Next Article in Journal
Cycling for Weight Loss May Clear Carbohydrates Rather Than Fat, Irrespective of Normal or Mildly Reduced Normobaric Oxygen
Previous Article in Journal
Ongoing Community-Based Whole-Food, Plant-Based Lifestyle Effectively Preserves Muscle Mass during Body Mass Loss
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Obesity Rodent Models Applied to Research with Food Products and Natural Compounds

by
Tânia Martins
1,2,3,
Tiago Ferreira
1,2,
Elisabete Nascimento-Gonçalves
1,2,
Catarina Castro-Ribeiro
1,2,
Sílvia Lemos
1,2,
Eduardo Rosa
1,2,
Luís Miguel Antunes
1,2,3 and
Paula Alexandra Oliveira
1,2,3,*
1
Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
2
Inov4Agro-Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, UTAD, 5000-801 Vila Real, Portugal
3
Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences, UTAD, 5000-801 Vila Real, Portugal
*
Author to whom correspondence should be addressed.
Obesities 2022, 2(2), 171-204; https://doi.org/10.3390/obesities2020015
Submission received: 8 February 2022 / Revised: 24 March 2022 / Accepted: 2 April 2022 / Published: 6 April 2022

Abstract

:
Obesity is a disease whose incidence has increased over the last few decades. Despite being a multifactorial disease, obesity results essentially from excessive intake of high-calorie foods associated with low physical activity. The demand for a pharmacological therapy using natural compounds as an alternative to synthetic drugs has increased. Natural compounds may have few adverse effects and high economic impact, as most of them can be extracted from underexploited plant species and food by-products. To test the potential anti-obesogenic effects of new natural substances, the use of preclinical animal models of obesity has been an important tool, among which rat and mouse models are the most used. Some animal models are monogenic, such as the db/db mice, ob/ob mice, Zucker fatty rat and Otsuka Long-Evans Tokushima fatty rat. There are also available chemical models using the neurotoxin monosodium glutamate that induces lesions in the ventromedial hypothalamus nucleus, resulting in the development of obesity. However, the most widely used are the obesity models induced by high-fat diets. The aim of this review was to compile detail studies on the anti-obesity effects of natural compounds or their derivatives on rodent models of obesity as well as a critical analysis of the data.

1. Introduction

Obesity is defined as the excessive accumulation of fat in adipose tissue, resulting from an imbalance in consumption patterns, energy expenditure, and physical activity [1]. Despite the number of studies and available information to prevent or treat obesity, it has been considered a worldwide emerging health problem which continues to increase. The World Health Organization (WHO) estimates that there are more than 1.9 billion overweight adults, of which at least a third are obese [2]. The causes of this disease are complex and multifactorial. However, there is a genetic predisposition for obesity. Factors such as lifestyle, cultural and environmental influences, and epigenetics play a fundamental role in the development of obesity (Figure 1) [3,4,5]. People with excessive weight also tend to have a higher risk of developing other non-communicable diseases, such as type 2 diabetes, hypertension, osteoarthritis, cardiovascular, neurodegenerative, gastrointestinal, and chronic respiratory diseases, and certain types of cancer, contributing to a higher risk of premature death [6].
Nowadays, two possible types of obesity treatments are available: physiological and pharmacological. The first, involves diet and behavior changes, but fails more frequently if it is not strictly followed by the patients. Currently, pharmacological therapy includes the following synthetic drugs: phentermine, diethylpropion, fluoxetine, sibutramine, orlistat and rimonabant [7]. Plants have been used by humans since primitive times for food and medicine. Due to the presumably adverse effects of synthetic food additives on human health and the increased consumer perception of this problem, there is currently a growing interest in natural compounds extracted from human diets [8,9]. Nutraceuticals and functional foods have become popular in human diets since their effects on preventing nutrition-related diseases and improving physical and mental well-being have been reported [10,11]. Crude extracts or isolated pure natural compounds have the potential to reduce body weight and prevent diet-induced obesity [7]. The use of animal models of obesity with dietary intervention is useful to understand the role of certain nutrients and the anti-obesity mechanisms of natural compounds [12]. In this review, we will focus on different animal models of obesity used to evaluate the effects of natural products or their derivatives and summarize the main anti-obesity outcomes.

2. Rodent Models of Obesity Used in Natural Compounds Research

Similar to other areas of biomedical research, rodents (rats and especially mice) are widely used as preclinical animal models to study the underlying mechanisms of obesity and to evaluate new therapeutic approaches [13,14,15,16]. The anatomical and physiological similarities between humans and rodents allow for the screening of potential natural compounds with therapeutic properties before applying new compounds on humans [11,17]. There are several animal models of obesity, but not all are used to study the effects of natural compounds. Some of the available models are monogenic (animals with single-gene disorders) such as the db/db mice, ob/ob mice, Zucker fatty rat and Otsuka Long-Evans Tokushima fatty rat, others are chemically induced models using the neurotoxin monosodium glutamate (MSG) which induces lesions in the ventromedial hypothalamus (VMH) nucleus, and the most widely used are the obesity models induced by diets (Figure 2).

2.1. db/db Mice

The db/db mouse (db stands for diabetes) [18] can be used to study the molecular basis of obesity; however, it is commonly used for studies concerning type 2 diabetes [19]. These db/db mice were developed by investigators from the Jackson Laboratories in 1966 and are phenotypically similar to the ob/ob mouse model. They exhibit a mutation in leptin receptor gene, in an autosomal recessive trait that encodes for a G-to-T point mutation, leading to defective leptin signaling [19,20]. The predominant mouse strain in which this mutation is maintained is the C57BL/KS [14,19]. The db/db mice are characterized by hyperphagia, a consequence of impaired leptin signing in the hypothalamus, develop early-onset obesity due to low energy expenditure, are insulin resistant, have decreased insulin levels and are hypothermic. Moreover, they develop dyslipidemia, hypogonadism, hyperglycemia, have growth hormone (GH) deficiency, and are infertile [18,20]. Regarding the analysis of natural products on obesity development, this model can become an option, especially when the compounds that are tested, such as Artemisia extract (artemether) and barley (Table 1), have supposedly a double effect on both obesity and diabetes [21,22].

