157. Heikkinen S, Argmann CA, Champy MF, Auwerx J. Evaluation of
in a prediabetic rat model. Neuroscience. 2013;250:565-577. glucose homeostasis. Curr Protoc Mol Biol. 2007;77(1): Chapter 29:
140. Pham N, Dhar A, Khalaj S, Desai K, Taghibiglou C. Down regulation Unit 29B.3.
of brain cellular prion protein in an animal model of insulin resis- 158. Wang Z, Yang Y, Xiang X, Zhu Y, Men J, He M. Estimation of the
tance: possible implication in increased prevalence of stroke in pre- normal range of blood glucose in rats. Wei Sheng Yan Jiu. 2010;
diabetics/diabetics. Biochem Biophys Res Commun. 2014;448(2): 39(2):133-137. 142
151-156. 159. Vander Werf R, Walter C, Bietiger W, et al. Beneficial effects of
141. Alzamendi A, Giovambattista A, Raschia A, et al. Fructose-rich diet- cherry consumption as a dietary intervention for metabolic, hepatic
induced abdominal adipose tissue endocrine dysfunction in normal and vascular complications in type 2 diabetic rats. Cardiovasc
male rats. Endocrine. 2009;35(2):227-232. Diabetol. 2018;17(1):104.
142. Tappy L, Le KA, Tran C, Paquot N. Fructose and metabolic diseases: 160. Clee SM, Attie AD. The genetic landscape of type 2 diabetes in mice.
new findings, new questions. Nutrition. 2010;26(11–12):1044-1049. Endocr Rev. 2007;28(1):48-83.
143. Tran LT, Yuen VG, McNeill JH. The fructose-fed rat: a review on the 161. Leiter EH. Selecting the “right” mouse model for metabolic syn-
mechanisms of fructose-induced insulin resistance and hyperten- drome and type 2 diabetes research. Methods Mol Biol. 2009;560:
sion. Mol Cell Biochem. 2009;332(1–2):145-159. 1-17.
144. Miranda CA, Schonholzer TE, Kloppel E, et al. Repercussions of low 162. Benede-Ubieto R, Estevez-Vazquez O, Ramadori P, Cubero FJ,
fructose-drinking water in male rats. An Acad Bras Cienc. 2019;91(1): Nevzorova YA. Guidelines and considerations for metabolic
e20170705. tolerance tests in mice. Diabetes Metab Syndr Obes. 2020;13:
145. Francini F, Massa ML, Polo MP, Villagarcia H, Castro MC, 439-450.
Gagliardino JJ. Control of liver glucokinase activity: a potential new 163. Kale VP, Joshi GS, Gohil PB, Jain MR. Effect of fasting duration on
target for incretin hormones? Peptides. 2015;74:57-63. clinical pathology results in Wistar rats. Vet Clin Pathol. 2009;38(3):
146. Kowalski GM, Kraakman MJ, Mason SA, Murphy AJ, Bruce CR. Res- 361-366.
olution of glucose intolerance in long-term high-fat, high-sucrose- 164. Srinivasan K, Ramarao P. Animal models in type 2 diabetes research:
fed mice. J Endocrinol. 2017;233(3):269-279. an overview. Indian J Med Res. 2007;125(3):451-472.
147. Gamede M, Mabuza L, Ngubane P, Khathi A. The effects of plant- 165. Chatzigeorgiou A, Halapas A, Kalafatakis K, Kamper E. The use of
derived oleanolic acid on selected parameters of glucose homeosta- animal models in the study of diabetes mellitus. In Vivo. 2009;23(2):
sis in a diet-induced pre-diabetic rat model. Molecules. 2018; 245-258.
23(4):794. 166. Kuwabara WMT, Panveloski-Costa AC, Yokota CNF, et al. Compari-
148. Panchal SK, Brown L. Rodent Models for Metabolic Syndrome son of Goto–Kakizaki rats and high fat diet-induced obese rats: are
Research. Journal of Biomedicine and Biotechnology. 2011;2011 they reliable models to study type 2 diabetes mellitus? PLoS ONE.
1–14. https://doi.org/10.1155/2011/351982 2017;12(12):e0189622.
