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.

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




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