2.2. ob/ob Mice

Developed at Jackson Laboratories in 1966, the ob/ob mice are homozygous for the obese spontaneous mutation in the leptin gene that prevents the secretion of bioactive leptin resulting in leptin deficiency [1,14,18]. Leptin is a hormone derived from adipocytes, and its deficiency leads to an increase in appetite and the development of severe obesity [23,24,25]. The homozygous mutant mice are typically of the C57BL/6J strain. These mice present a pronounced obese phenotype characterized by uncontrollable food intake, type 2 diabetes, hyperphagia, hyperleptinemia, insulin resistance and fatty liver, hepatic steatosis, hypogonadism, and hypothyroidism [19,20]. The ob/ob mice are a better model for studying obesity than db/db mice because they have a longer lifetime and fewer clinical signs [26]. The effects of natural compounds such as celastrol, genistein, cannabinoid Δ9-tetrahydrocannabivarin and cinnamon extract on the ob/ob mouse model can be analyzed in Table 1 [27,28,29,30].

2.3. Zucker Fatty Rat

The Zucker fatty rat (ZFR), or Zucker (fa/fa), has a similar phenotype to ob/ob and db/db mice, as it has a homozygous missense mutation (fatty, fa) in the long form of the leptin receptor, which makes it insensitive to leptin [31,32]. Defects in the leptin receptor caused by this mutation result in the development of early-onset obesity due to hyperphagia, reducing the energy expenditure and leading to morbid obesity [14]. These rats are less likely to acquire diabetes, although ZFR has a high level of insulin resistance and fertility is reduced [14,20]
The ZFR is the most used model for studying several genetic characteristics associated with obesity [33,34], although it has also been used to study the anti-obesogenic effects of natural compounds such as the extracts of red wine [35], rosemary [36], green tea [37], bilberries, and purple potatoes [38] (Table 1).

2.4. Otsuka Long-Evans Tokushima Fatty Rat

The Otsuka Long-Evans Tokushima fatty (OLETF) rat is a spontaneous cholecystokinin type A (CCK-A) receptor knockout and does not respond to CCK, a peptide-derived hormone that functions as a peripheral satiety signal [20,39]. As a result, these rats are hyperphagic beginning several weeks after birth and develop mild obesity [34], which is a consequence of increased food intake and meal sizes, leading to an increase in body weight [40]. Moreover, OLETF rats develop diabetes, which manifests as polyuria and polydipsia [20]. At approximately 8 weeks of age, OLETF rats display hyperinsulinemia, and insulin resistance is observed at 12 weeks of age. Later, hyperplasia of pancreatic islets and hypertriglyceridemia develop [18].
The effects of gambigyeongsinhwan [41], gyeongshingangjeehwan [42], fermented mushroom milk-supplemented dietary fiber [43], or soy β-conglycinin [44] on obesity were evaluated in this animal model (Table 1).

2.5. Monosodium Glutamate (MSG)-Induced Obesity Model

The VMH nucleus, located in the hypothalamus, is associated with the regulation of eating behavior and satiety, with the use of rats with VMH lesions being one of the first models developed to induce obesity in rodents [14,20,45]. Bilateral lesions of the VMH nucleus lead to the development of hyperphagia, vagal hyperactivity, sympathetic hypoactivity, enlarged pancreatic islets, and hyperinsulinemia and can be caused by using the neurotoxin MSG [46,47]. MSG administration also causes lesions in the arcuate nucleus of the hypothalamus and circumventricular neurons. To induce obesity, MSG can be administered subcutaneously or intraperitoneally (2–4 mg/g of body weight) during the neonatal period, 4–10 times [48]. Adult rats who received MSG in the neonatal period developed endocrine dysfunction syndromes, which are characterized by obesity development, disturbances in the regulation of caloric balance, reduced growth, behavioral changes, and hypogonadism [47,49]. This obesity model was used to study the anti-obesogenic effects of Roselle (Hibiscus sabdariffa L.) [50] (Table 1).
Table 1. Anti-obesity effects of natural products in monogenic and neurotoxin monosodium glutamate (MSG)-induced obesity models.
Table 1. Anti-obesity effects of natural products in monogenic and neurotoxin monosodium glutamate (MSG)-induced obesity models.
Food Product/PlantBioactive CompoundsStrain/Obesity ModelDose and TreatmentObserved EffectsReferences
Artemisia extractArtemetherC57BL/KsJ db/db mice ♂200 mg/kg (oral gavage), for 2 weeks↓ Food intake and weight increase rate[21]
↓ Fasting blood glucose levels
↑ Tolerance to glucose
↑ Insulin sensitivity
↑ Insulin secretion
Improved hyperinsulinemia
Ameliorated islet vacuolar degeneration and hepatic steatosis
↓ Apoptosis of pancreatic beta cells
BarleyN.A.db/db mice (BKS.Cg-+Leprdb/+Leprdb/OlaHsd—fat, black, homozygous) ♂88% (w/w; mixed with the diet), for 8 weeks↓ Plasma insulin and resistin levels[22]
↓ TC levels in the liver
Bilberries (Vaccinium myrtillus)Nonacylated anthocyanin extractZucker (fa/fa) rats ♂, fed with HFD25 mg/kg/day (oral gavage), for 8 weeks↓ Fasting plasma glucose level[38]
↓ Levels of branched-chain amino acids
Improved lipid profiles
Cannabis sativaCannabinoid
Δ9-tetrahydrocannabivarin
C57BL/6 ob/ob mice ♀0.1, 0.5, 2.5 and 12.5 mg/kg/day (oral gavage), for 30 days↓ Liver TG concentration (only for 12.5 mg/kg)[27]
Cinnamon extract (Cinnamomum zeylanicum)N.A.B6.V-Lepob/J mice [on a C57BL/6J background (ob/ob)] ♂ 4.5 mL/kg (equates to 0.8 g/kg) (in drinking water), for 6 weeks↑ Insulin sensitivity and glucose tolerance[28]
↓ Hepatic levels of TG
↓ Fat accumulation in the liver
↑ Liver glycogen content
Improvement of insulin-stimulated locomotor activity
Celastraceae family members (including Tripterygium wilfordii)Celastrol (tripterine)C57BL/6J ob/ob mice ♂, fed with HFD3 mg/kg/day (mixed with the HFD), for 6 weeks↓ B.w.[30]
↓ Liver weight
↓ TG levels in the liver
↑ Glucose clearance
Downregulation of intestinal lipid transporters
↑ Lipid excretion in feces
Green teaPolyphenolsZucker (fa/fa) rats ♂, fed with HFD200 mg/kg/day (oral gavage), for 8 weeks↓ B.w. gain [37]
↓ Visceral fat
↓ Fasting serum insulin, glucose and lipids levels
Liriope platyphylla (dry roots)Aqueous extractOLETF rats5 or 10% (15 mL/g b.w./day; oral gavage), for 2 weeks↓ Abdominal fat mass [51]
↓ Glucose concentration
↑ Insulin production (only for 10% concentration)
↓ Expression of Glut-1
Mix of Curcuma longa L., Alnus japonica and Massa Medicata FermentataGambigyeongsinhwanOLETF rats ♂250 or 500 mg/kg/day (oral gavage), for 8 weeks ↓ B.w. gain[41]
↓ Adipose tissue weight and visceral adipocyte size
↑ mRNA levels PPARα in adipose tissue
Mix of edible mushrooms (Lentinus edodes, Ganoderma lucidum, Pleurotus ostreatus and Flammulina velutipes) in fermented milkN.A.OLETF rats ♂10 and 20% (v/w; mixed with the diet), for 6 weeks↓ B.w., [43]
↓ Perirenal fat, visceral and epididymal fat (only for 20% concentration),
↓TG and FFA levels
Mix of Liriope platyphylla, Platycodon grandiflorum, Schisandra chinensis, and Ephedra sinicaGyeongshingangjeehwanOLETF rats ♂121.7 mg/kg/day (oral gavage), for 8 weeks ↓ Visceral WAT weight [42]
↓ Size adipocytes in mesenteric WAT
↓ mRNA expression levels of adipocyte marker genes (PPARγ, aP2 and leptin) in visceral WAT
↑ mRNA expression levels of PPARα target genes in visceral WAT
↓ Plasma levels of FFA, TG, insulin and glucose
Purple Potato (Solanum tuberosum)Acylated anthocyanin extractZucker (fa/fa) rats ♂, fed with HFD25 mg/kg/day (oral gavage), for 8 weeks↓ Levels of branched-chain amino acids[38]
improved lipid profiles
↑ Glutamine/glutamate ratio
↓ Glycerol levels and metabolites involved in glycolysis
Red Wine (ProvinolsTM)Polyphenol extract (70% Polyphenols)Zucker (fa/fa) rats20 mg/kg/day (mixed with the diet), for 8 weeks↓ Plasma levels of glucose, fructosamine, TG, TC and LDL-cholesterol[35]
↑ NO
↑ eNOS activity
↓Superoxide anion
Roselle (Hibiscus sabdariffa L.) aqueous extractAnthocyanins Swiss Webster (CFW) mice ♂ induced by MSG120 mg/kg/day (60 mg/kg/day by oral gavage plus 60 mg/kg/day dissolved in tap water given ad libitum), for 60 days↓ B.w. gain[50]
↓ Glycemia
↑ ALT levels
Rosemary (Rosmarinus officinalis L.) extractCarnosic acid and carnosol Zucker (fa/fa) rats ♀0.5% (w/w; mixed with the diet), for 64 days↓ B.w. gain, [36]
↓ Serum TG, TC and insulin levels
Lipase activity inhibition in the stomach
Soy products, grains and legumesGenisteinob/ob mice ♀0.06% (w/w; mixed with the diet), for 4 weeks↓B.w. gain[25]
Downregulation of SOD activity
↑ iNOS expression in mesenteric artery perivascular adipose tissue
♂, male; ♀, female; ↓, decrease; ↑, increase; ALT, alanine aminotransferase; aP2, adipocyte fatty acid-binding protein; B.w., body weight; eNOS, endothelial nitric oxide-synthase; FFA, free fatty acids; Glut-1, glucose transporter 1; iNOS, inducible nitric oxide synthase; LDL, low density lipoprotein; MSG, monosodium glutamate; N.A., not applicable; NO, nitric oxide; PPAR-α, peroxisome proliferator-activated receptor alpha; PPARγ, peroxisome proliferator-activated receptor gamma; SOD, superoxide dismutase; TC, total cholesterol; TG, triglycerides; WAT, white adipose tissue; weeks, weeks.