149. Lomba A, Milagro FI, Garcia-Diaz DF, Marti A, Campion J, 167. Stark AH, Timar B, Madar Z. Adaptation of Sprague Dawley rats to
Martinez JA. Obesity induced by a pair-fed high fat sucrose diet: long-term feeding of high fat or high fructose diets. Eur J Nutr.
methylation and expression pattern of genes related to energy 2000;39(5):229-234.
homeostasis. Lipids Health Dis. 2010;9:60. 168. Patel J, Iyer A, Brown L. Evaluation of the chronic complications of
150. Chun MR, Lee YJ, Kim KH, et al. Differential effects of high- diabetes in a high fructose diet in rats. Indian J Biochem Biophys.
carbohydrate and high-fat diet composition on muscle insulin resis- 2009;46(1):66-72.
tance in rats. J Korean Med Sci. 2010;25(7):1053-1059. 169. Rohlmann A, Basli K, Bergmann G. Analysis of the tension of the
151. Sato A, Kawano H, Notsu T, et al. Antiobesity effect of elbow joint before and after resection of the radius head. Biomed
eicosapentaenoic acid in high-fat/high-sucrose diet-induced obesity: Tech (Berl). 1986;31(10):230-239.
importance of hepatic lipogenesis. Diabetes. 2010;59(10):2495- 170. Matheus VA, Monteiro L, Oliveira RB, Maschio DA, Collares-
2504. Buzato CB. Butyrate reduces high-fat diet-induced metabolic
PREGUIÇA ET AL. 27

alterations, hepatic steatosis and pancreatic beta cell and intestinal expression in different models of diabetic C57BL/6 mice. Diabetol
barrier dysfunctions in prediabetic mice. Exp Biol Med (Maywood). Metab Syndr. 2019;11:65.
2017;242(12):1214-1226. 189. Fang X, Xia T, Xu F, Wu H, Ma Z, Zhao X, Gu X. Isoflurane aggra-
171. Surwit RS, Kuhn CM, Cochrane C, McCubbin JA, Feinglos MN. Diet- vates peripheral and central insulin resistance in high-fat diet/-
induced type II diabetes in C57BL/6J mice. Diabetes. 1988;37(9): streptozocin-induced type 2 diabetic mice. Brain Research. 2020;
1163-1167. 1727:146511. https://doi.org/10.1016/j.brainres.2019.146511
172. Guo A, Daniels NA, Thuma J, McCall KD, Malgor R, Schwartz FL. 190. Srinivasan K, Viswanad B, Asrat L, Kaul CL, Ramarao P. Combination
Diet is critical for prolonged glycemic control after short-term insulin of high-fat diet-fed and low-dose streptozotocin-treated rat: a
treatment in high-fat diet-induced type 2 diabetic male mice. PLoS model for type 2 diabetes and pharmacological screening. Pharmacol
ONE. 2015;10(1):e0117556. Res. 2005;52(4):313-320.
173. Kleindienst A, Battault S, Belaidi E, et al. Exercise does not activate 191. Barriere DA, Noll C, Roussy G, et al. Combination of high-fat/high-
the β3 adrenergic receptor-eNOS pathway, but reduces inducible fructose diet and low-dose streptozotocin to model long-term type-
NOS expression to protect the heart of obese diabetic mice. Basic 2 diabetes complications. Sci Rep. 2018;8(1):424.
Res Cardiol. 2016;111(4):40. 192. Obrosov A, Shevalye H, Coppey LJ, Yorek MA. Effect of tempol on
174. Winters WD, Huo YS, Yao DL. Inhibition of the progression of type peripheral neuropathy in diet-induced obese and high-fat fed/low-
2 diabetes in the C57BL/6J mouse model by an anti-diabetes herbal dose streptozotocin-treated C57Bl6/J mice. Free Radic Res. 2017;
formula. Phytother Res. 2003;17(6):591-598. 51(4):360-367.
175. Chalkley SM, Hettiarachchi M, Chisholm DJ, Kraegen EW. Long- 193. Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L, Wymer M.
term high-fat feeding leads to severe insulin resistance but not dia- Global epidemiology of nonalcoholic fatty liver disease-Meta-
betes in Wistar rats. Am J Physiol Endocrinol Metab. 2002;282(6): analytic assessment of prevalence, incidence, and outcomes.
E1231-E1238. Hepatology. 2016;64(1):73-84.
