Thromb Haemost 2021; 121(06): 703-715
DOI: 10.1055/s-0040-1721388
Review Article

Cardiometabolic Syndrome: An Update on Available Mouse Models

Dimitra Aravani
1   Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
,
Eva Kassi
2   Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
,
Antonios Chatzigeorgiou*
1   Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
3   Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden, Germany
,
Styliani Vakrou*
1   Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
4   Department of Cardiology, “Laiko” General Hospital, Athens, Greece
› Author Affiliations
Funding This work was funded by a grant from the European Foundation for the Study of Diabetes (EFSD) to A.C.
 

Abstract

Cardiometabolic syndrome (CMS), a disease entity characterized by abdominal obesity, insulin resistance (IR), hypertension, and hyperlipidemia, is a global epidemic with approximately 25% prevalence in adults globally. CMS is associated with increased risk for cardiovascular disease (CVD) and development of diabetes. Due to its multifactorial etiology, the development of several animal models to simulate CMS has contributed significantly to the elucidation of the disease pathophysiology and the design of therapies. In this review we aimed to present the most common mouse models used in the research of CMS. We found that CMS can be induced either by genetic manipulation, leading to dyslipidemia, lipodystrophy, obesity and IR, or obesity and hypertension, or by administration of specific diets and drugs. In the last decade, the ob/ob and db/db mice were the most common obesity and IR models, whereas Ldlr−/− and Apoe−/− were widely used to induce hyperlipidemia. These mice have been used either as a single transgenic or combined with a different background with or without diet treatment. High-fat diet with modifications is the preferred protocol, generally leading to increased body weight, hyperlipidemia, and IR. A plethora of genetically engineered mouse models, diets, drugs, or synthetic compounds that are available have advanced the understanding of CMS. However, each researcher should carefully select the most appropriate model and validate its consistency. It is important to consider the differences between strains of the same animal species, different animals, and most importantly differences to human when translating results.


#

Introduction

The cardiometabolic syndrome (CMS) was first described over 90 years ago by Kylin as a combination of hyperglycemia, hypertension, and gout.[1] Later on, Vague pointed out an association between abdominal adiposity and increased risk of type 2 diabetes and CVD.[2] CMS is now defined as a disease entity characterized by the simultaneous occurrence of at least three of the following medical conditions: abdominal obesity, raised plasma glucose, elevated blood pressure, raised triglycerides (TGs) or reduced high-density lipoprotein (HDL) cholesterol.[3] Additional components of CMS which are interconnected through cytokine release from adipose tissue are (1) a proinflammatory state, suggested by elevated C-reactive protein (CRP) and (2) a prothrombotic state, characterized by increased plasma plasminogen activator inhibitor-1 and fibrinogen.[4] Microalbuminuria, increased inflammation/oxidative stress, endothelial dysfunction, nonalcoholic steatohepatitis, abnormalities in the coagulation system, and enhanced cardiovascular tissue renin–angiotensin–aldosterone system (RAAS) contribute to the pathophysiology of CMS.[5] [6] The accumulation of the above metabolic and underlying risk factors promotes the development of atherosclerotic CVD and increases the risk for type 2 diabetes, stroke, and cognitive impairment.[6] [7] [8]

The prevalence of metabolic syndrome in United States has been estimated to be around 35% in both sexes of adults, whereas it increases to around 50% in the age group over 60 years old and particularly in women.[9] [10] [11] Increasingly high prevalence of CMS is seen in populations of different countries, confirming the global burden of the disease.[9] [12] [13] Several prospective cohorts and meta-analysis studies demonstrate that CMS is associated with increased relative risk for cardiovascular outcomes (twofold), diabetes (3.5–5-fold), and all-cause mortality (1.5-fold).[7] [14] In a longitudinal cohort study of 1 million Chinese, the prevalence of cardiometabolic multimorbidity increased from 2.41 to 5.94% in 5 years, indicating the rapid progression of cardiometabolic disease.[15]

Because of the multifactorial nature of this syndrome and the lack of specific treatment,[16] it is essential to elucidate the exact pathophysiologic mechanisms.

Animal models have traditionally been used in research for CVDs in an effort to translate mechanistic and therapeutic findings into human disease.[17] [18] [19] Yet it is critical and rather challenging to choose the appropriate model that would best reflect the human pathophysiology, since animal models present only a fraction of the CMS features at a given time.

Rodent models and particularly mice have been the most popular animal models in the investigation of CMS. This is because of their obvious advantages over other animals, which include easy handling and relatively easy genetic engineering, short gestation and lifespan periods, and most importantly high genetic and physiological similarity to humans.[20] Moreover, they are responsive to experimental diets as they develop central obesity, hyperglycemia, and hypertension, some of the main characteristics of the CMS. The importance and popularity of mouse models are also reflected by the numerous studies that used mice as the reference animal. The database mouse genome informatics (MGI) has catalogued 169 mouse models matching the phenotype “metabolic syndrome,” which are linked to 265 references. Moreover, genetic variation identified in mice has been shown to affect the corresponding human phenotypes.[21]

The aim of this review is to summarize the best characterized mouse models for CMS research and present newer models that have been described during the past 10 years. For this purpose, we conducted a PubMed search using the terms cardiometabolic AND mouse models, with a time filter of 10 years. This search returned 292 results, of which we selected the most appropriate ones using the following criteria: original research articles and those reporting the main aspects of CMS. Similar number (277) was obtained using Scopus as a search engine. To complement our search, we also investigated the MGI database; we found 169 mouse models linked to CMS syndrome and the most relevant models are included in this review.


#

Overview of the Best Characterized Mouse Models

There are three main ways of modeling CMS in mice. These are genetic manipulation, diet/drug-induced protocols, and the combination of any of the above.

Genetic CMS Mouse Models

The strategies of genetic engineering are focused on alteration of lipid metabolism, weight regulation, glucose homeostasis, blood pressure regulation, and the suitable combination of the above phenotypes.

Mouse Models of Dyslipidemia

Low-density lipoprotein (LDL) receptor (Ldlr) and apolipoprotein e (Apoe)-deficient mice are the two best characterized dyslipidemic models. Their hyperlipidemic profile is due to the absence of either Ldlr or Apoe, both of which participate in cholesterol clearance.[22] [23] Ldlr −/− mice develop moderate hypercholesterolemia (total cholesterol ∼250 mg/dL) on a normal diet with lipoprotein profiles similar to those of humans (i.e., elevated LDL) but they are very responsive to atherogenic diet (1.25% cholesterol diet), developing large atherosclerotic lesions and hyperlipidemia. Furthermore, when Ldlr−/− mice are placed on a diet with greater than 20% fat content, they also become obese, display insulin resistance (IR), and impaired glucose tolerance.[24] Apoe−/− mice develop a more severe hyperlipidemia, with an increase in plasma cholesterol levels and TG levels, which leads to spontaneous atherosclerosis on a normal diet.[23] In many cases, Apoe −/− mice do not become obese, nor do they develop IR, even on a high-fat diet (HFD).[25] [26] However, there has been a case where Apoe−/− mice fed HFD (60% fat) for 17 weeks displayed increased body weight, glucose intolerance, and an increase in systemic inflammation, which indicates that modulation of the feeding protocol can have a significant biological effect.[27] In general, as summarized by the Jackson Laboratory and the Mouse Phenome Database which have phenotyped 8-week old male and female Apoe and Ldlr null mice against C57BL/6J mice after 6, 10, and 14 weeks of normal diet (6% fat), these transgenics develop a range of cardiovascular phenotypes. These include elevated plasma cholesterol and TG for both knockouts as well as increased LDL, TG, and ApoB levels in male Ldlr −/− . Caution should be taken however when comparing absolute lipid concentrations between Apoe −/− and Ldlr −/− as various factors could lead to misinterpretations, such as the mechanism of function for each model and differences in the major plasma lipoproteins. Specifically, in Apoe −/− mice the accumulating particles are predominantly apoB48-containing cholesterol ester particles, whereas in Ldlr −/− the major lipoprotein is the ApoB100-containing LDL. Other factors affecting the absolute lipid concentrations include sex, genetic variations, and differences in gut microbiota.[28]


#

Mouse Models of Obesity and IR

Visceral obesity is one of the requirements for CMS and has been positively associated with IR and hyperglycemia.[29] In agreement with human studies, most of the obesity mouse models manifest IR. Leptin deficiency is one of the gold standard models. A spontaneous mutation in leptin gene led to the description of ob/ob mice.[30] Leptin deficiency results in increased food intake, extreme obesity, and reduced energy expenditure. The effect on glucose metabolism is strain-dependent. A C57BL/6J background is characterized by mild hyperglycemia compared with severely diabetic ob/ob mice on the C57BL/KsJ background. Moreover ob/ob mice show increased HDL and reduced LDL, hence they are protected from atherosclerosis and they are also resistant to hypertension.[31] [32] A single autosomal mutation in the leptin receptor[33] causes almost identical phenotypes to leptin deficiency in those mice (db/db) which develop obesity, hyperinsulinemia, hyperglycemia, and increased total cholesterol (predominantly LDL/HDL particles) in the absence of hypertension.[34] An additional model with altered leptin signaling is the POUND mouse generated by Charles River, which lacks all leptin receptor isoforms. When fed Purina diet they became hyperinsulinemic and hyperglycemic, which was accompanied by increased leptin and cholesterol levels.[35] Another model used in CMS research is the agouti yellow obese mouse (Ay/a). In the brain the secreted agouti protein antagonizes the binding of the anorexigenic α-melanocyte-stimulating hormone (α-MSH) to its receptor (melanocortin 4 receptor, MC4-R). α-MSH reduces food intake, increases energy expenditure, and regulates glucose metabolism, thus spontaneous mutations that occur in the agouti gene lead to disruption of these regulatory mechanisms. Agouti mice present adult-onset obesity, hyperinsulinemia, glucose intolerance, hyperglycemia, and hypertension, which establishes it as a useful model of CMS.[36] [37] As mentioned above, hypothalamic MC4-R-mediated signaling plays a role in food intake and energy homeostasis; therefore, targeted disruption of MC4-R was used as an alternative approach. The human relevance of this model is supported by studies which revealed that MC4-R mutations are associated with hereditary obesity.[38] [39] The MC4-R null mouse has a behavioral obesity syndrome characterized by hyperphagia, hyperglycemia, hyperinsulinemia, reduced metabolism, increased lean mass, and altered hepatic lipid metabolism.[40] [41] It has been also shown that MC4-R −/− mice have elevated total plasma cholesterol and TG but tend to be hypotensive rather than hypertensive.[34] [42] Cho et al from Yale University[43] created a polygenic obesity-induced diabetic mouse model (RCS10) by combining different obesity risk-conferring quantitative trait loci (eQTLs) from two different strains; NZO/HlLt and NON/Lt. This is a type 2 diabetes model accompanied by adult-onset obesity, hyperglycemia, IR, and increased hepatic, intramuscular and heart TG at 13 weeks of age; however, data of the effect on blood pressure are lacking.[43]


#

Obesity and Hypertension Models

Adiponectin is another protein that has been associated with obesity and obesity-related diseases in humans.[44] It is secreted by adipocytes and low levels are detected in obesity, which can be reversed during weight loss. Adipo −/− mice fed high fat-high sucrose diet for short periods (2–4 weeks) develop elevated glucose, insulin and free fatty acids, plasma TNF-a, and hypertension.[45] [46] An established model of systemic lupus erythematosus (NZBWF1) was described by Ryan et al in 2006 as potentially important model to study obesity and IR. The NZBWF1 mouse phenotype showed increased weight, plasma insulin, but not glucose levels, increased plasma leptin, renal adipose tissue macrophage infiltration, and hypertension.[47]


#

Nonobese-Lipodystrophic Models

The lipodystrophic A-ZIP F-1 and the aP2-SREBP-1c mice are nonobese mouse models, with restricted adipose capacity concerning mainly the white adipose tissue (WAT) and may be used in the study of nonobese CMS and genetic forms of lipodystrophies.[48] [49] The A-ZIP F-1 mice were generated by enhancing the expression of A-ZIP/F (dominant negative protein of C/EBP transcription factors that regulate growth and differentiation of adipocytes) by using an adipose-specific P2 enhancer promote. This model, which has complete deficiency of WAT, shows that fat ablation can cause liver steatosis and diabetes, with reduced leptin and elevated serum glucose, insulin, free fatty acids and TG, and elevated blood pressure.[48] [50] The transgenic aP2-SREBP-1c mice, which overexpress sterol regulatory element binding proteins (SREBP) in adipose tissue, have decreased WAT but have hypertrophic brown adipose tissue containing cells resembling immature WAT. These mice are characterized by hyperglycemia, hepatic steatosis, and hyperinsulinemia potentiated by secreted frizzled-related protein.[49] [51]

To better recapitulate the various aspects of the metabolic syndrome combinations such as obesity and hyperlipidemia, obesity and hypertension have also been employed and reviewed in detail by Kennedy et al.[34] [Table 1] summarizes the mouse models described above.