2.6. Diet-Induced Obesity Models

Diet-induced obesity (DIO) rodent models are achieved by exposing rats and mice to specific diets that induce obesity, thereby reproducing dietary imbalances that are often the main cause of obesity in humans [13,20,52,53]. The main advantage of diets is that they can be standardized and control the nutrient percentages. There are a variety of diets that can be used for this purpose, such as high-fat diets (HFD), high-sugar diets (HSD), and high-sugar and high-fat (HSHF) diets, known as Western diets [16]. The HFD-induced obesity mouse model is one of the most used to understand the relationship between hyperlipidemic diets and the pathophysiology of obesity [52]. Within mouse strains, there are some more susceptible to DIO than others [13,54]; for example, the inbred C57BL/6 mouse strain is highly susceptible in contrast to SWR/J, A/J and CAST/Ei mouse strains, which tend to be resistant to these diets. In fact, the C57BL/6J mouse strain is the most used since when they are fed with HFD, these animals develop characteristics similar to humans with complex metabolic syndrome, such as obesity, high fat accumulation, insulin resistance, hyperglycemia, hyperlipidemia, hypertension, non-alcoholic fatty liver disease, and endothelium damage associated with cardiovascular diseases [18,20,52,55,56]. Some rat strains can also be used as models of HFD-induced obesity, such as the outbred Sprague Dawley, Wistar, or Long Evans rats [14]. However, Sprague Dawley rats showed diverse responses, with some animals showing DIO, while others showed resistance to the diet [13]. Detailed information on studies with natural compounds in HFD-induced obesity models is shown in Table 2.

3. Conclusions

Within the various models of obesity in rodents, HFD-induced obesity is one of the most used to study the potential benefits of compounds of natural origin in the obesity disease, as it best simulates the development of obesity in humans. Furthermore, depending on the stability of the compound, the administration of substances through their incorporation into the diet facilitates administration to animals while simulating the natural humans’ intake, the oral route. This is important because a practical way to ingest these natural compounds with anti-obesogenic properties can be through their incorporation into functional foods. There is no ideal rodent obesity model that can recap all the underlying mechanisms of obesity. Each model has advantages and disadvantages. Depending on the research objective, costs, and available model, researchers should select the option that best suits their needs. The translation and application of the results obtained from animal models of obesity, treated with potential therapeutic natural compounds, to humans are of great importance, as this can contribute to the resolution of this public health problem with theoretically minor adverse effects. However, this translation of knowledge has its limitations. For example, despite the physiological similarities, the metabolism between rodents and humans is different, which also translates into different effective doses. The same can also apply to the potentially toxic effects of a particular compound. Furthermore, it is difficult to mimic human disease in a given animal model, and doses and administration protocols are often not comparable. However, it is undeniable that the translation of knowledge from animal models to humans has been a very useful tool, allowing the testing of new pharmacological and therapeutic agents to respond to various human diseases.