176. Peterson R, Shaw WN, Neel MA, Little LA, Eichberg J. Zucker dia- 194. Cohen JC, Horton JD, Hobbs HH. Human fatty liver disease: old
betic fatty rat as a model for non-insulin-dependent diabetes questions and new insights. Science. 2011;332(6037):1519-1523.
mellitus. ILAR News. 1990;32:16-19. 195. Nakamura A, Terauchi Y. Lessons from mouse models of high-fat
177. Corsetti JP, Sparks JD, Peterson RG, Smith RL, Sparks CE. Effect of diet-induced NAFLD. Int J Mol Sci. 2013;14(11):21240-21257.
dietary fat on the development of non-insulin dependent diabetes 196. Ito M, Suzuki J, Tsujioka S, et al. Longitudinal analysis of murine
mellitus in obese Zucker diabetic fatty male and female rats. Athero- steatohepatitis model induced by chronic exposure to high-fat diet.
sclerosis. 2000;148(2):231-241. Hepatol Res. 2007;37(1):50-57.
178. Ferreira L, Teixeira-de-Lemos E, Pinto F, Parada B, Mega C, 197. Gauthier MS, Favier R, Lavoie JM. Time course of the development
Vala H, Pinto R, Garrido P, Sereno J, Fernandes R, Santos P, of non-alcoholic hepatic steatosis in response to high-fat diet-
Velada I, Melo A, Nunes S, Teixeira F, Reis F. Effects of Sitagliptin induced obesity in rats. Br J Nutr. 2006;95(2):273-281.
Treatment on Dysmetabolism, Inflammation, and Oxidative Stress 198. Omagari K, Kato S, Tsuneyama K, et al. Effects of a long-term high-
in an Animal Model of Type 2 Diabetes (ZDF Rat). Mediators of fat diet and switching from a high-fat to low-fat, standard diet on
Inflammation. 2010;2010:1–11. https://doi.org/10.1155/2010/ hepatic fat accumulation in Sprague–Dawley rats. Dig Dis Sci. 2008;
592760 53(12):3206-3212.
179. Uozumi K, Ohno N, Ishizuka K, Iwahashi M, Hanada S, Arima T. 199. McCuskey RS, Ito Y, Robertson GR, McCuskey MK, Perry M,
Adult T-cell leukaemia in patients with OKT4 epitope deficiency. Br Farrell GC. Hepatic microvascular dysfunction during evolution of
J Haematol. 1991;79(4):651-652. dietary steatohepatitis in mice. Hepatology. 2004;40(2):386-393.
180. Kalaki-Jouybari F, Shanaki M, Delfan M, Gorgani-Firouzjae S, 200. Machado MV, Michelotti GA, Xie G, et al. Correction: Mouse models
Khakdan S. High-intensity interval training (HIIT) alleviated NAFLD of diet-induced nonalcoholic steatohepatitis reproduce the hetero-
feature via miR-122 induction in liver of high-fat high-fructose diet geneity of the human disease. PLoS ONE. 2015;10(6):e0132315.
induced diabetic rats. Arch Physiol Biochem. 2018;1-8. 201. Wolf MJ, Adili A, Piotrowitz K, et al. Metabolic activation of
181. Balakumar M, Raji L, Prabhu D, et al. High-fructose diet is as intrahepatic CD8+ T cells and NKT cells causes nonalcoholic
detrimental as high-fat diet in the induction of insulin resistance and steatohepatitis and liver cancer via cross-talk with hepatocytes. Can-
diabetes mediated by hepatic/pancreatic endoplasmic reticulum cer Cell. 2014;26(4):549-564.
(ER) stress. Mol Cell Biochem. 2016;423(1–2):93-104. 202. Miura K, Kodama Y, Inokuchi S, et al. Toll-like receptor 9 promotes
182. Skovso S. Modeling type 2 diabetes in rats using high fat diet and steatohepatitis by induction of interleukin-1β in mice. Gastroenterol-
streptozotocin. J Diabetes Investig. 2014;5(4):349-358. ogy. 2010;139(1):323-334. e327
183. Mansor LS, Gonzalez ER, Cole MA, et al. Cardiac metabolism in a 203. Uto H, Nakanishi C, Ido A, et al. The peroxisome proliferator-
new rat model of type 2 diabetes using high-fat diet with low dose activated receptor-gamma agonist, pioglitazone, inhibits fat accumu-
streptozotocin. Cardiovasc Diabetol. 2013;12:136. lation and fibrosis in the livers of rats fed a choline-deficient, L-
184. Guo XX, Wang Y, Wang K, Ji BP, Zhou F. Stability of a type amino acid-defined diet. Hepatol Res. 2005;32(4):235-242.