Table 1

Genetic CMS mouse models and their metabolic phenotype

Mouse model

Genetic modification

Metabolic changes

Source

Obesity

Hyperglycemia

Insulin resistance

Hypertension

Dyslipidemia

ob/ob

Autosomal recessive mutation in leptin gene

✓ (strain dependent)

↑ Total cholesterol (LDL/HDL1)

[30] [31] [32]

db/db

Autosomal recessive mutation in leptin receptor gene

✓ (age, strain dependent)

↑ Total cholesterol (LDL/HDL1)

[33]

MC4-R−/−

Targeted disruption of MC4-R gene

(exacerbated on HFD)

↓blood pressure

↑ Hepatic TG, ↑ serum FFA

[40] [41]

Ay/a

Spontaneous mutations in agouti gene

(delayed onset)

↓Glucose tolerance (HFD + HS-induced)

(delayed onset)

↑ HDL

[36] [37]

Ldlr−/−

Deletion of Ldlr gene

(HFD-induced)

↓Glucose tolerance (HFD-induced)

(HFD-induced)

↑ LDL

[22] [24]

Apoe−/−

Deletion of Apoe gene

Generally resistant

Generally resistant

↑ Total cholesterol, triglycerides, VLDL, LDL, L

[23]

NZBWF1

Cross between New Zealand black (NZB) mouse with the New Zealand white (NZW) mouse

[47]

Adipo−/−2–4 weeks on diet

Adiponectin knockout

↑ VLDL

[45]

A-ZIP/F-1

Overexpression of A-ZIP/F in adipose tissue

[48] [50]

aP2-SREBP-1c

Overexpression of SREBP-1c in adipose tissue

↑ Hepatic TG

[49]

RCS10

Combination of obesity risk loci on 2 strains; NZO/HlLt and NON/Lt

(adult onset)

↑Hepatic and intramuscular TG

[43]

POUND mouse

Deletion of all leptin isoforms

↑ Total cholesterol

Charles River

Abbreviations: CMS, cardiometabolic syndrome; FFA, free fatty acids; HDL, high-density lipoprotein; LDL, low-density lipoprotein; VLDL, very low-density lipoprotein.


Note: ✓ and ✖ indicate the presence or absence respectively, whereas “–“ indicates no data.



#
#

Diet-Induced CMS Mouse Models

Numerous dietary interventional studies have been conducted to study the pathophysiology of CMS. As with the genetic models these include either a single experimental diet or a combination of diets. The most known approaches are high carbohydrate, high sucrose, high fructose, and HFDs. The mode of action for each diet, with emphasis on the affected biochemical pathways, has been beautifully reviewed by Wong et al[52] and is beyond the scope of this review. Here we will present some key characteristics of the main experimental diets. As far as HFD is concerned, numerous modifications exist, with fat concentrations ranging from 20 to 60%, originating either from plants (e.g., corn, safflower, or olive oil) or animals (e.g., beef, tallow, and lard).[53] The common main outcome of HFD is the increased formation of TG following re-esterification of free fatty acids after lipolysis.[52] Interestingly, HFD-related outcomes are fat-dose-dependent.[54] Fructose is one of the monosaccharides alongside glucose and galactose and it is only an intermediary molecule during glucose metabolism. Chronic fructose consumption is an established way to induce increased energy intake, body weight, adiposity, hypertriglyceridemia, hyperlipidemia, hypertension, glucose intolerance, and decreased insulin sensitivity in laboratory animals and can be more effective than glucose or starch-feeding.[52] [55] According to Wu et al, a low dose of fructose in drinking water (10%) is sufficient to induce CMS in animals.[24] As far as sucrose (table sugar)-enriched diet is concerned, it is widely used for induction of obesity, whole body IR, hyperglycemia, hypertriglyceridemia, and hypercholesterolemia. Amounts of sucrose supplementation range from 20 to 77%.[52] [56] However, sucrose appears to be less effective than the equivalent amount of fructose in inducing CMS as it is a disaccharide consisting of 50% fructose and 50% glucose. Besides, mouse response to daily sucrose consumption is strain-dependent, affected in particular by polymorphisms in the Tas1r3 sweet taste receptor gene.[57]

The effects of each diet or diet combination on mice are summarized in [Table 2].

Table 2

Diet-induced CMS mouse models, the diet treatment, and their metabolic phenotype

Mouse strain

Diet treatment

Metabolic changes

Obesity

Hyperglycemia

Hypertension

Dyslipidemia

Male C57BL/6 J mice

High-fat diet, 8 wk

Male AKR/J (AKR) mice

High-fat diet, 8 wk

Male and female C57Bl/6J mice

High-fat diet (60%), 20 wk

Male and female A/J mice

High-fat diet (60%), 20 wk

Male C57BL/6 mice

High-fat diet, 16 wk

Male C57BL/6 mice

High-fat diet, 40 wk

Female C57Bl/6 J mice

High-fat diet, 24 wk

C57BL/6 mice (wild type)

High-fat diet, 13 wk

Ldlr−/− mice

High-fat diet, 13 wk

Female C57BL/6NTac mice

High-fat diet, 12 wk

Male C57BL/6 mice

High-fat diet, 12 wk

Male C57BL/6 J mice

High-fat diet, 4 wk

Male C57BL/6 mice

High-fat diet, 16 wk

Male NMRI mice

Fructose drinking water (15%), 10 wk

Sucrose soft drink

(10%), 10 wk

Noncaloric soft drink, 10 wk

Abbreviation: CMS, cardiometabolic syndrome.


Note: ✓ and ✖ indicate the presence or absence respectively, whereas “–“ indicates no data. Source: Adapted and reprinted with permission from Wong SK, Chin K-Y, Suhaimi FH, Fairus A, Ima-Nirwana S. Animal models of metabolic syndrome: a review. Nutr Metab. (Lond) 2016;13:65.



#

Drug/Chemically Induced CMS Mouse Models

Streptozotocin administration is widely used to induce diabetes in mice. The mode of action for streptozotocin is via destruction of the β pancreatic cells. Streptozotocin is a glucose analogue that accumulates in β pancreatic cells via the GLUT2 glucose transporter. Type 1 diabetes manifestation occurs in mice treated with Streptozotocin.[58] Glucocorticoid-induced metabolic syndrome has also been reported. Glucocorticoids are hormones that regulate glucose homeostasis in various ways such as gluconeogenesis in liver, whereas in skeletal muscle and WAT they decrease glucose uptake and utilization by antagonizing insulin response. Therefore, mice treated with glucocorticoids display glucose intolerance and reduced insulin sensitivity. Apart from body weight gain, they present dyslipidemia, central, liver and skeletal muscle fat accumulation, and hypertension.[59]


#
#

Newer CMS Mouse Models Used in Cardiometabolic Research

Dietary Interventions

Diet-induced obesity models are repeatedly used to assess the effect of various factors on cardiometabolic phenotype. It seems that HFD administered for various intervals is one of the most preferable interventions used to induce CMS. This approach most frequently leads to weight gain, dyslipidemia, and IR. C57/BL/6J mice fed with 60% HFD for 11 weeks were used as a model of obesity to study the effects of angiotensin (1–7). These obese mice exhibited high blood pressure and reduced insulin sensitivity.[60] Similarly, Miranda et al utilized diet-induced obesity to investigate the effect of cannabinoid receptor 1 antagonists. In this study, mice were fed with 60% HFD for a longer period (16 weeks). HFD increased body weight, fasting glucose, and IR, but also promoted adipose tissue inflammation characterized by inflammatory M1 macrophage infiltration.[61] Another study confirmed the HFD-induced M1–M2 imbalance in WAT and the abnormalities in glycemic indexes and lipid profiles.[62]

Guo et al[63] fed 2-month-old male C57BL/6 mice with either HFD (45% energy from fat) including 7% sucrose or low-fat diet (LFD; 10% energy from fat). Control mice fed LFD were compared with various HFD groups which were supplemented with one of three fatty acids (EPA, DPA, and DHA) or none, after 6 weeks of intervention. The HFD group with no addition of fatty acids showed gathered characteristics of the CMS, in that mice were significantly obese with increased liver weight and epididymal adiposity, fatty liver, increased hepatic total cholesterol, high serum glucose, alanine transaminase, reduced adiponectin, and IR. Additionally, upregulation of inflammatory genes in the TLR-4/NF-κB pathway was observed. Supplementation of long-chain omega-3 polyunsaturated fatty acids could reverse the above effects. Blood pressure was not assessed in this model.[63] Leptin levels were also elevated after 8 weeks of HFD in a different study.[64] Similar phenotypes were reported by several studies using HFD as a mean of mimicking the CMS.[65] [66] [67] [68] [69] CMS-related serum biomarkers were investigated in HFD (60%) fed mice for an extended period (36 weeks). Besides increased body weight, dysregulated lipid metabolism (high total cholesterol and TG), raised blood glucose, insulin, glycated hemoglobin (HbA1c), and brain natriuretic peptide, there were also abnormalities in renin–angiotensin system. This was suggested by high serum angiotensin-converting enzyme, angiotensin II (Ang-II), Ang-II type 1 receptor, and aldosterone. Cardiac remodeling was also observed in the HFD group compared with normal diet (10% fat).[70] A combination of diet and drug-induced CMS has also been tested. Here mice were maintained on 60% HFD for 5 to 15 weeks alongside nitric oxide synthase inhibitor L-NAME. This double hit model[71] [72] developed several aspects of CMS, as evident by obesity, hyperglycemia, reduced insulin sensitivity, impaired glucose clearance (heightened fasting glucose, HbA1c), hypertension, and endothelial dysfunction. Various diet modifications have been also described, such as high fat (60%) diet with salt addition (7.25% NaCl), which was sufficient to cause most of the aspects of CMS including diabetes, hypertension, and secondary features such as cardiac dysfunction and vascular remodeling.[73] A combination of high fat (35%)–high sucrose (34%) long-term (4 months) feeding protocol caused body weight gain, impaired glucose homeostasis, increased hepatic and myocardial TG content, as well as cardiac dysfunction in male C57BL6R mice.[74] All the above studies indicate that HFD is a reliable and simple protocol that induces reproducible phenotypes. However, something that should be taken into consideration when it comes to HFD protocols is that mice may respond differently according to their genetic background[75] [76] or their gut microbiota adaptation to HFD. This adaptation involves transcriptional changes in hepatic miRNAs leading to a divergent metabolic phenotype. Subsequently, one can claim that hepatic lipid metabolism is modulated according to the response to HFD.[77] [78] Other limitations regarding HFD include disparities in phenotypes sometimes attributed to the modulation of the HFD protocol (i.e., duration of intervention, age of mice)[79] and the diet compositions particularly the type of fat and amount of carbohydrates, which can sway the balance between protection and lipotoxicity,[80] [81] sex-specific responsiveness to HFD[82] and strain specificity to sugar consumption,[57] behavioral effects[83] and intramyocellular lipid accumulation causing muscle defects.[84]

Although obesity-related complications such as IR, nonalcoholic fatty liver disease, and CVD share common pathophysiology mechanisms such as increased tissular lipid deposition, hypoxia, and low-grade inflammation, different lipid species and groups are implicated in the aforementioned processes.[85] For instance, in the case of atherosclerosis and CVD, it is widely accepted that the presence of ApoB-containing lipoproteins such as very low-density lipoprotein (VLDL), LDL, and lipoprotein-a is of determinant importance for the pathogenesis of such complications.[86] The adipose tissue, on the other hand, is predominantly characterized by the presence of triacylglycerols rather than cholesterol or phospholipids and, during obesity, specific triacylglycerol signatures, defined by different fatty acid compositions, are associated to the development of IR.[87] Cholesterol and especially cholesterol esters are considered important inducers of nonalcoholic fatty liver disease, independently of obesity and metabolic dysregulation.[88] Nevertheless, the progression of the disease into nonalcoholic steatohepatitis in both humans and mice depends on the existence of specific long-chain fatty acids that are toxic to the hepatocytes.[89] Considering that the different diets that are used in animal models to provoke CMS trigger the emergence of different metabolic signatures in vivo and finally simulate different CMS-related complications, and not all of them together, exceptional caution should be taken when animal models of CMS are used for the study of CVD.