Author Contributions

Conceptualization, T.M., T.F., E.N.-G., C.C.-R., S.L., E.R., L.M.A. and P.A.O.; writing—original draft preparation, T.M., T.F., E.N.-G., C.C.-R. and S.L.; writing—review and editing, T.M., T.F., E.N.-G., C.C.-R., S.L., L.M.A. and P.A.O.; supervision, L.M.A. and P.A.O.; funding acquisition, E.R. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the Portuguese Foundation for Science and Technology (FCT) and co-financed by the European Regional Development Fund (FEDER) through COMPETE 2020—Operational Competitiveness and Internationalization Programme (POCI), grant PTDC/ASP-HOR/29152/2017, POCI-01-0145-FEDER-029152 (VALORIZEBYPRODUCTS). This work was also supported by National Funds by FCT—Portuguese Foundation for Science and Technology, under the project UIDB/04033/2020. The authors acknowledge the financial support provided by the Portuguese Foundation for Science and Technology (FCT) through a Doctoral Grant (2020.04789.BD, Tiago Ferreira and BD/136747/2018, Elisabete Nascimento-Gonçalves).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Speakman, J.; Hambly, C.; Mitchell, S.; Król, E. The Contribution of Animal Models to the Study of Obesity. Lab. Anim. 2008, 42, 413–432. [Google Scholar] [CrossRef] [PubMed]
  2. WHO Overweight and Obesity—Children and Adolescents Aged 5–19. Available online: https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight (accessed on 18 January 2021).
  3. Novelli, E.L.B.; Diniz, Y.S.; Galhardi, C.M.; Ebaid, G.M.X.; Rodrigues, H.G.; Mani, F.; Fernandes, A.A.H.; Cicogna, A.C.; Novelli Filho, J.L.V.B. Anthropometrical Parameters and Markers of Obesity in Rats. Lab. Anim. 2007, 41, 111–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Kaila, B.; Raman, M. Obesity: A Review of Pathogenesis and Management Strategies. Can. J. Gastroenterol. 2008, 22, 61–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Mittwede, P.N.; Clemmer, J.S.; Bergin, P.F.; Xiang, L. Obesity and Critical Illness: Insights from Animal Models. Shock 2016, 45, 349–358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Lau, D.C.W.; Douketis, J.D.; Morrison, K.M.; Hramiak, I.M.; Sharma, A.M.; Ur, E.; for members of the Obesity Canada Clinical Practice Guidelines Expert Panel. 2006 Canadian Clinical Practice Guidelines on the Management and Prevention of Obesity in Adults and Children [Summary]. Can. Med. Assoc. J. 2007, 176, S1–S13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Mohamed, G.A.; Ibrahim, S.R.M.; Elkhayat, E.S.; El Dine, R.S. Natural Anti-Obesity Agents. Bull. Fac. Pharm. Cairo Univ. 2014, 52, 269–284. [Google Scholar] [CrossRef] [Green Version]
  8. Zhong, Y.; Wu, L.; Chen, X.; Huang, Z.; Hu, W. Effects of Food-Additive-Information on Consumers’ Willingness to Accept Food with Additives. Int. J. Environ. Res. Public Health 2018, 15, 2394. [Google Scholar] [CrossRef] [Green Version]
  9. Lourenço, S.C.; Moldão-Martins, M.; Alves, V.D. Antioxidants of Natural Plant Origins: From Sources to Food Industry Applications. Molecules 2019, 24, 4132. [Google Scholar] [CrossRef] [Green Version]
  10. Rodríguez-Pérez, C.; Segura-Carretero, A.; del Mar Contreras, M. Phenolic Compounds as Natural and Multifunctional Anti-Obesity Agents: A Review. Crit. Rev. Food Sci. Nutr. 2019, 59, 1212–1229. [Google Scholar] [CrossRef]
  11. Ferreira, T.; Nascimento-Gonçalves, E.; Gil Da Costa, R.M.; Rosa, E.; Alexandra Oliveira, P. Therapeutic and Toxicological Effects of Natural Compounds: Data from HPV16-Transgenic and ICR Mice (Review). World Acad. Sci. J. 2020, 2, 9. [Google Scholar] [CrossRef]
  12. Chalvon-Demersay, T.; Blachier, F.; Tomé, D.; Blais, A. Animal Models for the Study of the Relationships between Diet and Obesity: A Focus on Dietary Protein and Estrogen Deficiency. Front. Nutr. 2017, 4, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Speakman, J.; Hambly, C.; Mitchell, S.; Król, E. Animal Models of Obesity. Obes. Rev. 2007, 8, 55–61. [Google Scholar] [CrossRef] [PubMed]
  14. Lutz, T.A.; Woods, S.C. Overview of Animal Models of Obesity. Curr. Protoc. Pharmacol. 2012, 58, 5.61.1–5.61.18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Barrett, P.; Mercer, J.G.; Morgan, P.J. Preclinical Models for Obesity Research. Dis. Model. Mech. 2016, 9, 1245–1255. [Google Scholar] [CrossRef] [Green Version]
  16. Preguiça, I.; Alves, A.; Nunes, S.; Fernandes, R.; Gomes, P.; Viana, S.D.; Reis, F. Diet-induced Rodent Models of Obesity-related Metabolic Disorders—A Guide to a Translational Perspective. Obes. Rev. 2020, 21, e13081. [Google Scholar] [CrossRef]
  17. Bryda, E.C. The Mighty Mouse: The Impact of Rodents on Advances in Biomedical Research. Mo. Med. 2013, 110, 207–211. [Google Scholar]
  18. Kanasaki, K.; Koya, D. Biology of Obesity: Lessons from Animal Models of Obesity. J. Biomed. Biotechnol. 2011, 2011, 197636. [Google Scholar] [CrossRef] [Green Version]