2 diabetes rat model induced by high-fat diet feeding with low-dose 204. Kamada Y, Matsumoto H, Tamura S, et al. Hypoadiponectinemia
streptozotocin injection. J Zhejiang Univ Sci B. 2018;19(7):559-569. accelerates hepatic tumor formation in a nonalcoholic
185. Chen M, Xiao D, Liu W, et al. Intake of Ganoderma lucidum polysac- steatohepatitis mouse model. J Hepatol. 2007;47(4):556-564.
charides reverses the disturbed gut microbiota and metabolism in 205. Matsumoto M, Hada N, Sakamaki Y, et al. An improved mouse
type 2 diabetic rats. Int J Biol Macromol. 2019;155:890-902. model that rapidly develops fibrosis in non-alcoholic steatohepatitis.
186. Lee JY, Jeong EA, Kim KE, et al. TonEBP/NFAT5 haploinsufficiency Int J Exp Pathol. 2013;94(2):93-103.
attenuates hippocampal inflammation in high-fat diet/ 206. Matsuzawa N, Takamura T, Kurita S, et al. Lipid-induced oxidative
streptozotocin-induced diabetic mice. Sci Rep. 2017;7(1):7837. stress causes steatohepatitis in mice fed an atherogenic diet.
187. Leguina-Ruzzi A, Ortiz R, Velarde V. The streptozotocin-high fat diet Hepatology. 2007;46(5):1392-1403.
induced diabetic mouse model exhibits severe skin damage and 207. Charlton M, Krishnan A, Viker K, et al. Fast food diet mouse: novel
alterations in local lipid mediators. Biom J. 2018;41(5):328-332. small animal model of NASH with ballooning, progressive fibrosis,
188. Goodarzi G, Shirgir A, Alavi S, Khoshi A. Effect of insulin-glucose and high physiological fidelity to the human condition. Am J Physiol
metabolism compared with obesity on adipose omentin gene Gastrointest Liver Physiol. 2011;301(5):G825-G834.
28 PREGUIÇA ET AL.

208. Mamikutty N, Thent ZC, Haji SF. Fructose-drinking water induced 228. Ter Horst KW, Gilijamse PW, de Weijer BA, et al. Sexual dimorphism
nonalcoholic fatty liver disease and ultrastructural alteration of in hepatic, adipose tissue, and peripheral tissue insulin sensitivity in
hepatocyte mitochondria in male wistar rat. Biomed Res Int. 2015; obese humans. Front Endocrinol (Lausanne). 2015;6:182.
2015:895961. 229. Kautzky-Willer A, Harreiter J, Pacini G. Sex and gender differences
209. Spruss A, Kanuri G, Wagnerberger S, Haub S, Bischoff SC, in risk, pathophysiology and complications of type 2 diabetes
Bergheim I. Toll-like receptor 4 is involved in the development of mellitus. Endocr Rev. 2016;37(3):278-316.
fructose-induced hepatic steatosis in mice. Hepatology. 2009;50(4): 230. Macotela Y, Boucher J, Tran TT, Kahn CR. Sex and depot differences
1094-1104. in adipocyte insulin sensitivity and glucose metabolism. Diabetes.
210. Asgharpour A, Cazanave SC, Pacana T, et al. A diet-induced animal 2009;58(4):803-812.
model of non-alcoholic fatty liver disease and hepatocellular cancer. 231. Lee MJ, Wu Y, Fried SK. Adipose tissue heterogeneity: implication
J Hepatol. 2016;65(3):579-588. of depot differences in adipose tissue for obesity complications. Mol
211. Nishikawa S, Yasoshima A, Doi K, Nakayama H, Uetsuka K. Involve- Aspects Med. 2013;34(1):1-11.
ment of sex, strain and age factors in high fat diet-induced obesity 232. Stubbins RE, Holcomb VB, Hong J, Nunez NP. Estrogen modulates
in C57BL/6J and BALB/cA mice. Exp Anim. 2007;56(4):263-272. abdominal adiposity and protects female mice from obesity and
212. Choi JY, McGregor RA, Kwon EY, et al. The metabolic response to a impaired glucose tolerance. Eur J Nutr. 2012;51(7):861-870.
high-fat diet reveals obesity-prone and -resistant phenotypes in 233. Chen X, McClusky R, Chen J, et al. The number of X chromosomes
mice with distinct mRNA-seq transcriptome profiles. Int J Obes causes sex differences in adiposity in mice. PLoS Genet. 2012;8(5):
(Lond). 2016;40(9):1452-1460. e1002709.