#

Genetic Manipulations

In addition to diet-induced strategies, genetic mouse models have been extensively used as illustrated in the first part of this review. Genetic manipulations include either manipulation of known metabolic pathways or exploration of new genes that have been shown to be associated with human CMS. This approach allows researchers to dissect the genetic component of CMS, induce more severe phenotypes, and in some occasions shorten the study duration compared with long-term feeding protocols. Such an example is the human cholesteryl ester transfer protein (CETP), which has no homologous gene in mouse and has been found to be associated with human CMS.[90] CEPT is responsible for the exchange of TG and cholesterol esters between LDL and VLDL particles and HDL. CETP activity is associated with reduced HDL and increased risk of atherosclerosis. There is no mouse homologue of this gene, therefore overexpression of the human CETP has been applied to mimic the human conditions. In several studies, CETP overexpression is combined with Apoe deficiency conferring reduced HDL and reduced clearance of TG-rich lipoproteins. APOE*3-Leiden.CETP mouse is a model combining obesity, IR, and hyperlipidemia when treated with HFD and fructose. It is responsive to antidiabetic and lipid-lowering drugs evidenced by improved glucose regulation, IR, and dyslipidemia.[91] Moreover, transcriptome analysis revealed that these mice when fed HFD comprise distinct hepatic gene signatures and serum miRNAs.[92] Hemizygous CETP expression combined with Ldlr haploinsufficiency was also used as a model that reflects the lipoprotein metabolism and the risk of cardiometabolic disease in humans. These transgenic mice have perturbed lipid profiles showing increased LDL and decreased HDL. Glucose was not altered as a function of CETP expression and blood pressure and body weight data were not available. Thus this model lacks some of the main components of CMS.[93]

The Ldlr-deficient mouse treated with HFD is still commonly used and seems to be quite consistent as far as the lipid phenotype, hepatic inflammation, and atherosclerosis development are concerned.[94] [95] [96] [97] Ldlr −/− mice fed high fat sucrose enriched diet with 0.15% cholesterol for 12 weeks developed diet-induced obesity, increased fasting insulin and glucose levels, and dyslipidemia characterized by increased TG and total cholesterol. Accelerated liver inflammation was also detected by elevated expression of inflammatory genes such as Emr1, Tnfa, ll-6, Ccl2 as well as susceptibility to atherosclerosis. However, blood pressure was not evaluated in this mouse model.[94] Apoe-deficient mice appeared less frequently in the literature search we conducted. Crossing of heterozygous aromatase-deficient mice (Ar +/−) on the Apoe −/− background was used to generate metabolically altered mice (MetS-Tg). The double knockout animals displayed central obesity, increased body weight, elevated serum, and increased blood pressure. Glucose and insulin tolerance tests revealed glucose intolerance and IR. Detection of cytokines such as TNF-a, IL-6, and CRP indicates an inflammation state.[98] The same group reported almost identical results for MetS-Tg mice in a different study, which were characterized by increased body weight compared with wild types, no differences in plasma glucose but in glucose area under the curve, and higher mean arterial blood pressure.[99] The induction of hyperlipidemia has been traditionally achieved by time-consuming generation of knockout transgenics of Ldlr and Apoe. Recently, hyperlipidemia has been attempted via adeno-associated-virus-8 (AAV8)-mediated overexpression of PCSK9.[100] A single intravenous femoral injection of viral particles containing the human PCSK9(DY) gene, which is a gain-of-function mutation of PCSK9 under the liver-specific promoter HCR-hAAH, led to stable hepatic expression of this mutant protein. Mice expressing the mutant PCSK9 had increased serum cholesterol and particularly LDL from day 30 up to 1-year postinjection, under normal feeding conditions[100] and much earlier (day 7) when treated with HFD as reported by others.[101] Consistent with the elevated fraction of LDL, the hepatic expression of Ldlr was significantly reduced. HFD exacerbated the observed hyperlipidemia and atherogenicity. The potency of the adenovirus-mediated PCSK9 transexpression was also confirmed in three different genetic mouse strains. This fast and efficient way of introducing hypercholesterolemia in mice without the need of genetic manipulation has since been employed by several groups.[102] [103] [104]

Genetic ablation of GC-A guanylyl cyclase-A (GC-A) receptor in β pancreatic cells is a novel genetic mouse model used for the investigation of the role of atrial and brain-type natriuretic peptides and its GC-A receptor in glucose homeostasis. The disruption of GC-A receptor in β-cells combined with diet-induced obesity (60% HFD) was sufficient to cause arterial hypertension, but not obesity (compared with wild types on HFD), increased area under the curve during glucose tolerance test which was maintained from 8 to 18 weeks of feeding, and raised blood insulin.[105]

Another genetic model utilizing the neuropeptide Y (NPY) was first described in 2008.[106] NPY is a co-transmitter expressed in the brain and released alongside noradrenaline in response to sympathetic stimulation. It is also expressed in neurons innervating the vasculature cardiomyocytes and endocardium, and is involved in physiological processes including vasoconstriction, cardiac remodeling, and angiogenesis. Overexpression of NPY in noradrenergic neurons of the central and peripheral nervous system leads to increased WAT weight (subcutaneous, epididymal, and retroperitoneal fat pads) but not overall body weight, increased liver TG, and fasting glucose. These changes occurred in the absence of modified diet. The relevance of NPY in cardiometabolic animal modeling is also supported by human studies, where genetic polymorphisms found in NPY receptors (Y1, Y2, Y5) are associated with cardiometabolic risk factors and early-onset CVD risk.[107] [108] Deletion of protein kinase D2 (PRKD2) triggers hyperinsulinemia which precedes IR and precipitates metabolic changes. Increased β-cell insulin secretion was mediated by increased expression and activation of L-Ca2+ channels and subsequent high glucose and membrane depolarization. The authors have focused on insulin secretion regulation rather than lipid metabolism and blood pressure data which are not reported.[109]

Besides the known obesity-induced diabetes models, ob/ob and db/db, which are widely used with or without a diet combination,[110] [111] [112] [113] a very recent study has designed a new mouse model, which recapitulates maturity-onset diabetes of the young (MODY).[114] MODY4 mouse is essentially a pancreatic and duodenal homeobox 1 (Pdx1) haploinsufficient animal. Pdx1 is a transcription factor that regulates β-pancreatic cell maturation. Pdx1+/− mice were further challenged by genetic inhibition of the stress-responsive IKK/NF-κB signaling pathway resulting in a double transgenic IKK2-DNPdx1 animal. Transgenics presented rapid onset of diabetes characterized by hyperglycemia, hyperinsulinemia, and loss of β-cell mass.[114] Although this is a model of diabetes, it could be potentially combined with high fat or other dietary intervention to introduce hyperlipidemia and/or obesity.


#

Other Models for CMS Simulation

The model of maternal separation and early weaning (MSEW) together with high fat feeding predispose female offspring to an early onset of cardiometabolic risk factors, including hyperinsulinemia, glucose intolerance, and hypercholesterolemia as well as increased weight and fat mass compared with MSEW and non-MSEW male counterparts. Both male and female MSEW mice fed HFD also exhibited an increase in blood pressure compared with controls, non-MSEW.[115] Other studies that link maternal feeding status and the cardiometabolic profile of offspring have been reported. Saad et al allocated pregnant C57BL/6J mice to fructose solution or water from day 1 of pregnancy. The fructose protocol promoted hypoglycemia as indicated by intraperitoneal glucose testing in both sexes and higher mean arterial pressure compared with controls but had no effect on lipid levels. Moreover, there was a female-specific effect on various other parameters following fructose intake compared with the control group. Female mice had higher weights with visceral adiposity, hepatic fat accumulation, IR, and high serum leptin.[116] Similarly, another group concluded that maternal diet-induced obesity coupled with postweaning obesogenic diet worsened offspring hyperinsulinemia, hyperleptinemia, fat deposition, hypertension risk, and cardiac fibrosis.[117] A recent study has implicated maternal diet in offspring malfunctioning metabolism. Maternal high-fructose feeding caused multigenerational hypertension, which was characterized by increased mRNA expression of the RAAS genes, as well as the expression of renin in the kidneys. The authors conclude that high fructose intake during pregnancy activated the RAAS and epigenetically induced changes in the cardiometabolic system of the offspring.[118] While maternal nutrition status can affect the progeny, a different study reported that paternal cholestasis (10 weeks of 0.5% cholic acid-supplemented diet prior to mating) resulted in metabolic defects in male offspring. Male pups of cholestatic fathers further treated with western diet for 18 weeks had raised fasting insulin, hepatic TG content, and serum cholesterol levels compared with diet-matched controls. At 25 weeks of age, male offspring showed hypertension as well.[119]

The above studies suggest that the prenatal and perinatal periods are as important as the postnatal lifestyle for the onset of CMS and—as highlighted by others—it is important to understand the long-term effects of treatment during pregnancy as it can have significant effects on offspring.[120]

It is also worth mentioning that wild-derived inbred strains have been suggested to be informative about our knowledge of CMS. These inbred strains arose from inbreeding of wild mice caught around the world. Karunakaran and Clee have extensively reviewed the characteristic of these wild-derived species (Cast, Molf, PWD, PWK, Spret, WSB), which are generally obesity-resistant but they show a spectrum of abnormalities in lipid and glucose metabolism.[21]

Finally, in [Table 3] we present additional mouse models that carry the main CMS components after searching the MGI database for mouse models under the phenotype metabolic syndrome.[121] [122] [123] [124] [125] [126] [127] [128] [129] [130] [131] [132] [133] [134] Moreover, the MGI search confirmed already mentioned models such as HFD-induced, ob/ob, KKAy/a, streptozotocin-induced, genetic strains (PWD, PCD1), NPY, and triple knockouts (ob/ob; Apoe −/− on ApoB100 background).

Table 3

Additional genetic CMS mouse models as reported in MGI database and their metabolic phenotype

Mouse model

Genetic modification

Metabolic changes

Source

GNB3/+

BAC transgenic mice that carry an extra copy of human GNB3

↑ Fasting plasma glucose, insulin, and C-peptide, triglycerides, cholesterol, phospholipids, and glucose intolerance

[121]

SNAP-25b

Targeted deletion of SNAP-25b

Hyperglycemia, liver steatosis, and adipocyte hypertrophy (HFD exacerbated)

[122]

Het-MλKO

Heterozygous muscle-specific PKC-λ knockout

Hyperglycemia, hyperinsulinemia, abdominal obesity, hepatosteatosis, hypertriglyceridemia, and hypercholesterolemia

[123] [124]

Tyk2−/−

Tyrosine kinase 2 knockout

Obesity, ↑ plasma insulin, cholesterol, and FFA

[125]

AdiposeENPP1-TG

Targeted overexpression of human ENPP1 in adipocytes

↑ Plasma FFA, triglycerides, glucose, insulin, fatty liver (HFD-induced)

[133]

Ankrd26

Partial inactivation of the Ankrd26 gene

Obesity, ↑ plasma insulin

[126]

PrRP-deficient mice

Targeted disruption of PrRP gene

Late-onset obesity and adiposity, ↑ blood glucose

[127]

Timo/Timo

Targeted disruption of Bdnf gene

Obesity, hepatic steatosis, ↑ leptin, insulin, glucose cholesterol, LDL cholesterol

[128]

Neil1−/−

Targeted deletion of Neil1

Severe obesity, dyslipidemia, fatty liver disease, and tendency to hyperinsulinemia

[129]

GPCR12 KO

Deletion of GPCR12 gene

↑ Body weight, body fat mass, hepatic steatosis, dyslipidemia

[130]

RS1−/−

Targeted deletion of RS1 gene

Obesity, ↑ total fat, serum cholesterol, and leptin

[131]

PLSCR3−/−

Targeted deletion of PLSCR3 gene

↑ Abdominal fat, FFA, triglycerides, and leptin, ↓ leptin, insulin resistance, glucose intolerance

[132]

11β HSD-1 TG

Selective overexpression of 11β HSD-1 in adipose

Visceral obesity (HFD exacerbated) insulin resistance, hyperlipidemia, hyperphagia despite hyperleptinemia

[134]

Abbreviations: CMS, cardiometabolic syndrome; FFA, free fatty acids; HFD, high-fat diet; LDL, low-density lipoprotein; MGI, mouse genome informatics.



#
#

Conclusion

CMS is a global epidemic with increasing prevalence. Due to its complex etiology—a combination of genetic and environmental factors—and pathophysiology, it is challenging to address effective therapies. Animal models have traditionally guided research and shed light on the involved mechanisms and candidate therapeutics.

In the first part of the review, we summarized the best characterized mouse models for CMS; the last comprehensive reviews focusing on CMS mouse models date back in 2010.[34] [135] In the second part we reviewed the PubMed and MGI database and described mouse models that have been developed in the last 10 years. Also, we identified the models that are repeatedly used, confirming their utility and contribution to the field. We conclude that ob/ob and db/db mice are the most common obesity and IR models, whereas Ldlr −/− and Apoe −/− are widely used to induce hyperlipidemia. These mice have been extensively used either as a single transgenic or combined with a different background with or without diet treatment. As far as diet-induced models are concerned, we found that HFD (40–60%) with modifications (sucrose, salt addition) is the preferred protocol, which generally leads to increased body weight, hyperlipidemia, and IR.

The significance of mouse as a proxy organism to study the human CMS has been extensively documented through numerous studies. The generation of genetically engineered mouse models and the idea of altered diet consumption date back to early 1990s. An important advantage is the availability of inbred strains, which provide a constant genetic background (within strains) that allows us to study the effects of various manipulations (gene–gene, gene–environmental) on CMS development. However, there are defined genetic differences among different inbred strains, which can alter their responsiveness threshold. In the context of CMS, it is proven that susceptibility to atherosclerosis, hyperlipidemia, hypertension, and diet adaptation varies among different mouse strains.[31] [136] [137] [138] Thus, this should be taken into account when interpreting or comparing inter-study results. Furthermore, for diet-induced CMS the selection of proper control diet is crucial as cardiometabolic alterations were found to be underestimated in low fat diet-treated controls.[139] Another emerging issue that needs to be addressed is the impact of sex in the study of CMS. Traditionally, male mice are preferred over females because of the known development of a more severe disease phenotype and because of the concern that the estrous cycle induces variability in traits that complicate experimental designs; however, the latter has been debated.[140] The current scientific view encourages the study of both sexes since it can differentially affect metabolic phenotype and give ground to sex-based treatments of metabolic diseases.[141]

In conclusion, the plethora of genetically engineered mouse models, diets, drugs, or synthetic compounds that are available nowadays facilitates the research of CMS. However, each researcher should carefully select the most appropriate model for his/her research and validate the consistency of the chosen model. Overall, when translating discoveries across strains and species it is important to consider the differences between strains of the same animal species (e.g., not all mice are equivalent to C57/BL mice) and, most importantly, the differences between human and animal models.[142]


#
#

Conflict of Interest

None declared.