  19. Suleiman, J.B.; Mohamed, M.; Bakar, A.B.A. A Systematic Review on Different Models of Inducing Obesity in Animals: Advantages and Limitations. J. Adv. Vet. Anim. Res. 2020, 7, 103–114. [Google Scholar] [CrossRef]
  20. Kleinert, M.; Clemmensen, C.; Hofmann, S.M.; Moore, M.C.; Renner, S.; Woods, S.C.; Huypens, P.; Beckers, J.; de Angelis, M.H.; Schürmann, A.; et al. Animal Models of Obesity and Diabetes Mellitus. Nat. Rev. Endocrinol. 2018, 14, 140–162. [Google Scholar] [CrossRef] [Green Version]
  21. Guo, Y.; Fu, W.; Xin, Y.; Bai, J.; Peng, H.; Fu, L.; Liu, J.; Li, L.; Ma, Y.; Jiang, H. Antidiabetic and Antiobesity Effects of Artemether in Db/Db Mice. BioMed Res. Int. 2018, 2018, 8639523. [Google Scholar] [CrossRef]
  22. Garcia-Mazcorro, J.F.; Mills, D.A.; Murphy, K.; Noratto, G. Effect of Barley Supplementation on the Fecal Microbiota, Caecal Biochemistry, and Key Biomarkers of Obesity and Inflammation in Obese Db/Db Mice. Eur. J. Nutr. 2018, 57, 2513–2528. [Google Scholar] [CrossRef] [PubMed]
  23. Crujeiras, A.B.; Carreira, M.C.; Cabia, B.; Andrade, S.; Amil, M.; Casanueva, F.F. Leptin Resistance in Obesity: An Epigenetic Landscape. Life Sci. 2015, 140, 57–63. [Google Scholar] [CrossRef] [PubMed]
  24. Liu, J.; Lee, J.; Andres, M.; Hernandez, S.; Mazitschek, R.; Ozcan, U.; Hospital, G. Treatment of Obesity with Celastrol. Treatment 2016, 161, 999–1011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Smith, J.T.; Acohido, B.V.; Clifton, D.K.; Steiner, R.A. KiSS-1 Neurones Are Direct Targets for Leptin in the Ob/Ob Mouse. J. Neuroendocrinol. 2006, 18, 298–303. [Google Scholar] [CrossRef]
  26. Ritskes-Hoitinga, M.; Tobin, G.; Jensen, T.L.; Mikkelsen, L.F. Nutrition of the Laboratory Mouse. In The Laboratory Mouse; Elsevier: Amsterdam, The Netherlands, 2012; pp. 567–599. ISBN 978-0-12-382008-2. [Google Scholar]
  27. Wargent, E.T.; Zaibi, M.S.; Silvestri, C.; Hislop, D.C.; Stocker, C.J.; Stott, C.G.; Guy, G.W.; Duncan, M.; Di Marzo, V.; Cawthorne, M.A. The Cannabinoid Δ9-Tetrahydrocannabivarin (THCV) Ameliorates Insulin Sensitivity in Two Mouse Models of Obesity. Nutr. Diabetes 2013, 3, e68. [Google Scholar] [CrossRef] [PubMed]
  28. Sartorius, T.; Peter, A.; Schulz, N.; Drescher, A.; Bergheim, I.; Machann, J.; Schick, F.; Siegel-Axel, D.; Schürmann, A.; Weigert, C.; et al. Cinnamon Extract Improves Insulin Sensitivity in the Brain and Lowers Liver Fat in Mouse Models of Obesity. PLoS ONE 2014, 9, e92358. [Google Scholar] [CrossRef] [Green Version]
  29. Simperova, A.; Al-Nakkash, L.; Faust, J.J.; Sweazea, K.L. Genistein Supplementation Prevents Weight Gain but Promotes Oxidative Stress and Inflammation in the Vasculature of Female Obese ob/ob Mice. Nutr. Res. 2016, 36, 789–797. [Google Scholar] [CrossRef]
  30. Hua, H.; Zhang, Y.; Zhao, F.; Chen, K.; Wu, T.; Liu, Q.; Huang, S.; Zhang, A.; Jia, Z. Celastrol Inhibits Intestinal Lipid Absorption by Reprofiling the Gut Microbiota to Attenuate High-Fat Diet-Induced Obesity. iScience 2021, 24, 102077. [Google Scholar] [CrossRef]
  31. van der Spek, R.; Kreier, F.; Fliers, E.; Kalsbeek, A. Circadian Rhythms in White Adipose Tissue. In Progress in Brain Research; Elsevier: Amsterdam, The Netherlands, 2012; Volume 199, pp. 183–201. ISBN 978-0-444-59427-3. [Google Scholar]
  32. Kitada, M.; Ogura, Y.; Koya, D. Rodent Models of Diabetic Nephropathy: Their Utility and Limitations. Int. J. Nephrol. Renov. Dis. 2016, 9, 279–290. [Google Scholar] [CrossRef] [Green Version]
  33. Yorek, M.A. Alternatives to the Streptozotocin-Diabetic Rodent, 1st ed.; Elsevier Inc.: Amsterdam, The Netherlands, 2016; Volume 127, ISBN 9780128039151. [Google Scholar]
  34. Owens, D.R. Spontaneous, Surgically and Chemically Induced Models of Disease. In The laboratory Rat; Academic Press: Cambridge, MA, USA, 2006; pp. 711–732. [Google Scholar] [CrossRef]
  35. Agouni, A.; Lagrue-Lak-Hal, A.-H.; Mostefai, H.A.; Tesse, A.; Mulder, P.; Rouet, P.; Desmoulin, F.; Heymes, C.; Martínez, M.C.; Andriantsitohaina, R. Red Wine Polyphenols Prevent Metabolic and Cardiovascular Alterations Associated with Obesity in Zucker Fatty Rats (Fa/Fa). PLoS ONE 2009, 4, e5557. [Google Scholar] [CrossRef] [Green Version]
  36. Romo Vaquero, M.; Yáñez-Gascón, M.-J.; García Villalba, R.; Larrosa, M.; Fromentin, E.; Ibarra, A.; Roller, M.; Tomás-Barberán, F.; Espín de Gea, J.C.; García-Conesa, M.-T. Inhibition of Gastric Lipase as a Mechanism for Body Weight and Plasma Lipids Reduction in Zucker Rats Fed a Rosemary Extract Rich in Carnosic Acid. PLoS ONE 2012, 7, e39773. [Google Scholar] [CrossRef] [Green Version]