213. Svenson KL, Van Smith R, Magnani PA, et al. Multiple trait measure- 234. Link JC, Hasin-Brumshtein Y, Cantor RM, et al. Diet, gonadal sex,
ments in 43 inbred mouse strains capture the phenotypic diversity and sex chromosome complement influence white adipose tissue
characteristic of human populations. J Appl Physiol (1985). 2007; miRNA expression. BMC Genomics. 2017;18(1):89.
102(6):2369-2378. 235. Jackman MR, Kramer RE, MacLean PS, Bessesen DH. Trafficking of
214. Kebede MA, Attie AD. Insights into obesity and diabetes at the dietary fat in obesity-prone and obesity-resistant rats. Am J Physiol
intersection of mouse and human genetics. Trends Endocrinol Metab. Endocrinol Metab. 2006;291(5):E1083-E1091.
2014;25(10):493-501. 236. Jackman MR, MacLean PS, Bessesen DH. Energy expenditure in
215. Tuttle AH, Philip VM, Chesler EJ, Mogil JS. Comparing phenotypic obesity-prone and obesity-resistant rats before and after the intro-
variation between inbred and outbred mice. Nat Methods. 2018; duction of a high-fat diet. Am J Physiol Regul Integr Comp Physiol.
15(12):994-996. 2010;299(4):R1097-R1105.
216. Ding C, Guo J, Su Z. The status of research into resistance to diet- 237. Nadal-Casellas A, Proenza AM, Llado I, Gianotti M. Sex-dependent
induced obesity. Horm Metab Res. 2015;47(6):404-410. differences in rat hepatic lipid accumulation and insulin sensitivity in
217. Smith RL, Soeters MR, Wust RCI, Houtkooper RH. Metabolic flexi- response to diet-induced obesity. Biochem Cell Biol. 2012;90(2):
bility as an adaptation to energy resources and requirements in 164-172.
health and disease. Endocr Rev. 2018;39(4):489-517. 238. Medrikova D, Jilkova ZM, Bardova K, Janovska P, Rossmeisl M,
218. Bardova K, Horakova O, Janovska P, et al. Early differences in meta- Kopecky J. Sex differences during the course of diet-induced obe-
bolic flexibility between obesity-resistant and obesity-prone mice. sity in mice: adipose tissue expandability and glycemic control. Int J
Biochimie. 2016;124:163-170. Obes (Lond). 2012;36(2):262-272.
219. Poret JM, Souza-Smith F, Marcell SJ, et al. High fat diet consump- 239. Tokuyama Y, Sturis J, DePaoli AM, et al. Evolution of β-cell dysfunc-
tion differentially affects adipose tissue inflammation and adipocyte tion in the male Zucker diabetic fatty rat. Diabetes. 1995;44(12):
size in obesity-prone and obesity-resistant rats. Int J Obes (Lond). 1447-1457.
2018;42(3):535-541. 240. McCullough LD, de Vries GJ, Miller VM, Becker JB, Sandberg K,
220. Hong J, Stubbins RE, Smith RR, Harvey AE, Nunez NP. Differential McCarthy MM. NIH initiative to balance sex of animals in preclinical
susceptibility to obesity between male, female and ovariectomized studies: generative questions to guide policy, implementation, and
female mice. Nutr J. 2009;8:11. metrics. Biol Sex Differ. 2014;5:15.
221. Giles ED, Jackman MR, MacLean PS. Modeling diet-induced obesity 241. Mauvais-Jarvis F, Arnold AP, Reue K. A guide for the design of pre-
with obesity-prone rats: implications for studies in females. Front clinical studies on sex differences in metabolism. Cell Metab. 2017;
Nutr. 2016;3:50. 25(6):1216-1230.