* These authors contributed equally to this work as senior authors.


  • References

  • 1 Kylin E. Studien ueber das hypertonie-hyperglyka. Zentralblatt Fuer Inn Med 1923; 44: 105-127
  • 2 Vague J. The degree of masculine differentiation of obesities: a factor determining predisposition to diabetes, atherosclerosis, gout, and uric calculous disease. Am J Clin Nutr 1956; 4 (01) 20-34
  • 3 Alberti KGMM, Eckel RH, Grundy SM. et al; International Diabetes Federation Task Force on Epidemiology and Prevention, Hational Heart, Lung, and Blood Institute, American Heart Association; World Heart Federation, International Atherosclerosis Society, International Association for the Study of Obesity. Harmonizing the metabolic syndrome: a joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; and International Association for the Study of Obesity. Circulation 2009; 120 (16) 1640-1645
  • 4 Grundy SM, Brewer Jr HB, Cleeman JI, Smith Jr SC, Lenfant C. American Heart Association, National Heart, Lung, and Blood Institute. Definition of metabolic syndrome: report of the National Heart, Lung, and Blood Institute/American Heart Association conference on scientific issues related to definition. Circulation 2004; 109 (03) 433-438
  • 5 Castro JP, El-Atat FA, McFarlane SI, Aneja A, Sowers JR. Cardiometabolic syndrome: pathophysiology and treatment. Curr Hypertens Rep 2003; 5 (05) 393-401
  • 6 Grundy SM, Cleeman JI, Daniels SR. et al; American Heart Association, National Heart, Lung, and Blood Institute. Diagnosis and management of the metabolic syndrome: an American Heart Association/National Heart, Lung, and Blood Institute Scientific statement. Circulation 2005; 112 (17) 2735-2752
  • 7 Mottillo S, Filion KB, Genest J. et al. The metabolic syndrome and cardiovascular risk a systematic review and meta-analysis. J Am Coll Cardiol 2010; 56 (14) 1113-1132
  • 8 Yates KF, Sweat V, Yau PL, Turchiano MM, Convit A. Impact of metabolic syndrome on cognition and brain: a selected review of the literature. Arterioscler Thromb Vasc Biol 2012; 32 (09) 2060-2067
  • 9 Aguilar M, Bhuket T, Torres S, Liu B, Wong RJ. Prevalence of the metabolic syndrome in the United States, 2003-2012. JAMA 2015; 313 (19) 1973-1974
  • 10 Sherling DH, Perumareddi P, Hennekens CH. Metabolic syndrome. J Cardiovasc Pharmacol Ther 2017; 22 (04) 365-367
  • 11 Pradhan AD. Sex differences in the metabolic syndrome: implications for cardiovascular health in women. Clin Chem 2014; 60 (01) 44-52
  • 12 Sigit FS, Tahapary DL, Trompet S. et al. The prevalence of metabolic syndrome and its association with body fat distribution in middle-aged individuals from Indonesia and the Netherlands: a cross-sectional analysis of two population-based studies. Diabetol Metab Syndr 2020; 12: 2
  • 13 Saklayen MG. The global epidemic of the metabolic syndrome. Curr Hypertens Rep 2018; 20 (02) 12
  • 14 Ford ES, Li C, Sattar N. Metabolic syndrome and incident diabetes: current state of the evidence. Diabetes Care 2008; 31 (09) 1898-1904
  • 15 Zhang D, Tang X, Shen P. et al. Multimorbidity of cardiometabolic diseases: prevalence and risk for mortality from one million Chinese adults in a longitudinal cohort study. BMJ Open 2019; 9 (03) e024476
  • 16 Chatzigeorgiou A, Kandaraki E, Papavassiliou AG, Koutsilieris M. Peripheral targets in obesity treatment: a comprehensive update. Obes Rev 2014; 15 (06) 487-503
  • 17 Vakrou S, Abraham MR. Hypertrophic cardiomyopathy: a heart in need of an energy bar?. Front Physiol 2014; 5: 309
  • 18 Halapas A, Papalois A, Stauropoulou A. et al. In vivo models for heart failure research. In Vivo 2008; 22 (06) 767-780
  • 19 Santhekadur PK, Kumar DP, Sanyal AJ. Preclinical models of non-alcoholic fatty liver disease. J Hepatol 2018; 68 (02) 230-237
  • 20 Perlman RL. Mouse models of human disease: An evolutionary perspective. Evol Med Public Health 2016; 2016 (01) 170-176
  • 21 Karunakaran S, Clee SM. Genetics of metabolic syndrome: potential clues from wild-derived inbred mouse strains. Physiol Genomics 2018; 50 (01) 35-51
  • 22 Ishibashi S, Brown MS, Goldstein JL, Gerard RD, Hammer RE, Herz J. Hypercholesterolemia in low density lipoprotein receptor knockout mice and its reversal by adenovirus-mediated gene delivery. J Clin Invest 1993; 92 (02) 883-893
  • 23 Plump AS, Smith JD, Hayek T. et al. Severe hypercholesterolemia and atherosclerosis in apolipoprotein E-deficient mice created by homologous recombination in ES cells. Cell 1992; 71 (02) 343-353
  • 24 Wu L, Vikramadithyan R, Yu S. et al. Addition of dietary fat to cholesterol in the diets of LDL receptor knockout mice: effects on plasma insulin, lipoproteins, and atherosclerosis. J Lipid Res 2006; 47 (10) 2215-2222
  • 25 Gao J, Katagiri H, Ishigaki Y. et al. Involvement of apolipoprotein E in excess fat accumulation and insulin resistance. Diabetes 2007; 56 (01) 24-33
  • 26 Hofmann SM, Perez-Tilve D, Greer TM. et al. Defective lipid delivery modulates glucose tolerance and metabolic response to diet in apolipoprotein E-deficient mice. Diabetes 2008; 57 (01) 5-12
  • 27 King VL, Hatch NW, Chan H-W, de Beer MC, de Beer FC, Tannock LR. A murine model of obesity with accelerated atherosclerosis. Obesity (Silver Spring) 2010; 18 (01) 35-41
  • 28 Getz GS, Reardon CA. Do the Apoe−/− and Ldlr−/−mice yield the same insight on atherogenesis?. Arterioscler Thromb Vasc Biol 2016; 36 (09) 1734-1741
  • 29 Papaetis GS, Papakyriakou P, Panagiotou TN. Central obesity, type 2 diabetes and insulin: exploring a pathway full of thorns. Arch Med Sci 2015; 11 (03) 463-482
  • 30 Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature 1994; 372 (6505): 425-432
  • 31 Haluzik M, Colombo C, Gavrilova O. et al. Genetic background (C57BL/6J versus FVB/N) strongly influences the severity of diabetes and insulin resistance in ob/ob mice. Endocrinology 2004; 145 (07) 3258-3264
  • 32 Qiu J, Ogus S, Mounzih K, Ewart-Toland A, Chehab FF. Leptin-deficient mice backcrossed to the BALB/cJ genetic background have reduced adiposity, enhanced fertility, normal body temperature, and severe diabetes. Endocrinology 2001; 142 (08) 3421-3425
  • 33 Hummel KP, Dickie MM, Coleman DL. Diabetes, a new mutation in the mouse. Science 1966; 153 (3740): 1127-1128
  • 34 Kennedy AJ, Ellacott KLJ, King VL, Hasty AH. Mouse models of the metabolic syndrome. Dis Model Mech 2010; 3 (3–4): 156-166
  • 35 Charles River. The C57BL/6NCrl-Leprdb-lb/Crl mouse: a model for metabolic syndrome/pre-diabetes. Accessed April 21, 2020 at: https://www.criver.com/sites/default/files/resources/rm_rm_r_POUND_MOUSE_fact_sheet.pdf
  • 36 Frigeri LG, Wolff GL, Teguh C. Differential responses of yellow Avy/A and agouti A/a (BALB/c X VY) F1 hybrid mice to the same diets: glucose tolerance, weight gain, and adipocyte cellularity. Int J Obes 1988; 12 (04) 305-320
  • 37 Mark AL, Shaffer RA, Correia ML, Morgan DA, Sigmund CD, Haynes WG. Contrasting blood pressure effects of obesity in leptin-deficient ob/ob mice and agouti yellow obese mice. J Hypertens 1999; 17 (12, Pt 2): 1949-1953
  • 38 Yeo GS, Farooqi IS, Aminian S, Halsall DJ, Stanhope RG, O'Rahilly S. A frameshift mutation in MC4R associated with dominantly inherited human obesity. Nat Genet 1998; 20 (02) 111-112
  • 39 Vaisse C, Clement K, Durand E, Hercberg S, Guy-Grand B, Froguel P. Melanocortin-4 receptor mutations are a frequent and heterogeneous cause of morbid obesity. J Clin Invest 2000; 106 (02) 253-262
  • 40 Huszar D, Lynch CA, Fairchild-Huntress V. et al. Targeted disruption of the melanocortin-4 receptor results in obesity in mice. Cell 1997; 88 (01) 131-141
  • 41 Lede V, Meusel A, Garten A. et al. Altered hepatic lipid metabolism in mice lacking both the melanocortin type 4 receptor and low density lipoprotein receptor. PLoS One 2017; 12 (02) e0172000
  • 42 Iqbal J, Li X, Chang BH-J. et al. An intrinsic gut leptin-melanocortin pathway modulates intestinal microsomal triglyceride transfer protein and lipid absorption. J Lipid Res 2010; 51 (07) 1929-1942
  • 43 Cho Y-R, Kim H-J, Park S-Y. et al. Hyperglycemia, maturity-onset obesity, and insulin resistance in NONcNZO10/LtJ males, a new mouse model of type 2 diabetes. Am J Physiol Endocrinol Metab 2007; 293 (01) E327-E336
  • 44 Nigro E, Scudiero O, Monaco ML. et al. New insight into adiponectin role in obesity and obesity-related diseases. BioMed Res Int 2014; 2014: 658913
  • 45 Ouchi N, Ohishi M, Kihara S. et al. Association of hypoadiponectinemia with impaired vasoreactivity. Hypertension 2003; 42 (03) 231-234
  • 46 Ohashi K, Kihara S, Ouchi N. et al. Adiponectin replenishment ameliorates obesity-related hypertension. Hypertension 2006; 47 (06) 1108-1116
  • 47 Ryan MJ, McLemore Jr GR, Hendrix ST. Insulin resistance and obesity in a mouse model of systemic lupus erythematosus. Hypertension 2006; 48 (05) 988-993
  • 48 Moitra J, Mason MM, Olive M. et al. Life without white fat: a transgenic mouse. Genes Dev 1998; 12 (20) 3168-3181
  • 49 Shimomura I, Hammer RE, Richardson JA. et al. Insulin resistance and diabetes mellitus in transgenic mice expressing nuclear SREBP-1c in adipose tissue: model for congenital generalized lipodystrophy. Genes Dev 1998; 12 (20) 3182-3194
  • 50 Takemori K, Gao Y-J, Ding L. et al. Elevated blood pressure in transgenic lipoatrophic mice and altered vascular function. Hypertension 2007; 49 (02) 365-372
  • 51 Hörbelt T, Knebel B, Fahlbusch P. et al. The adipokine sFRP4 induces insulin resistance and lipogenesis in the liver. Biochim Biophys Acta Mol Basis Dis 2019; 1865 (10) 2671-2684
  • 52 Wong SK, Chin K-Y, Suhaimi FH, Fairus A, Ima-Nirwana S. Animal models of metabolic syndrome: a review. Nutr Metab (Lond) 2016; 13: 65
  • 53 Buettner R, Schölmerich J, Bollheimer LC. High-fat diets: modeling the metabolic disorders of human obesity in rodents. Obesity (Silver Spring) 2007; 15 (04) 798-808
  • 54 Ghibaudi L, Cook J, Farley C, van Heek M, Hwa JJ. Fat intake affects adiposity, comorbidity factors, and energy metabolism of sprague-dawley rats. Obes Res 2002; 10 (09) 956-963
  • 55 Johnson RJ, Segal MS, Sautin Y. et al. Potential role of sugar (fructose) in the epidemic of hypertension, obesity and the metabolic syndrome, diabetes, kidney disease, and cardiovascular disease. Am J Clin Nutr 2007; 86 (04) 899-906
  • 56 Rasool S, Geetha T, Broderick TL, Babu JR. High fat with high sucrose diet leads to obesity and induces myodegeneration. Front Physiol 2018; 9: 1054
  • 57 Glendinning JI, Breinager L, Kyrillou E, Lacuna K, Rocha R, Sclafani A. Differential effects of sucrose and fructose on dietary obesity in four mouse strains. Physiol Behav 2010; 101 (03) 331-343
  • 58 Furman BL. Streptozotocin-induced diabetic models in mice and rats. Curr Protoc Pharmacol 2015; 70: 1-20
  • 59 Fransson L, Franzén S, Rosengren V, Wolbert P, Sjöholm Å, Ortsäter H. β-Cell adaptation in a mouse model of glucocorticoid-induced metabolic syndrome. J Endocrinol 2013; 219 (03) 231-241
  • 60 Loloi J, Miller AJ, Bingaman SS, Silberman Y, Arnold AC. Angiotensin-(1-7) contributes to insulin-sensitizing effects of angiotensin-converting enzyme inhibition in obese mice. Am J Physiol Endocrinol Metab 2018; 315 (06) E1204-E1211
  • 61 Miranda K, Mehrpouya-Bahrami P, Nagarkatti PS, Nagarkatti M. Cannabinoid receptor 1 blockade attenuates obesity and adipose tissue type 1 inflammation through miR-30e-5p regulation of delta-like-4 in macrophages and consequently downregulation of Th1 cells. Front Immunol 2019; 10: 1049
  • 62 Shan B, Wang X, Wu Y. et al. The metabolic ER stress sensor IRE1α suppresses alternative activation of macrophages and impairs energy expenditure in obesity. Nat Immunol 2017; 18 (05) 519-529
  • 63 Guo X-F, Sinclair AJ, Kaur G, Li D. Differential effects of EPA, DPA and DHA on cardio-metabolic risk factors in high-fat diet fed mice. Prostaglandins Leukot Essent Fatty Acids 2018; 136: 47-55
  • 64 Hecker PA, O'Shea KM, Galvao TF, Brown BH, Stanley WC. Role of adiponectin in the development of high fat diet-induced metabolic abnormalities in mice. Horm Metab Res 2011; 43 (02) 100-105
  • 65 Kalupahana NS, Claycombe K, Newman SJ. et al. Eicosapentaenoic acid prevents and reverses insulin resistance in high-fat diet-induced obese mice via modulation of adipose tissue inflammation. J Nutr 2010; 140 (11) 1915-1922
  • 66 Flachs P, Mohamed-Ali V, Horakova O. et al. Polyunsaturated fatty acids of marine origin induce adiponectin in mice fed a high-fat diet. Diabetologia 2006; 49 (02) 394-397
  • 67 Hiel S, Neyrinck AM, Rodriguez J. et al. Inulin improves postprandial hypertriglyceridemia by modulating gene expression in the small intestine. Nutrients 2018; 10 (05) E532
  • 68 Katunga LA, Gudimella P, Efird JT. et al. Obesity in a model of gpx4 haploinsufficiency uncovers a causal role for lipid-derived aldehydes in human metabolic disease and cardiomyopathy. Mol Metab 2015; 4 (06) 493-506
  • 69 Laurila P-P, Soronen J, Kooijman S. et al. USF1 deficiency activates brown adipose tissue and improves cardiometabolic health. Sci Transl Med 2016; 8 (323) 323ra13
  • 70 Jin N, Wang Y, Liu L, Xue F, Jiang T, Xu M. Dysregulation of the renin-angiotensin system and cardiometabolic status in mice fed a long-term high-fat diet. Med Sci Monit 2019; 25: 6605-6614
  • 71 Cordero-Herrera I, Kozyra M, Zhuge Z. et al. AMP-activated protein kinase activation and NADPH oxidase inhibition by inorganic nitrate and nitrite prevent liver steatosis. Proc Natl Acad Sci U S A 2019; 116 (01) 217-226
  • 72 Schiattarella GG, Altamirano F, Tong D. et al. Nitrosative stress drives heart failure with preserved ejection fraction. Nature 2019; 568 (7752): 351-356