  37. Tan, Y.; Kim, J.; Cheng, J.; Ong, M.; Lao, W.-G.; Jin, X.-L.; Lin, Y.-G.; Xiao, L.; Zhu, X.-Q.; Qu, X.-Q. Green Tea Polyphenols Ameliorate Non-Alcoholic Fatty Liver Disease through Upregulating AMPK Activation in High Fat Fed Zucker Fatty Rats. World J. Gastroenterol. 2017, 23, 3805. [Google Scholar] [CrossRef] [PubMed]
  38. Chen, K.; Wei, X.; Zhang, J.; Pariyani, R.; Jokioja, J.; Kortesniemi, M.; Linderborg, K.M.; Heinonen, J.; Sainio, T.; Zhang, Y.; et al. Effects of Anthocyanin Extracts from Bilberry (Vaccinium Myrtillus L.) and Purple Potato (Solanum Tuberosum L. Var. ‘Synkeä Sakari’) on the Plasma Metabolomic Profile of Zucker Diabetic Fatty Rats. J. Agric. Food Chem. 2020, 68, 9436–9450. [Google Scholar] [CrossRef] [PubMed]
  39. Bi, S.; Moran, T.H. Actions of CCK in the Controls of Food Intake and Body Weight: Lessons from the CCK-A Receptor Deficient OLETF Rat. Neuropeptides 2002, 36, 171–181. [Google Scholar] [CrossRef] [PubMed]
  40. Schwarzer, M. Models to Investigate Cardiac Metabolism Michael. In The Scientist’s Guide to Cardiac Metabolism; Elsevier Inc.: Amsterdam, The Netherlands, 2016; Volume 45, p. 114. ISBN 9780128023945. [Google Scholar]
  41. Sung Roh, J.; Lee, H.; Woo, S.; Yoon, M.; Kim, J.; Dong Park, S.; Shik Shin, S.; Yoon, M. Herbal Composition Gambigyeongsinhwan (4) from Curcuma Longa, Alnus Japonica, and Massa Medicata Fermentata Inhibits Lipid Accumulation in 3T3-L1 Cells and Regulates Obesity in Otsuka Long-Evans Tokushima Fatty Rats. J. Ethnopharmacol. 2015, 171, 287–294. [Google Scholar] [CrossRef] [PubMed]
  42. Shin, S.S.; Jung, Y.S.; Yoon, K.H.; Choi, S.; Hong, Y.; Park, D.; Lee, H.; Seo, B., II; Lee, H.Y.; Yoon, M. The Korean Traditional Medicine Gyeongshingangjeehwan Inhibits Adipocyte Hypertrophy and Visceral Adipose Tissue Accumulation by Activating PPARα Actions in Rat White Adipose Tissues. J. Ethnopharmacol. 2010, 127, 47–54. [Google Scholar] [CrossRef]
  43. Jeon, B.S.; Park, J.W.; Kim, B.K.; Kim, H.K.; Jung, T.S.; Hahm, J.R.; Kim, D.R.; Cho, Y.S.; Cha, J.Y. Fermented Mushroom Milk-Supplemented Dietary Fibre Prevents the Onset of Obesity and Hypertriglyceridaemia in Otsuka Long-Evans Tokushima Fatty Rats. Diabetes Obes. Metab. 2005, 7, 709–715. [Google Scholar] [CrossRef]
  44. Wanezaki, S.; Tachibana, N.; Nagata, M.; Saito, S.; Nagao, K.; Yanagita, T.; Kohno, M. Soy β-Conglycinin Improves Obesity-Induced Metabolic Abnormalities in a Rat Model of Nonalcoholic Fatty Liver Disease. Obes. Res. Clin. Pract. 2015, 9, 168–174. [Google Scholar] [CrossRef]
  45. Sun, H.; Zhao, P.; Liu, W.; Li, L.; Ai, H.; Ma, X. Ventromedial Hypothalamic Nucleus in Regulation of Stress-Induced Gastric Mucosal Injury in Rats. Sci. Rep. 2018, 8, 10170. [Google Scholar] [CrossRef]
  46. Kiba, T.; Tanaka, K.; Numata, K.; Hoshino, M.; Misugi, K.; Inoue, S. Ventromedial Hypothalamic Lesion-Induced Vagal Hyperactivity Stimulates Rat Pancreatic Cell Proliferation. Gastroenterology 1996, 110, 885–893. [Google Scholar] [CrossRef]
  47. Parasuraman, S.; Wen, L.E. Animal Model for Obesity-An Overview. Syst. Rev. Pharm. 2016, 6, 9–12. [Google Scholar] [CrossRef]
  48. Von Diemen, V.; Trindade, E.N.; Trindade, M.R.M. Experimental Model to Induce Obesity in Rats. Acta Cir. Bras. 2006, 21, 425–429. [Google Scholar] [CrossRef] [PubMed]
  49. Miśkowiak, B.; Partyka, M. Effects of Neonatal Treatment with MSG (Monosodium Glutamate) on Hypothalamo-Pituitary-Thyroid Axis in Adult Male Rats. Histol. Histopathol. 1993, 8, 731–734. [Google Scholar] [PubMed]
  50. Alarcon-Aguilar, F.J.; Zamilpa, A.; Perez-Garcia, M.D.; Almanza-Perez, J.C.; Romero-Nuñez, E.; Campos-Sepulveda, E.A.; Vazquez-Carrillo, L.I.; Roman-Ramos, R. Effect of Hibiscus Sabdariffa on Obesity in MSG Mice. J. Ethnopharmacol. 2007, 114, 66–71. [Google Scholar] [CrossRef]
  51. Kim, J.-E.; Hwang, I.-S.; Choi, S.-I.; Hye-Ryun, L.; Young-Ju, L.; Jun-Seo, G.; Hee-Seob, L.; Son, H.-J.; Jang, M.-J.; Lee, S.-H.; et al. Aqueous Extract of Liriope Platyphylla, a Traditional Chinese Medicine, Significantly Inhibits Abdominal Fat Accumulation and Improves Glucose Regulation in OLETF Type II Diabetes Model Rats. Lab Anim. Res 2012, 28, 181–191. [Google Scholar] [CrossRef] [Green Version]
  52. Hernández-Granados, M.J.; Ramírez-Emiliano, J.; Franco-Robles, E. Rodent Models of Obesity and Diabetes. In Experimental Animal Models of Human Diseases—An Effective Therapeutic Strategy; Bartholomew, I., Ed.; InTech: London, UK, 2018; ISBN 978-1-78923-164-9. [Google Scholar]
  53. Geiger, B.M.; Pothos, E.N. Translating Animal Models of Obesity and Diabetes to the Clinic. In Handbook of Behavioral Neuroscience; Elsevier: Amsterdam, The Netherlands, 2019; Volume 29, pp. 1–16. ISBN 978-0-12-803161-2. [Google Scholar]
  54. Panchal, S.K.; Brown, L. Rodent Models for Metabolic Syndrome Research. J. Biomed. Biotechnol. 2011, 2011, 1–14. [Google Scholar] [CrossRef] [Green Version]