222. Franconi F, Seghieri G, Canu S, Straface E, Campesi I, Malorni W. 242. Houtkooper RH, Argmann C, Houten SM, et al. The metabolic foot-
Are the available experimental models of type 2 diabetes appropri- print of aging in mice. Sci Rep. 2011;1:134.
ate for a gender perspective? Pharmacol Res. 2008;57(1):6-18. 243. van der Heijden RA, Bijzet J, Meijers WC, et al. Obesity-induced
223. Parks BW, Sallam T, Mehrabian M, et al. Genetic architecture of chronic inflammation in high fat diet challenged C57BL/6J mice is
insulin resistance in the mouse. Cell Metab. 2015;21(2):334-347. associated with acceleration of age-dependent renal amyloidosis. Sci
224. Wang S, Yehya N, Schadt EE, Wang H, Drake TA, Lusis AJ. Genetic Rep. 2015;5:16474.
and genomic analysis of a fat mass trait with complex inheritance 244. Bray GA, Lovejoy JC, Smith SR, et al. The influence of different fats
reveals marked sex specificity. PLoS Genet. 2006;2(2):e15. and fatty acids on obesity, insulin resistance and inflammation.
225. Collaboration NCDRF. Trends in adult body-mass index in 200 coun- J Nutr. 2002;132(9):2488-2491.
tries from 1975 to 2014: a pooled analysis of 1698 population- 245. Stanhope KL, Goran MI, Bosy-Westphal A, et al. Pathways and
based measurement studies with 19.2 million participants. Lancet. mechanisms linking dietary components to cardiometabolic disease:
2016;387(10026):1377-1396. thinking beyond calories. Obes Rev. 2018;19(9):1205-1235.
226. Hoeg LD, Sjoberg KA, Jeppesen J, et al. Lipid-induced insulin resis- 246. Alvheim AR, Malde MK, Osei-Hyiaman D, et al. Dietary linoleic acid
tance affects women less than men and is not accompanied by elevates endogenous 2-AG and anandamide and induces obesity.
inflammation or impaired proximal insulin signaling. Diabetes. 2011; Obesity (Silver Spring). 2012;20(10):1984-1994.
60(1):64-73. 247. Ailhaud G, Massiera F, Weill P, Legrand P, Alessandri JM, Guesnet P.
227. Mascarenhas-Melo F, Marado D, Palavra F, et al. Diabetes abrogates Temporal changes in dietary fats: role of n−6 polyunsaturated fatty
sex differences and aggravates cardiometabolic risk in postmeno- acids in excessive adipose tissue development and relationship to
pausal women. Cardiovasc Diabetol. 2013;12:61. obesity. Prog Lipid Res. 2006;45(3):203-236.
PREGUIÇA ET AL. 29

248. Vimaleswaran KS, Berry DJ, Lu C, et al. Causal relationship between 263. Ganeshan K, Chawla A. Warming the mouse to model human dis-
obesity and vitamin D status: bi-directional Mendelian randomiza- eases. Nat Rev Endocrinol. 2017;13(8):458-465.
tion analysis of multiple cohorts. PLoS Med. 2013;10(2):e1001383. 264. Karp CL. Unstressing intemperate models: how cold stress under-
249. Showalter MR, Nonnecke EB, Linderholm AL, et al. Obesogenic diets mines mouse modeling. J Exp Med. 2012;209(6):1069-1074.
alter metabolism in mice. PLoS ONE. 2018;13(1):e0190632. 265. Cannon B, Nedergaard J. Nonshivering thermogenesis and its ade-
250. Yang SC, Lin SH, Chang JS, Chien YW. High fat diet with a high quate measurement in metabolic studies. J Exp Biol. 2011;214(Pt 2):
monounsaturated fatty acid and polyunsaturated/saturated fatty 242-253.
acid ratio suppresses body fat accumulation and weight gain in 266. Tian XY, Ganeshan K, Hong C, et al. Thermoneutral housing acceler-
obese hamsters. Nutrients. 2017;9(10):1148. ates metabolic inflammation to potentiate atherosclerosis but not
251. Takeuchi H, Matsuo T, Tokuyama K, Shimomura Y, Suzuki M. Diet- insulin resistance. Cell Metab. 2016;23(1):165-178.
induced thermogenesis is lower in rats fed a lard diet than in those 267. Giles DA, Moreno-Fernandez ME, Stankiewicz TE, et al. The-
fed a high oleic acid safflower oil diet, a safflower oil diet or a lin- rmoneutral housing exacerbates nonalcoholic fatty liver disease in
seed oil diet. J Nutr. 1995;125(4):920-925. mice and allows for sex-independent disease modeling. Nat Med.