  • 73 Mendes-Junior LG, Freitas-Lima LC, Oliveira JR. et al. The usefulness of short-term high-fat/high salt diet as a model of metabolic syndrome in mice. Life Sci 2018; 209: 341-348
  • 74 Abdesselam I, Pepino P, Troalen T. et al. Time course of cardiometabolic alterations in a high fat high sucrose diet mice model and improvement after GLP-1 analog treatment using multimodal cardiovascular magnetic resonance. J Cardiovasc Magn Reson 2015; 17: 95
  • 75 Wang C-Y, Liao JK. A mouse model of diet-induced obesity and insulin resistance. Methods Mol Biol 2012; 821: 421-433
  • 76 Sims EK, Hatanaka M, Morris DL. et al. Divergent compensatory responses to high-fat diet between C57BL6/J and C57BLKS/J inbred mouse strains. Am J Physiol Endocrinol Metab 2013; 305 (12) E1495-E1511
  • 77 Blasco-Baque V, Coupé B, Fabre A. et al. Associations between hepatic miRNA expression, liver triacylglycerols and gut microbiota during metabolic adaptation to high-fat diet in mice. Diabetologia 2017; 60 (04) 690-700
  • 78 de Fourmestraux V, Neubauer H, Poussin C. et al. Transcript profiling suggests that differential metabolic adaptation of mice to a high fat diet is associated with changes in liver to muscle lipid fluxes. J Biol Chem 2004; 279 (49) 50743-50753
  • 79 Heydemann A. An overview of murine high fat diet as a model for type 2 diabetes mellitus. J Diabetes Res 2016; 2016: 2902351
  • 80 Stanley WC, Dabkowski ER, Ribeiro Jr RF, O'Connell KA. Dietary fat and heart failure: moving from lipotoxicity to lipoprotection. Circ Res 2012; 110 (05) 764-776
  • 81 Takahashi M, Ikemoto S, Ezaki O. Effect of the fat/carbohydrate ratio in the diet on obesity and oral glucose tolerance in C57BL/6J mice. J Nutr Sci Vitaminol (Tokyo) 1999; 45 (05) 583-593
  • 82 Ingvorsen C, Karp NA, Lelliott CJ. The role of sex and body weight on the metabolic effects of high-fat diet in C57BL/6N mice. Nutr Diabetes 2017; 7 (04) e261
  • 83 Eudave DM, BeLow MN, Flandreau EI. Effects of high fat or high sucrose diet on behavioral-response to social defeat stress in mice. Neurobiol Stress 2018; 9: 1-8
  • 84 Messa GAM, Piasecki M, Hurst J, Hill C, Tallis J, Degens H. The impact of a high-fat diet in mice is dependent on duration and age, and differs between muscles. J Exp Biol 2020; 223 (Pt 6): jeb217117
  • 85 Chatzigeorgiou A, Chavakis T. Immune cells and metabolism. Handb Exp Pharmacol 2016; 233: 221-249
  • 86 Shapiro MD, Fazio S. Apolipoprotein B-containing lipoproteins and atherosclerotic cardiovascular disease. F1000 Res 2017; 6: 134
  • 87 Al-Sulaiti H, Diboun I, Banu S. et al. Triglyceride profiling in adipose tissues from obese insulin sensitive, insulin resistant and type 2 diabetes mellitus individuals. J Transl Med 2018; 16 (01) 175
  • 88 Tu LN, Showalter MR, Cajka T. et al. Metabolomic characteristics of cholesterol-induced non-obese nonalcoholic fatty liver disease in mice. Sci Rep 2017; 7 (01) 6120
  • 89 Chiappini F, Coilly A, Kadar H. et al. Metabolism dysregulation induces a specific lipid signature of nonalcoholic steatohepatitis in patients. Sci Rep 2017; 7: 46658
  • 90 Garcia-Rios A, Alcala-Diaz JF, Gomez-Delgado F. et al. Beneficial effect of CETP gene polymorphism in combination with a Mediterranean diet influencing lipid metabolism in metabolic syndrome patients: CORDIOPREV study. Clin Nutr 2018; 37 (01) 229-234
  • 91 van den Hoek AM, van der Hoorn JWA, Maas AC. et al. APOE*3Leiden.CETP transgenic mice as model for pharmaceutical treatment of the metabolic syndrome. Diabetes Obes Metab 2014; 16 (06) 537-544
  • 92 Nasias D, Evangelakos I, Nidris V. et al. Significant changes in hepatic transcriptome and circulating miRNAs are associated with diet-induced metabolic syndrome in apoE3L.CETP mice. J Cell Physiol 2019; 234 (11) 20485-20500
  • 93 Casquero AC, Berti JA, Teixeira LLS, de Oliveira HCF. Chronic exercise reduces CETP and mesterolone treatment counteracts exercise benefits on plasma lipoproteins profile: studies in transgenic mice. Lipids 2017; 52 (12) 981-990
  • 94 Neuhofer A, Wernly B, Leitner L. et al. An accelerated mouse model for atherosclerosis and adipose tissue inflammation. Cardiovasc Diabetol 2014; 13: 23
  • 95 Soares e Silva AK, de Oliveira Cipriano Torres D, dos Santos Gomes FO. et al. LPSF/GQ-02 inhibits the development of hepatic steatosis and inflammation in a mouse model of non-alcoholic fatty liver disease (NAFLD). PLoS One 2015; 10 (04) e0123787
  • 96 Gromovsky AD, Schugar RC, Brown AL. et al. Δ-5 Fatty acid desaturase FADS1 impacts metabolic disease by balancing proinflammatory and proresolving lipid mediators. Arterioscler Thromb Vasc Biol 2018; 38 (01) 218-231
  • 97 Aravani D, Morris GE, Jones PD. et al. HHIPL1, a gene at the 14q32 coronary artery disease locus, positively regulates hedgehog signaling and promotes atherosclerosis. Circulation 2019; 140 (06) 500-513
  • 98 Scott NJA, Cameron VA, Raudsepp S. et al. Generation and characterization of a mouse model of the metabolic syndrome: apolipoprotein E and aromatase double knockout mice. Am J Physiol Endocrinol Metab 2012; 302 (05) E576-E584
  • 99 Scott NJA, Ellmers LJ, Pilbrow AP. et al. Metabolic and blood pressure effects of walnut supplementation in a mouse model of the metabolic syndrome. Nutrients 2017; 9 (07) E722
  • 100 Roche-Molina M, Sanz-Rosa D, Cruz FM. et al. Induction of sustained hypercholesterolemia by single adeno-associated virus-mediated gene transfer of mutant hPCSK9. Arterioscler Thromb Vasc Biol 2015; 35 (01) 50-59
  • 101 Bjørklund MM, Hollensen AK, Hagensen MK. et al. Induction of atherosclerosis in mice and hamsters without germline genetic engineering. Circ Res 2014; 114 (11) 1684-1689
  • 102 Davel AP, Lu Q, Moss ME. et al. Sex-specific mechanisms of resistance vessel endothelial dysfunction induced by cardiometabolic risk factors. J Am Heart Assoc 2018; 7 (04) e007675
  • 103 Goettsch C, Hutcheson JD, Hagita S. et al. A single injection of gain-of-function mutant PCSK9 adeno-associated virus vector induces cardiovascular calcification in mice with no genetic modification. Atherosclerosis 2016; 251: 109-118
  • 104 Kumar S, Kang D-W, Rezvan A, Jo H. Accelerated atherosclerosis development in C57Bl6 mice by overexpressing AAV-mediated PCSK9 and partial carotid ligation. Lab Invest 2017; 97 (08) 935-945
  • 105 Tauscher S, Nakagawa H, Völker K. et al. β Cell-specific deletion of guanylyl cyclase A, the receptor for atrial natriuretic peptide, accelerates obesity-induced glucose intolerance in mice. Cardiovasc Diabetol 2018; 17 (01) 103
  • 106 Ruohonen ST, Pesonen U, Moritz N. et al. Transgenic mice overexpressing neuropeptide Y in noradrenergic neurons: a novel model of increased adiposity and impaired glucose tolerance. Diabetes 2008; 57 (06) 1517-1525
  • 107 Wei Z, Zhang K, Wen G. et al. Heredity and cardiometabolic risk: naturally occurring polymorphisms in the human neuropeptide Y(2) receptor promoter disrupt multiple transcriptional response motifs. J Hypertens 2013; 31 (01) 123-133
  • 108 Tan CMJ, Green P, Tapoulal N, Lewandowski AJ, Leeson P, Herring N. The role of neuropeptide Y in cardiovascular health and disease. Front Physiol 2018; 9: 1281
  • 109 Xiao Y, Wang C, Chen J-Y. et al. Deficiency of PRKD2 triggers hyperinsulinemia and metabolic disorders. Nat Commun 2018; 9 (01) 2015
  • 110 Rockwood S, Broderick TL, Al-Nakkash L. Feeding obese diabetic mice a genistein diet induces thermogenic and metabolic change. J Med Food 2018; 21 (04) 332-339
  • 111 Wu H-K, Zhang Y, Cao C-M. et al. Glucose-sensitive myokine/cardiokine mg53 regulates systemic insulin response and metabolic homeostasis. Circulation 2019; 139 (07) 901-914
  • 112 Widjaja AA, Singh BK, Adami E. et al. Inhibiting interleukin 11 signaling reduces hepatocyte death and liver fibrosis, inflammation, and steatosis in mouse models of nonalcoholic steatohepatitis. Gastroenterology 2019; 157 (03) 777.e14-792.e14
  • 113 Adingupu DD, Göpel SO, Grönros J. et al. SGLT2 inhibition with empagliflozin improves coronary microvascular function and cardiac contractility in prediabetic ob/ob−/− mice. Cardiovasc Diabetol 2019; 18 (01) 16
  • 114 Trojanowski B, Salem HH, Neubauer H. et al. Elevated β-cell stress levels promote severe diabetes development in mice with MODY4. J Endocrinol 2020; 244 (02) 323-337
  • 115 Murphy MO, Herald JB, Leachman J, Villasante Tezanos A, Cohn DM, Loria AS. A model of neglect during postnatal life heightens obesity-induced hypertension and is linked to a greater metabolic compromise in female mice. Int J Obes 2018; 42 (07) 1354-1365
  • 116 Saad AF, Dickerson J, Kechichian TB. et al. High-fructose diet in pregnancy leads to fetal programming of hypertension, insulin resistance, and obesity in adult offspring. Am J Obstet Gynecol 2016; 215 (03) 378.e1-378.e6
  • 117 Loche E, Blackmore HL, Carpenter AA. et al. Maternal diet-induced obesity programmes cardiac dysfunction in male mice independently of post-weaning diet. Cardiovasc Res 2018; 114 (10) 1372-1384
  • 118 Seong HY, Cho HM, Kim M, Kim I. Maternal high-fructose intake induces multigenerational activation of the renin-angiotensin-aldosterone system. Hypertension 2019; 74 (03) 518-525
  • 119 Pataia V, Papacleovoulou G, Nikolova V. et al. Paternal cholestasis exacerbates obesity-associated hypertension in male offspring but is prevented by paternal ursodeoxycholic acid treatment. Int J Obes 2019; 43 (02) 319-330
  • 120 Mills V, Plows JF, Zhao H. et al. Effect of sildenafil citrate treatment in the eNOS knockout mouse model of fetal growth restriction on long-term cardiometabolic outcomes in male offspring. Pharmacol Res 2018; 137: 122-134
  • 121 Ozdemir AC, Wynn GM, Vester A. et al. GNB3 overexpression causes obesity and metabolic syndrome. PLoS One 2017; 12 (12) e0188763
  • 122 Valladolid-Acebes I, Daraio T, Brismar K. et al. Replacing SNAP-25b with SNAP-25a expression results in metabolic disease. Proc Natl Acad Sci U S A 2015; 112 (31) E4326-E4335
  • 123 Farese RV, Sajan MP, Yang H. et al. Muscle-specific knockout of PKC-lambda impairs glucose transport and induces metabolic and diabetic syndromes. J Clin Invest 2007; 117 (08) 2289-2301
  • 124 Sajan MP, Nimal S, Mastorides S. et al. Correction of metabolic abnormalities in a rodent model of obesity, metabolic syndrome, and type 2 diabetes mellitus by inhibitors of hepatic protein kinase C-ι. Metabolism 2012; 61 (04) 459-469
  • 125 Derecka M, Gornicka A, Koralov SB. et al. Tyk2 and Stat3 regulate brown adipose tissue differentiation and obesity. Cell Metab 2012; 16 (06) 814-824
  • 126 Bera TK, Liu X-F, Yamada M. et al. A model for obesity and gigantism due to disruption of the Ankrd26 gene. Proc Natl Acad Sci U S A 2008; 105 (01) 270-275
  • 127 Takayanagi Y, Matsumoto H, Nakata M. et al. Endogenous prolactin-releasing peptide regulates food intake in rodents. J Clin Invest 2008; 118 (12) 4014-4024
  • 128 Sha H, Xu J, Tang J. et al. Disruption of a novel regulatory locus results in decreased Bdnf expression, obesity, and type 2 diabetes in mice. Physiol Genomics 2007; 31 (02) 252-263
  • 129 Vartanian V, Lowell B, Minko IG. et al. The metabolic syndrome resulting from a knockout of the NEIL1 DNA glycosylase. Proc Natl Acad Sci U S A 2006; 103 (06) 1864-1869
  • 130 Bjursell M, Gerdin A-K, Jönsson M. et al. G protein-coupled receptor 12 deficiency results in dyslipidemia and obesity in mice. Biochem Biophys Res Commun 2006; 348 (02) 359-366
  • 131 Osswald C, Baumgarten K, Stümpel F. et al. Mice without the regulator gene Rsc1A1 exhibit increased Na+-D-glucose cotransport in small intestine and develop obesity. Mol Cell Biol 2005; 25 (01) 78-87
  • 132 Wiedmer T, Zhao J, Li L. et al. Adiposity, dyslipidemia, and insulin resistance in mice with targeted deletion of phospholipid scramblase 3 (PLSCR3). Proc Natl Acad Sci U S A 2004; 101 (36) 13296-13301
  • 133 Pan W, Ciociola E, Saraf M. et al. Metabolic consequences of ENPP1 overexpression in adipose tissue. Am J Physiol Endocrinol Metab 2011; 301 (05) E901-E911
  • 134 Masuzaki H, Paterson J, Shinyama H. et al. A transgenic model of visceral obesity and the metabolic syndrome. Science 2001; 294 (5549): 2166-2170
  • 135 Varga O, Harangi M, Olsson IA, Hansen AK. Contribution of animal models to the understanding of the metabolic syndrome: a systematic overview. Obes Rev 2010; 11 (11) 792-807
  • 136 Liang Y-Q, Isono M, Okamura T, Takeuchi F, Kato N. Alterations of lipid metabolism, blood pressure and fatty liver in spontaneously hypertensive rats transgenic for human cholesteryl ester transfer protein. Hypertens Res 2020; 43: 655-666
  • 137 Getz GS, Reardon CA. Animal models of atherosclerosis. Arterioscler Thromb Vasc Biol 2012; 32 (05) 1104-1115
  • 138 Zhang G, Byun HR, Ying Z. et al. Differential metabolic and multi-tissue transcriptomic responses to fructose consumption among genetically diverse mice. Biochim Biophys Acta Mol Basis Dis 2020; 1866 (01) 165569
  • 139 González-Blázquez R, Alcalá M, Fernández-Alfonso MS. et al. Relevance of control diet choice in metabolic studies: impact in glucose homeostasis and vascular function. Sci Rep 2020; 10 (01) 2902
  • 140 Prendergast BJ, Onishi KG, Zucker I. Female mice liberated for inclusion in neuroscience and biomedical research. Neurosci Biobehav Rev 2014; 40: 1-5
  • 141 Mauvais-Jarvis F, Arnold AP, Reue K. A guide for the design of pre-clinical studies on sex differences in metabolism. Cell Metab 2017; 25 (06) 1216-1230
  • 142 Libby P. Murine “model” monotheism: an iconoclast at the altar of mouse. Circ Res 2015; 117 (11) 921-925