  55. Yang, Y.; Smith, D.L.; Keating, K.D.; Allison, D.B.; Nagy, T.R. Variations in Body Weight, Food Intake and Body Composition after Long-Term High-Fat Diet Feeding in C57BL/6J Mice: Variations in Diet-Induced Obese C57BL/6J Mice. Obesity 2014, 22, 2147–2155. [Google Scholar] [CrossRef]
  56. Glastras, S.J.; Chen, H.; Teh, R.; McGrath, R.T.; Chen, J.; Pollock, C.A.; Wong, M.G.; Saad, S. Mouse Models of Diabetes, Obesity and Related Kidney Disease. PLoS ONE 2016, 11, e0162131. [Google Scholar] [CrossRef]
  57. Eom, J.; Thomas, S.S.; Sung, N.-Y.; Kim, D.-S.; Cha, Y.-S.; Kim, K.-A. Abeliophyllum Distichum Ameliorates High-Fat Diet-Induced Obesity in C57BL/6J Mice by Upregulating the AMPK Pathway. Nutrients 2020, 12, 3320. [Google Scholar] [CrossRef]
  58. Cha, Y.-S.; Rhee, S.-J.; Heo, Y.-R. Acanthopanax Senticosus Extract Prepared from Cultured Cells Decreases Adiposity and Obesity Indices in C57BL/6J Mice Fed a High Fat Diet. J. Med. Food 2004, 7, 422–429. [Google Scholar] [CrossRef]
  59. Yoshizumi, K.; Hirano, K.; Ando, H.; Hirai, Y.; Ida, Y.; Tsuji, T.; Tanaka, T.; Satouchi, K.; Terao, J. Lupane-Type Saponins from Leaves of Acanthopanax Sessiliflorus and Their Inhibitory Activity on Pancreatic Lipase. J. Agric. Food Chem. 2006, 54, 335–341. [Google Scholar] [CrossRef] [PubMed]
  60. Liu, W.; Zheng, Y.; Han, L.; Wang, H.; Saito, M.; Ling, M.; Kimura, Y.; Feng, Y. Saponins (Ginsenosides) from Stems and Leaves of Panax Quinquefolium Prevented High-Fat Diet-Induced Obesity in Mice. Phytomedicine 2008, 15, 1140–1145. [Google Scholar] [CrossRef] [PubMed]
  61. Park, Y.S. Platycodon Grandiflorum> Extract Represses Up-Regulated Adipocyte Fatty Acid Binding Protein Triggered by a High Fat Feeding in Obese Rats. World J. Gastroenterol. 2007, 13, 3493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Ae Yoon, N.; Park, J.; Yeon Jeong, J.; Rashidova, N.; Ryu, J.; Seob Roh, G.; Joon Kim, H.; Jae Cho, G.; Sung Choi, W.; Hoon Lee, D.; et al. Anti-Obesity Activity of Ethanol Extract from Bitter Melon in Mice Fed High-Fat Diet. Dev. Reprod. 2019, 23, 129–138. [Google Scholar] [CrossRef] [Green Version]
  63. Benn, T.; Kim, B.; Park, Y.-K.; Wegner, C.J.; Harness, E.; Nam, T.-G.; Kim, D.-O.; Lee, J.S.; Lee, J.-Y. Polyphenol-Rich Blackcurrant Extract Prevents Inflammation in Diet-Induced Obese Mice. J. Nutr. Biochem. 2014, 25, 1019–1025. [Google Scholar] [CrossRef]
  64. Uchiyama, S.; Taniguchi, Y.; Saka, A.; Yoshida, A.; Yajima, H. Prevention of Diet-Induced Obesity by Dietary Black Tea Polyphenols Extract in Vitro and in Vivo. Nutrition 2011, 27, 287–292. [Google Scholar] [CrossRef]
  65. Ikarashi, N.; Toda, T.; Okaniwa, T.; Ito, K.; Ochiai, W.; Sugiyama, K. Anti-Obesity and Anti-Diabetic Effects of Acacia Polyphenol in Obese Diabetic KKAy Mice Fed High-Fat Diet. Evid. Based Complement. Alternat. Med. 2011, 2011, 1–10. [Google Scholar] [CrossRef] [Green Version]
  66. Aborehab, N.M.; El Bishbishy, M.H.; Waly, N.E. Resistin Mediates Tomato and Broccoli Extract Effects on Glucose Homeostasis in High Fat Diet-Induced Obesity in Rats. BMC Complement. Altern. Med. 2016, 16, 225. [Google Scholar] [CrossRef] [Green Version]
  67. Zandani, G.; Anavi-Cohen, S.; Tsybina-Shimshilashvili, N.; Sela, N.; Nyska, A.; Madar, Z. Broccoli Florets Supplementation Improves Insulin Sensitivity and Alters Gut Microbiome Population—A Steatosis Mice Model Induced by High-Fat Diet. Front. Nutr. 2021, 8, 680241. [Google Scholar] [CrossRef]
  68. Li, X.; Tian, S.; Wang, Y.; Liu, J.; Wang, J.; Lu, Y. Broccoli Microgreens Juice Reduces Body Weight by Enhancing Insulin Sensitivity and Modulating Gut Microbiota in High-Fat Diet-Induced C57BL/6J Obese Mice. Eur. J. Nutr. 2021, 60, 3829–3839. [Google Scholar] [CrossRef]
  69. Nagata, N.; Xu, L.; Kohno, S.; Ushida, Y.; Aoki, Y.; Umeda, R.; Fuke, N.; Zhuge, F.; Ni, Y.; Nagashimada, M.; et al. Glucoraphanin Ameliorates Obesity and Insulin Resistance Through Adipose Tissue Browning and Reduction of Metabolic Endotoxemia in Mice. Diabetes 2017, 66, 1222–1236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Kang, M.-C.; Lee, H.-G.; Kim, H.-S.; Song, K.-M.; Chun, Y.-G.; Lee, M.H.; Kim, B.-K.; Jeon, Y.-J. Anti-Obesity Effects of Sargassum Thunbergii via Downregulation of Adipogenesis Gene and Upregulation of Thermogenic Genes in High-Fat Diet-Induced Obese Mice. Nutrients 2020, 12, 3325. [Google Scholar] [CrossRef] [PubMed]
  71. Kim, I.; Choi, J.; Lee, M.; Kwon, C.; Nam, T. Anti-Obesity Effects of Pectinase and Cellulase Enzyme-treated Eckloniaïcava Extract in High-fat Diet-fed C57BL/6N Mice. Int. J. Mol. Med. 2017, 41, 924–934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Maeda, H.; Hosokawa, M.; Sashima, T.; Murakami-Funayama, K.; Miyashita, K. Anti-obesity and anti-diabetic effects of fucoxanthin on diet-induced obesity conditions in a murine model. Mol. Med. Rep. 2009, 2, 897–902. [Google Scholar] [CrossRef]