252. Piers LS, Walker KZ, Stoney RM, Soares MJ, O'Dea K. The influence 2017;23(7):829-838.
of the type of dietary fat on postprandial fat oxidation rates: mono- 268. Speakman JR, Keijer J. Not so hot: optimal housing temperatures for
unsaturated (olive oil) vs saturated fat (cream). Int J Obes Relat mice to mimic the thermal environment of humans. Mol Metab.
Metab Disord. 2002;26(6):814-821. 2012;2(1):5-9.
253. Jones PJ, Jew S, AbuMweis S. The effect of dietary oleic, linoleic, 269. Kumar Singh A, Cabral C, Kumar R, et al. Beneficial effects of dietary
and linolenic acids on fat oxidation and energy expenditure in polyphenols on gut microbiota and strategies to improve delivery
healthy men. Metabolism. 2008;57(9):1198-1203. efficiency. Nutrients. 2019;11(9):2216.
254. Marques C, Mega C, Goncalves A, et al. Sitagliptin prevents inflam- 270. Fernandes R, Viana SD, Nunes S, Reis F. Diabetic gut microbiota
mation and apoptotic cell death in the kidney of type 2 diabetic ani- dysbiosis as an inflammaging and immunosenescence condition that
mals. Mediators Inflamm. 2014;2014:538737. fosters progression of retinopathy and nephropathy. Biochim
255. Mega C, de Lemos ET, Vala H, et al. Diabetic nephropathy ameliora- Biophys Acta Mol Basis Dis. 2019;1865(7):1876-1897.
tion by a low-dose sitagliptin in an animal model of type 2 diabetes 271. Turnbaugh PJ, Ridaura VK, Faith JJ, Rey FE, Knight R, Gordon JI.
(Zucker diabetic fatty rat). Exp Diabetes Res. 2011;2011:162092. The effect of diet on the human gut microbiome: a metagenomic
256. Goncalves A, Leal E, Paiva A, et al. Protective effects of the analysis in humanized gnotobiotic mice. Sci Transl Med. 2009;1(6):
dipeptidyl peptidase IV inhibitor sitagliptin in the blood-retinal bar- 6ra14.
rier in a type 2 diabetes animal model. Diabetes Obes Metab. 2012; 272. David LA, Maurice CF, Carmody RN, et al. Diet rapidly and repro-
14(5):454-463. ducibly alters the human gut microbiome. Nature. 2014;505(7484):
257. Warden CH, Fisler JS. Comparisons of diets used in animal models 559-563.
of high-fat feeding. Cell Metab. 2008;7(4):277. 273. Yang Q, Liang Q, Balakrishnan B, Belobrajdic DP, Feng QJ,
258. Benoit B, Plaisancie P, Awada M, et al. High-fat diet action on adi- Zhang W. Role of dietary nutrients in the modulation of gut micro-
posity, inflammation, and insulin sensitivity depends on the control biota: a narrative review. Nutrients. 2020;12(2):381.
low-fat diet. Nutr Res. 2013;33(11):952-960. 274. Wallis KF, Melnyk SB, Miousse IR. Sex-specific effects of dietary
259. Chassaing B, Miles-Brown J, Pellizzon M, et al. Lack of soluble fiber methionine restriction on the intestinal microbiome. Nutrients.
drives diet-induced adiposity in mice. Am J Physiol Gastrointest Liver 2020;12(3):781.
Physiol. 2015;309(7):G528-G541.
260. Pellizzon MA, Ricci MR. The common use of improper control diets
in diet-induced metabolic disease research confounds data interpre-
tation: the fiber factor. Nutr Metab (Lond). 2018;15:3.
261. Dalby MJ, Ross AW, Walker AW, Morgan PJ. Dietary uncoupling of How to cite this article: Preguiça I, Alves A, Nunes S, et al.
gut microbiota and energy harvesting from obesity and glucose tol- Diet-induced rodent models of obesity-related metabolic
erance in mice. Cell Rep. 2017;21(6):1521-1533.
disorders—A guide to a translational perspective. Obesity
262. Ussar S, Griffin NW, Bezy O, et al. Interactions between gut micro-
biota, host genetics and diet modulate the predisposition to obesity Reviews. 2020;1–29. https://doi.org/10.1111/obr.13081
and metabolic syndrome. Cell Metab. 2015;22(3):516-530.

You might also like