Address for correspondence

Antonios Chatzigeorgiou, MD, PhD
Department of Physiology, Medical School, National and Kapodistrian University of Athens
75 Mikras Asias Street, 11527 Athens
Greece   
Styliani Vakrou, MD, PhD
Department of Physiology, Medical School, National and Kapodistrian University of Athens
75 Mikras Asias Street, 11527 Athens
Greece   

Publication History

Received: 03 June 2020

Accepted: 24 October 2020

Article published online:
06 December 2020

© 2020. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References

  • 1 Kylin E. Studien ueber das hypertonie-hyperglyka. Zentralblatt Fuer Inn Med 1923; 44: 105-127
  • 2 Vague J. The degree of masculine differentiation of obesities: a factor determining predisposition to diabetes, atherosclerosis, gout, and uric calculous disease. Am J Clin Nutr 1956; 4 (01) 20-34
  • 3 Alberti KGMM, Eckel RH, Grundy SM. et al; International Diabetes Federation Task Force on Epidemiology and Prevention, Hational Heart, Lung, and Blood Institute, American Heart Association; World Heart Federation, International Atherosclerosis Society, International Association for the Study of Obesity. Harmonizing the metabolic syndrome: a joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; and International Association for the Study of Obesity. Circulation 2009; 120 (16) 1640-1645
  • 4 Grundy SM, Brewer Jr HB, Cleeman JI, Smith Jr SC, Lenfant C. American Heart Association, National Heart, Lung, and Blood Institute. Definition of metabolic syndrome: report of the National Heart, Lung, and Blood Institute/American Heart Association conference on scientific issues related to definition. Circulation 2004; 109 (03) 433-438
  • 5 Castro JP, El-Atat FA, McFarlane SI, Aneja A, Sowers JR. Cardiometabolic syndrome: pathophysiology and treatment. Curr Hypertens Rep 2003; 5 (05) 393-401
  • 6 Grundy SM, Cleeman JI, Daniels SR. et al; American Heart Association, National Heart, Lung, and Blood Institute. Diagnosis and management of the metabolic syndrome: an American Heart Association/National Heart, Lung, and Blood Institute Scientific statement. Circulation 2005; 112 (17) 2735-2752
  • 7 Mottillo S, Filion KB, Genest J. et al. The metabolic syndrome and cardiovascular risk a systematic review and meta-analysis. J Am Coll Cardiol 2010; 56 (14) 1113-1132
  • 8 Yates KF, Sweat V, Yau PL, Turchiano MM, Convit A. Impact of metabolic syndrome on cognition and brain: a selected review of the literature. Arterioscler Thromb Vasc Biol 2012; 32 (09) 2060-2067
  • 9 Aguilar M, Bhuket T, Torres S, Liu B, Wong RJ. Prevalence of the metabolic syndrome in the United States, 2003-2012. JAMA 2015; 313 (19) 1973-1974
  • 10 Sherling DH, Perumareddi P, Hennekens CH. Metabolic syndrome. J Cardiovasc Pharmacol Ther 2017; 22 (04) 365-367
  • 11 Pradhan AD. Sex differences in the metabolic syndrome: implications for cardiovascular health in women. Clin Chem 2014; 60 (01) 44-52
  • 12 Sigit FS, Tahapary DL, Trompet S. et al. The prevalence of metabolic syndrome and its association with body fat distribution in middle-aged individuals from Indonesia and the Netherlands: a cross-sectional analysis of two population-based studies. Diabetol Metab Syndr 2020; 12: 2
  • 13 Saklayen MG. The global epidemic of the metabolic syndrome. Curr Hypertens Rep 2018; 20 (02) 12
  • 14 Ford ES, Li C, Sattar N. Metabolic syndrome and incident diabetes: current state of the evidence. Diabetes Care 2008; 31 (09) 1898-1904
  • 15 Zhang D, Tang X, Shen P. et al. Multimorbidity of cardiometabolic diseases: prevalence and risk for mortality from one million Chinese adults in a longitudinal cohort study. BMJ Open 2019; 9 (03) e024476
  • 16 Chatzigeorgiou A, Kandaraki E, Papavassiliou AG, Koutsilieris M. Peripheral targets in obesity treatment: a comprehensive update. Obes Rev 2014; 15 (06) 487-503
  • 17 Vakrou S, Abraham MR. Hypertrophic cardiomyopathy: a heart in need of an energy bar?. Front Physiol 2014; 5: 309
  • 18 Halapas A, Papalois A, Stauropoulou A. et al. In vivo models for heart failure research. In Vivo 2008; 22 (06) 767-780
  • 19 Santhekadur PK, Kumar DP, Sanyal AJ. Preclinical models of non-alcoholic fatty liver disease. J Hepatol 2018; 68 (02) 230-237
  • 20 Perlman RL. Mouse models of human disease: An evolutionary perspective. Evol Med Public Health 2016; 2016 (01) 170-176
  • 21 Karunakaran S, Clee SM. Genetics of metabolic syndrome: potential clues from wild-derived inbred mouse strains. Physiol Genomics 2018; 50 (01) 35-51
  • 22 Ishibashi S, Brown MS, Goldstein JL, Gerard RD, Hammer RE, Herz J. Hypercholesterolemia in low density lipoprotein receptor knockout mice and its reversal by adenovirus-mediated gene delivery. J Clin Invest 1993; 92 (02) 883-893
  • 23 Plump AS, Smith JD, Hayek T. et al. Severe hypercholesterolemia and atherosclerosis in apolipoprotein E-deficient mice created by homologous recombination in ES cells. Cell 1992; 71 (02) 343-353
  • 24 Wu L, Vikramadithyan R, Yu S. et al. Addition of dietary fat to cholesterol in the diets of LDL receptor knockout mice: effects on plasma insulin, lipoproteins, and atherosclerosis. J Lipid Res 2006; 47 (10) 2215-2222
  • 25 Gao J, Katagiri H, Ishigaki Y. et al. Involvement of apolipoprotein E in excess fat accumulation and insulin resistance. Diabetes 2007; 56 (01) 24-33
  • 26 Hofmann SM, Perez-Tilve D, Greer TM. et al. Defective lipid delivery modulates glucose tolerance and metabolic response to diet in apolipoprotein E-deficient mice. Diabetes 2008; 57 (01) 5-12
  • 27 King VL, Hatch NW, Chan H-W, de Beer MC, de Beer FC, Tannock LR. A murine model of obesity with accelerated atherosclerosis. Obesity (Silver Spring) 2010; 18 (01) 35-41
  • 28 Getz GS, Reardon CA. Do the Apoe−/− and Ldlr−/−mice yield the same insight on atherogenesis?. Arterioscler Thromb Vasc Biol 2016; 36 (09) 1734-1741
  • 29 Papaetis GS, Papakyriakou P, Panagiotou TN. Central obesity, type 2 diabetes and insulin: exploring a pathway full of thorns. Arch Med Sci 2015; 11 (03) 463-482
  • 30 Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature 1994; 372 (6505): 425-432
  • 31 Haluzik M, Colombo C, Gavrilova O. et al. Genetic background (C57BL/6J versus FVB/N) strongly influences the severity of diabetes and insulin resistance in ob/ob mice. Endocrinology 2004; 145 (07) 3258-3264
  • 32 Qiu J, Ogus S, Mounzih K, Ewart-Toland A, Chehab FF. Leptin-deficient mice backcrossed to the BALB/cJ genetic background have reduced adiposity, enhanced fertility, normal body temperature, and severe diabetes. Endocrinology 2001; 142 (08) 3421-3425
  • 33 Hummel KP, Dickie MM, Coleman DL. Diabetes, a new mutation in the mouse. Science 1966; 153 (3740): 1127-1128
  • 34 Kennedy AJ, Ellacott KLJ, King VL, Hasty AH. Mouse models of the metabolic syndrome. Dis Model Mech 2010; 3 (3–4): 156-166
  • 35 Charles River. The C57BL/6NCrl-Leprdb-lb/Crl mouse: a model for metabolic syndrome/pre-diabetes. Accessed April 21, 2020 at: https://www.criver.com/sites/default/files/resources/rm_rm_r_POUND_MOUSE_fact_sheet.pdf
  • 36 Frigeri LG, Wolff GL, Teguh C. Differential responses of yellow Avy/A and agouti A/a (BALB/c X VY) F1 hybrid mice to the same diets: glucose tolerance, weight gain, and adipocyte cellularity. Int J Obes 1988; 12 (04) 305-320
  • 37 Mark AL, Shaffer RA, Correia ML, Morgan DA, Sigmund CD, Haynes WG. Contrasting blood pressure effects of obesity in leptin-deficient ob/ob mice and agouti yellow obese mice. J Hypertens 1999; 17 (12, Pt 2): 1949-1953
  • 38 Yeo GS, Farooqi IS, Aminian S, Halsall DJ, Stanhope RG, O'Rahilly S. A frameshift mutation in MC4R associated with dominantly inherited human obesity. Nat Genet 1998; 20 (02) 111-112
  • 39 Vaisse C, Clement K, Durand E, Hercberg S, Guy-Grand B, Froguel P. Melanocortin-4 receptor mutations are a frequent and heterogeneous cause of morbid obesity. J Clin Invest 2000; 106 (02) 253-262
  • 40 Huszar D, Lynch CA, Fairchild-Huntress V. et al. Targeted disruption of the melanocortin-4 receptor results in obesity in mice. Cell 1997; 88 (01) 131-141
  • 41 Lede V, Meusel A, Garten A. et al. Altered hepatic lipid metabolism in mice lacking both the melanocortin type 4 receptor and low density lipoprotein receptor. PLoS One 2017; 12 (02) e0172000
  • 42 Iqbal J, Li X, Chang BH-J. et al. An intrinsic gut leptin-melanocortin pathway modulates intestinal microsomal triglyceride transfer protein and lipid absorption. J Lipid Res 2010; 51 (07) 1929-1942
  • 43 Cho Y-R, Kim H-J, Park S-Y. et al. Hyperglycemia, maturity-onset obesity, and insulin resistance in NONcNZO10/LtJ males, a new mouse model of type 2 diabetes. Am J Physiol Endocrinol Metab 2007; 293 (01) E327-E336
  • 44 Nigro E, Scudiero O, Monaco ML. et al. New insight into adiponectin role in obesity and obesity-related diseases. BioMed Res Int 2014; 2014: 658913
  • 45 Ouchi N, Ohishi M, Kihara S. et al. Association of hypoadiponectinemia with impaired vasoreactivity. Hypertension 2003; 42 (03) 231-234
  • 46 Ohashi K, Kihara S, Ouchi N. et al. Adiponectin replenishment ameliorates obesity-related hypertension. Hypertension 2006; 47 (06) 1108-1116
  • 47 Ryan MJ, McLemore Jr GR, Hendrix ST. Insulin resistance and obesity in a mouse model of systemic lupus erythematosus. Hypertension 2006; 48 (05) 988-993
  • 48 Moitra J, Mason MM, Olive M. et al. Life without white fat: a transgenic mouse. Genes Dev 1998; 12 (20) 3168-3181
  • 49 Shimomura I, Hammer RE, Richardson JA. et al. Insulin resistance and diabetes mellitus in transgenic mice expressing nuclear SREBP-1c in adipose tissue: model for congenital generalized lipodystrophy. Genes Dev 1998; 12 (20) 3182-3194
  • 50 Takemori K, Gao Y-J, Ding L. et al. Elevated blood pressure in transgenic lipoatrophic mice and altered vascular function. Hypertension 2007; 49 (02) 365-372
  • 51 Hörbelt T, Knebel B, Fahlbusch P. et al. The adipokine sFRP4 induces insulin resistance and lipogenesis in the liver. Biochim Biophys Acta Mol Basis Dis 2019; 1865 (10) 2671-2684
  • 52 Wong SK, Chin K-Y, Suhaimi FH, Fairus A, Ima-Nirwana S. Animal models of metabolic syndrome: a review. Nutr Metab (Lond) 2016; 13: 65
  • 53 Buettner R, Schölmerich J, Bollheimer LC. High-fat diets: modeling the metabolic disorders of human obesity in rodents. Obesity (Silver Spring) 2007; 15 (04) 798-808
  • 54 Ghibaudi L, Cook J, Farley C, van Heek M, Hwa JJ. Fat intake affects adiposity, comorbidity factors, and energy metabolism of sprague-dawley rats. Obes Res 2002; 10 (09) 956-963
  • 55 Johnson RJ, Segal MS, Sautin Y. et al. Potential role of sugar (fructose) in the epidemic of hypertension, obesity and the metabolic syndrome, diabetes, kidney disease, and cardiovascular disease. Am J Clin Nutr 2007; 86 (04) 899-906
  • 56 Rasool S, Geetha T, Broderick TL, Babu JR. High fat with high sucrose diet leads to obesity and induces myodegeneration. Front Physiol 2018; 9: 1054
  • 57 Glendinning JI, Breinager L, Kyrillou E, Lacuna K, Rocha R, Sclafani A. Differential effects of sucrose and fructose on dietary obesity in four mouse strains. Physiol Behav 2010; 101 (03) 331-343
  • 58 Furman BL. Streptozotocin-induced diabetic models in mice and rats. Curr Protoc Pharmacol 2015; 70: 1-20
  • 59 Fransson L, Franzén S, Rosengren V, Wolbert P, Sjöholm Å, Ortsäter H. β-Cell adaptation in a mouse model of glucocorticoid-induced metabolic syndrome. J Endocrinol 2013; 219 (03) 231-241
  • 60 Loloi J, Miller AJ, Bingaman SS, Silberman Y, Arnold AC. Angiotensin-(1-7) contributes to insulin-sensitizing effects of angiotensin-converting enzyme inhibition in obese mice. Am J Physiol Endocrinol Metab 2018; 315 (06) E1204-E1211
  • 61 Miranda K, Mehrpouya-Bahrami P, Nagarkatti PS, Nagarkatti M. Cannabinoid receptor 1 blockade attenuates obesity and adipose tissue type 1 inflammation through miR-30e-5p regulation of delta-like-4 in macrophages and consequently downregulation of Th1 cells. Front Immunol 2019; 10: 1049
  • 62 Shan B, Wang X, Wu Y. et al. The metabolic ER stress sensor IRE1α suppresses alternative activation of macrophages and impairs energy expenditure in obesity. Nat Immunol 2017; 18 (05) 519-529
  • 63 Guo X-F, Sinclair AJ, Kaur G, Li D. Differential effects of EPA, DPA and DHA on cardio-metabolic risk factors in high-fat diet fed mice. Prostaglandins Leukot Essent Fatty Acids 2018; 136: 47-55
  • 64 Hecker PA, O'Shea KM, Galvao TF, Brown BH, Stanley WC. Role of adiponectin in the development of high fat diet-induced metabolic abnormalities in mice. Horm Metab Res 2011; 43 (02) 100-105
  • 65 Kalupahana NS, Claycombe K, Newman SJ. et al. Eicosapentaenoic acid prevents and reverses insulin resistance in high-fat diet-induced obese mice via modulation of adipose tissue inflammation. J Nutr 2010; 140 (11) 1915-1922
  • 66 Flachs P, Mohamed-Ali V, Horakova O. et al. Polyunsaturated fatty acids of marine origin induce adiponectin in mice fed a high-fat diet. Diabetologia 2006; 49 (02) 394-397
  • 67 Hiel S, Neyrinck AM, Rodriguez J. et al. Inulin improves postprandial hypertriglyceridemia by modulating gene expression in the small intestine. Nutrients 2018; 10 (05) E532
  • 68 Katunga LA, Gudimella P, Efird JT. et al. Obesity in a model of gpx4 haploinsufficiency uncovers a causal role for lipid-derived aldehydes in human metabolic disease and cardiomyopathy. Mol Metab 2015; 4 (06) 493-506
  • 69 Laurila P-P, Soronen J, Kooijman S. et al. USF1 deficiency activates brown adipose tissue and improves cardiometabolic health. Sci Transl Med 2016; 8 (323) 323ra13
  • 70 Jin N, Wang Y, Liu L, Xue F, Jiang T, Xu M. Dysregulation of the renin-angiotensin system and cardiometabolic status in mice fed a long-term high-fat diet. Med Sci Monit 2019; 25: 6605-6614
  • 71 Cordero-Herrera I, Kozyra M, Zhuge Z. et al. AMP-activated protein kinase activation and NADPH oxidase inhibition by inorganic nitrate and nitrite prevent liver steatosis. Proc Natl Acad Sci U S A 2019; 116 (01) 217-226
  • 72 Schiattarella GG, Altamirano F, Tong D. et al. Nitrosative stress drives heart failure with preserved ejection fraction. Nature 2019; 568 (7752): 351-356
  • 73 Mendes-Junior LG, Freitas-Lima LC, Oliveira JR. et al. The usefulness of short-term high-fat/high salt diet as a model of metabolic syndrome in mice. Life Sci 2018; 209: 341-348
  • 74 Abdesselam I, Pepino P, Troalen T. et al. Time course of cardiometabolic alterations in a high fat high sucrose diet mice model and improvement after GLP-1 analog treatment using multimodal cardiovascular magnetic resonance. J Cardiovasc Magn Reson 2015; 17: 95
  • 75 Wang C-Y, Liao JK. A mouse model of diet-induced obesity and insulin resistance. Methods Mol Biol 2012; 821: 421-433
  • 76 Sims EK, Hatanaka M, Morris DL. et al. Divergent compensatory responses to high-fat diet between C57BL6/J and C57BLKS/J inbred mouse strains. Am J Physiol Endocrinol Metab 2013; 305 (12) E1495-E1511
  • 77 Blasco-Baque V, Coupé B, Fabre A. et al. Associations between hepatic miRNA expression, liver triacylglycerols and gut microbiota during metabolic adaptation to high-fat diet in mice. Diabetologia 2017; 60 (04) 690-700
  • 78 de Fourmestraux V, Neubauer H, Poussin C. et al. Transcript profiling suggests that differential metabolic adaptation of mice to a high fat diet is associated with changes in liver to muscle lipid fluxes. J Biol Chem 2004; 279 (49) 50743-50753
  • 79 Heydemann A. An overview of murine high fat diet as a model for type 2 diabetes mellitus. J Diabetes Res 2016; 2016: 2902351
  • 80 Stanley WC, Dabkowski ER, Ribeiro Jr RF, O'Connell KA. Dietary fat and heart failure: moving from lipotoxicity to lipoprotection. Circ Res 2012; 110 (05) 764-776
  • 81 Takahashi M, Ikemoto S, Ezaki O. Effect of the fat/carbohydrate ratio in the diet on obesity and oral glucose tolerance in C57BL/6J mice. J Nutr Sci Vitaminol (Tokyo) 1999; 45 (05) 583-593
  • 82 Ingvorsen C, Karp NA, Lelliott CJ. The role of sex and body weight on the metabolic effects of high-fat diet in C57BL/6N mice. Nutr Diabetes 2017; 7 (04) e261
  • 83 Eudave DM, BeLow MN, Flandreau EI. Effects of high fat or high sucrose diet on behavioral-response to social defeat stress in mice. Neurobiol Stress 2018; 9: 1-8
  • 84 Messa GAM, Piasecki M, Hurst J, Hill C, Tallis J, Degens H. The impact of a high-fat diet in mice is dependent on duration and age, and differs between muscles. J Exp Biol 2020; 223 (Pt 6): jeb217117
  • 85 Chatzigeorgiou A, Chavakis T. Immune cells and metabolism. Handb Exp Pharmacol 2016; 233: 221-249
  • 86 Shapiro MD, Fazio S. Apolipoprotein B-containing lipoproteins and atherosclerotic cardiovascular disease. F1000 Res 2017; 6: 134
  • 87 Al-Sulaiti H, Diboun I, Banu S. et al. Triglyceride profiling in adipose tissues from obese insulin sensitive, insulin resistant and type 2 diabetes mellitus individuals. J Transl Med 2018; 16 (01) 175
  • 88 Tu LN, Showalter MR, Cajka T. et al. Metabolomic characteristics of cholesterol-induced non-obese nonalcoholic fatty liver disease in mice. Sci Rep 2017; 7 (01) 6120
  • 89 Chiappini F, Coilly A, Kadar H. et al. Metabolism dysregulation induces a specific lipid signature of nonalcoholic steatohepatitis in patients. Sci Rep 2017; 7: 46658
  • 90 Garcia-Rios A, Alcala-Diaz JF, Gomez-Delgado F. et al. Beneficial effect of CETP gene polymorphism in combination with a Mediterranean diet influencing lipid metabolism in metabolic syndrome patients: CORDIOPREV study. Clin Nutr 2018; 37 (01) 229-234
  • 91 van den Hoek AM, van der Hoorn JWA, Maas AC. et al. APOE*3Leiden.CETP transgenic mice as model for pharmaceutical treatment of the metabolic syndrome. Diabetes Obes Metab 2014; 16 (06) 537-544
  • 92 Nasias D, Evangelakos I, Nidris V. et al. Significant changes in hepatic transcriptome and circulating miRNAs are associated with diet-induced metabolic syndrome in apoE3L.CETP mice. J Cell Physiol 2019; 234 (11) 20485-20500
  • 93 Casquero AC, Berti JA, Teixeira LLS, de Oliveira HCF. Chronic exercise reduces CETP and mesterolone treatment counteracts exercise benefits on plasma lipoproteins profile: studies in transgenic mice. Lipids 2017; 52 (12) 981-990
  • 94 Neuhofer A, Wernly B, Leitner L. et al. An accelerated mouse model for atherosclerosis and adipose tissue inflammation. Cardiovasc Diabetol 2014; 13: 23
  • 95 Soares e Silva AK, de Oliveira Cipriano Torres D, dos Santos Gomes FO. et al. LPSF/GQ-02 inhibits the development of hepatic steatosis and inflammation in a mouse model of non-alcoholic fatty liver disease (NAFLD). PLoS One 2015; 10 (04) e0123787
  • 96 Gromovsky AD, Schugar RC, Brown AL. et al. Δ-5 Fatty acid desaturase FADS1 impacts metabolic disease by balancing proinflammatory and proresolving lipid mediators. Arterioscler Thromb Vasc Biol 2018; 38 (01) 218-231
  • 97 Aravani D, Morris GE, Jones PD. et al. HHIPL1, a gene at the 14q32 coronary artery disease locus, positively regulates hedgehog signaling and promotes atherosclerosis. Circulation 2019; 140 (06) 500-513
  • 98 Scott NJA, Cameron VA, Raudsepp S. et al. Generation and characterization of a mouse model of the metabolic syndrome: apolipoprotein E and aromatase double knockout mice. Am J Physiol Endocrinol Metab 2012; 302 (05) E576-E584
  • 99 Scott NJA, Ellmers LJ, Pilbrow AP. et al. Metabolic and blood pressure effects of walnut supplementation in a mouse model of the metabolic syndrome. Nutrients 2017; 9 (07) E722
  • 100 Roche-Molina M, Sanz-Rosa D, Cruz FM. et al. Induction of sustained hypercholesterolemia by single adeno-associated virus-mediated gene transfer of mutant hPCSK9. Arterioscler Thromb Vasc Biol 2015; 35 (01) 50-59
  • 101 Bjørklund MM, Hollensen AK, Hagensen MK. et al. Induction of atherosclerosis in mice and hamsters without germline genetic engineering. Circ Res 2014; 114 (11) 1684-1689
  • 102 Davel AP, Lu Q, Moss ME. et al. Sex-specific mechanisms of resistance vessel endothelial dysfunction induced by cardiometabolic risk factors. J Am Heart Assoc 2018; 7 (04) e007675
  • 103 Goettsch C, Hutcheson JD, Hagita S. et al. A single injection of gain-of-function mutant PCSK9 adeno-associated virus vector induces cardiovascular calcification in mice with no genetic modification. Atherosclerosis 2016; 251: 109-118
  • 104 Kumar S, Kang D-W, Rezvan A, Jo H. Accelerated atherosclerosis development in C57Bl6 mice by overexpressing AAV-mediated PCSK9 and partial carotid ligation. Lab Invest 2017; 97 (08) 935-945
  • 105 Tauscher S, Nakagawa H, Völker K. et al. β Cell-specific deletion of guanylyl cyclase A, the receptor for atrial natriuretic peptide, accelerates obesity-induced glucose intolerance in mice. Cardiovasc Diabetol 2018; 17 (01) 103
  • 106 Ruohonen ST, Pesonen U, Moritz N. et al. Transgenic mice overexpressing neuropeptide Y in noradrenergic neurons: a novel model of increased adiposity and impaired glucose tolerance. Diabetes 2008; 57 (06) 1517-1525
  • 107 Wei Z, Zhang K, Wen G. et al. Heredity and cardiometabolic risk: naturally occurring polymorphisms in the human neuropeptide Y(2) receptor promoter disrupt multiple transcriptional response motifs. J Hypertens 2013; 31 (01) 123-133
  • 108 Tan CMJ, Green P, Tapoulal N, Lewandowski AJ, Leeson P, Herring N. The role of neuropeptide Y in cardiovascular health and disease. Front Physiol 2018; 9: 1281
  • 109 Xiao Y, Wang C, Chen J-Y. et al. Deficiency of PRKD2 triggers hyperinsulinemia and metabolic disorders. Nat Commun 2018; 9 (01) 2015
  • 110 Rockwood S, Broderick TL, Al-Nakkash L. Feeding obese diabetic mice a genistein diet induces thermogenic and metabolic change. J Med Food 2018; 21 (04) 332-339
  • 111 Wu H-K, Zhang Y, Cao C-M. et al. Glucose-sensitive myokine/cardiokine mg53 regulates systemic insulin response and metabolic homeostasis. Circulation 2019; 139 (07) 901-914
  • 112 Widjaja AA, Singh BK, Adami E. et al. Inhibiting interleukin 11 signaling reduces hepatocyte death and liver fibrosis, inflammation, and steatosis in mouse models of nonalcoholic steatohepatitis. Gastroenterology 2019; 157 (03) 777.e14-792.e14
  • 113 Adingupu DD, Göpel SO, Grönros J. et al. SGLT2 inhibition with empagliflozin improves coronary microvascular function and cardiac contractility in prediabetic ob/ob−/− mice. Cardiovasc Diabetol 2019; 18 (01) 16
  • 114 Trojanowski B, Salem HH, Neubauer H. et al. Elevated β-cell stress levels promote severe diabetes development in mice with MODY4. J Endocrinol 2020; 244 (02) 323-337
  • 115 Murphy MO, Herald JB, Leachman J, Villasante Tezanos A, Cohn DM, Loria AS. A model of neglect during postnatal life heightens obesity-induced hypertension and is linked to a greater metabolic compromise in female mice. Int J Obes 2018; 42 (07) 1354-1365
  • 116 Saad AF, Dickerson J, Kechichian TB. et al. High-fructose diet in pregnancy leads to fetal programming of hypertension, insulin resistance, and obesity in adult offspring. Am J Obstet Gynecol 2016; 215 (03) 378.e1-378.e6
  • 117 Loche E, Blackmore HL, Carpenter AA. et al. Maternal diet-induced obesity programmes cardiac dysfunction in male mice independently of post-weaning diet. Cardiovasc Res 2018; 114 (10) 1372-1384
  • 118 Seong HY, Cho HM, Kim M, Kim I. Maternal high-fructose intake induces multigenerational activation of the renin-angiotensin-aldosterone system. Hypertension 2019; 74 (03) 518-525
  • 119 Pataia V, Papacleovoulou G, Nikolova V. et al. Paternal cholestasis exacerbates obesity-associated hypertension in male offspring but is prevented by paternal ursodeoxycholic acid treatment. Int J Obes 2019; 43 (02) 319-330
  • 120 Mills V, Plows JF, Zhao H. et al. Effect of sildenafil citrate treatment in the eNOS knockout mouse model of fetal growth restriction on long-term cardiometabolic outcomes in male offspring. Pharmacol Res 2018; 137: 122-134
  • 121 Ozdemir AC, Wynn GM, Vester A. et al. GNB3 overexpression causes obesity and metabolic syndrome. PLoS One 2017; 12 (12) e0188763
  • 122 Valladolid-Acebes I, Daraio T, Brismar K. et al. Replacing SNAP-25b with SNAP-25a expression results in metabolic disease. Proc Natl Acad Sci U S A 2015; 112 (31) E4326-E4335
  • 123 Farese RV, Sajan MP, Yang H. et al. Muscle-specific knockout of PKC-lambda impairs glucose transport and induces metabolic and diabetic syndromes. J Clin Invest 2007; 117 (08) 2289-2301
  • 124 Sajan MP, Nimal S, Mastorides S. et al. Correction of metabolic abnormalities in a rodent model of obesity, metabolic syndrome, and type 2 diabetes mellitus by inhibitors of hepatic protein kinase C-ι. Metabolism 2012; 61 (04) 459-469
  • 125 Derecka M, Gornicka A, Koralov SB. et al. Tyk2 and Stat3 regulate brown adipose tissue differentiation and obesity. Cell Metab 2012; 16 (06) 814-824
  • 126 Bera TK, Liu X-F, Yamada M. et al. A model for obesity and gigantism due to disruption of the Ankrd26 gene. Proc Natl Acad Sci U S A 2008; 105 (01) 270-275
  • 127 Takayanagi Y, Matsumoto H, Nakata M. et al. Endogenous prolactin-releasing peptide regulates food intake in rodents. J Clin Invest 2008; 118 (12) 4014-4024
  • 128 Sha H, Xu J, Tang J. et al. Disruption of a novel regulatory locus results in decreased Bdnf expression, obesity, and type 2 diabetes in mice. Physiol Genomics 2007; 31 (02) 252-263
  • 129 Vartanian V, Lowell B, Minko IG. et al. The metabolic syndrome resulting from a knockout of the NEIL1 DNA glycosylase. Proc Natl Acad Sci U S A 2006; 103 (06) 1864-1869
  • 130 Bjursell M, Gerdin A-K, Jönsson M. et al. G protein-coupled receptor 12 deficiency results in dyslipidemia and obesity in mice. Biochem Biophys Res Commun 2006; 348 (02) 359-366
  • 131 Osswald C, Baumgarten K, Stümpel F. et al. Mice without the regulator gene Rsc1A1 exhibit increased Na+-D-glucose cotransport in small intestine and develop obesity. Mol Cell Biol 2005; 25 (01) 78-87
  • 132 Wiedmer T, Zhao J, Li L. et al. Adiposity, dyslipidemia, and insulin resistance in mice with targeted deletion of phospholipid scramblase 3 (PLSCR3). Proc Natl Acad Sci U S A 2004; 101 (36) 13296-13301
  • 133 Pan W, Ciociola E, Saraf M. et al. Metabolic consequences of ENPP1 overexpression in adipose tissue. Am J Physiol Endocrinol Metab 2011; 301 (05) E901-E911
  • 134 Masuzaki H, Paterson J, Shinyama H. et al. A transgenic model of visceral obesity and the metabolic syndrome. Science 2001; 294 (5549): 2166-2170
  • 135 Varga O, Harangi M, Olsson IA, Hansen AK. Contribution of animal models to the understanding of the metabolic syndrome: a systematic overview. Obes Rev 2010; 11 (11) 792-807
  • 136 Liang Y-Q, Isono M, Okamura T, Takeuchi F, Kato N. Alterations of lipid metabolism, blood pressure and fatty liver in spontaneously hypertensive rats transgenic for human cholesteryl ester transfer protein. Hypertens Res 2020; 43: 655-666
  • 137 Getz GS, Reardon CA. Animal models of atherosclerosis. Arterioscler Thromb Vasc Biol 2012; 32 (05) 1104-1115
  • 138 Zhang G, Byun HR, Ying Z. et al. Differential metabolic and multi-tissue transcriptomic responses to fructose consumption among genetically diverse mice. Biochim Biophys Acta Mol Basis Dis 2020; 1866 (01) 165569
  • 139 González-Blázquez R, Alcalá M, Fernández-Alfonso MS. et al. Relevance of control diet choice in metabolic studies: impact in glucose homeostasis and vascular function. Sci Rep 2020; 10 (01) 2902
  • 140 Prendergast BJ, Onishi KG, Zucker I. Female mice liberated for inclusion in neuroscience and biomedical research. Neurosci Biobehav Rev 2014; 40: 1-5
  • 141 Mauvais-Jarvis F, Arnold AP, Reue K. A guide for the design of pre-clinical studies on sex differences in metabolism. Cell Metab 2017; 25 (06) 1216-1230
  • 142 Libby P. Murine “model” monotheism: an iconoclast at the altar of mouse. Circ Res 2015; 117 (11) 921-925