  73. Oh, J.-H.; Kim, J.; Lee, Y. Anti-Inflammatory and Anti-Diabetic Effects of Brown Seaweeds in High-Fat Diet-Induced Obese Mice. Nutr. Res. Pract. 2016, 10, 42. [Google Scholar] [CrossRef] [Green Version]
  74. Lee, M.; Hou, J.G.; Begum, S.; Wang, Y.B.; Oh, D.S.; Wi, A.J.; Yoon, B.-S.; Sung, C.-G. Anti-Obesity Effects of Sparassis Crispa on High-Fat Diet-Induced Obese Mice. J. Life Sci. 2014, 24, 952–958. [Google Scholar] [CrossRef] [Green Version]
  75. Han, L.-K.; Sumiyoshi, M.; Zhang, J.; Liu, M.-X.; Zhang, X.-F.; Zheng, Y.-N.; Okuda, H.; Kimura, Y. Anti-Obesity Action Of Salix Matsudana Leaves (Part 1). Anti-Obesity Action by Polyphenols of Salix Matsudana in High Fat-Diet Treated Rodent Animals. Phytother. Res. 2003, 17, 1188–1194. [Google Scholar] [CrossRef]
  76. Choi, E.H.; Yang, H.P.; Chun, H.S. Chitooligosaccharide Ameliorates Diet-Induced Obesity in Mice and Affects Adipose Gene Expression Involved in Adipogenesis and Inflammation. Nutr. Res. 2012, 32, 218–228. [Google Scholar] [CrossRef]
  77. Pan, H.; Yang, Q.; Huang, G.; Ding, C.; Cao, P.; Huang, L.; Xiao, T.; Guo, J.; Su, Z. Hypolipidemic Effects of Chitosan and Its Derivatives in Hyperlipidemic Rats Induced by a High-Fat Diet. Food Nutr. Res. 2016, 60, 31137. [Google Scholar] [CrossRef] [Green Version]
  78. Tao, Y.; Zhang, H.-L.; Hu, Y.-M.; Wan, S.; Su, Z.-Q. Preparation of Chitosan and Water-Soluble Chitosan Microspheres via Spray-Drying Method to Lower Blood Lipids in Rats Fed with High-Fat Diets. Int. J. Mol. Sci. 2013, 14, 4174–4184. [Google Scholar] [CrossRef]
  79. de Melo, C.L.; Queiroz, M.G.R.; Arruda Filho, A.C.V.; Rodrigues, A.M.; de Sousa, D.F.; Almeida, J.G.L.; Pessoa, O.D.L.; Silveira, E.R.; Menezes, D.B.; Melo, T.S.; et al. Betulinic Acid, a Natural Pentacyclic Triterpenoid, Prevents Abdominal Fat Accumulation in Mice Fed a High-Fat Diet. J. Agric. Food Chem. 2009, 57, 8776–8781. [Google Scholar] [CrossRef] [PubMed]
  80. Min, S.Y.; Yang, H.; Seo, S.G.; Shin, S.H.; Chung, M.-Y.; Kim, J.; Lee, S.J.; Lee, H.J.; Lee, K.W. Cocoa Polyphenols Suppress Adipogenesis In Vitro and Obesity In Vivo by Targeting Insulin Receptor. Int. J. Obes. 2013, 37, 584–592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Zhu, Z.; Jiang, W.; Thompson, H.J. Edible Dry Bean Consumption (Phaseolus Vulgaris L.) Modulates Cardiovascular Risk Factors and Diet-Induced Obesity in Rats and Mice. Br. J. Nutr. 2012, 108, S66–S73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Jayaprakasam, B.; Olson, L.K.; Schutzki, R.E.; Tai, M.-H.; Nair, M.G. Amelioration of Obesity and Glucose Intolerance in High-Fat-Fed C57BL/6 Mice by Anthocyanins and Ursolic Acid in Cornelian Cherry (Cornus Mas). J. Agric. Food Chem. 2006, 54, 243–248. [Google Scholar] [CrossRef]
  83. Jo, Y.; Choi, K.-M.; Liu, Q.; Kim, S.; Ji, H.-J.; Kim, M.; Shin, S.-K.; Do, S.-G.; Shin, E.; Jung, G.; et al. Anti-Obesity Effect of 6,8-Diprenylgenistein, an Isoflavonoid of Cudrania Tricuspidata Fruits in High-Fat Diet-Induced Obese Mice. Nutrients 2015, 7, 10480–10490. [Google Scholar] [CrossRef]
  84. Asai, A.; Miyazawa, T. Dietary Curcuminoids Prevent High-Fat Diet–Induced Lipid Accumulation in Rat Liver and Epididymal Adipose Tissue. J. Nutr. 2001, 131, 2932–2935. [Google Scholar] [CrossRef] [Green Version]
  85. Ejaz, A.; Wu, D.; Kwan, P.; Meydani, M. Curcumin Inhibits Adipogenesis in 3T3-L1 Adipocytes and Angiogenesis and Obesity in C57/BL Mice. J. Nutr. 2009, 139, 919–925. [Google Scholar] [CrossRef]
  86. Kwon, C.-S.; Sohn, H.Y.; Kim, S.H.; Kim, J.H.; Son, K.H.; Lee, J.S.; Lim, J.K.; Kim, J.-S. Anti-Obesity Effect of Dioscorea Nipponica Makino with Lipase-Inhibitory Activity in Rodents. Biosci. Biotechnol. Biochem. 2003, 67, 1451–1456. [Google Scholar] [CrossRef] [Green Version]
  87. Jeong, E.J.; Jegal, J.; Ahn, J.; Kim, J.; Yang, M.H. Anti-Obesity Effect of Dioscorea Oppositifolia Extract in High-Fat Diet-Induced Obese Mice and Its Chemical Characterization. Biol. Pharm. Bull. 2016, 39, 409–414. [Google Scholar] [CrossRef] [Green Version]
  88. Belguith-Hadriche, O.; Ammar, S.; del Mar Contreras, M.; Turki, M.; Segura-Carretero, A.; El Feki, A.; Makni-Ayedi, F.; Bouaziz, M. Antihyperlipidemic and Antioxidant Activities of Edible Tunisian Ficus carica L. Fruits in High Fat Diet-Induced Hyperlipidemic Rats. Plant Foods Hum. Nutr. 2016, 71, 183–189. [Google Scholar] [CrossRef]
  89. Ibarra, A.; Bai, N.; He, K.; Bily, A.; Cases, J.; Roller, M.; Sang, S. Fraxinus Excelsior Seed Extract FraxiPureTM Limits Weight Gains and Hyperglycemia in High-Fat Diet-Induced Obese Mice. Phytomedicine 2011, 18, 479–485. [Google Scholar] [CrossRef] [PubMed]
  90. Xia, D.-Z.; Yu, X.-F.; Wang, H.-M.; Ren, Q.-Y.; Chen, B.-M. Anti-Obesity and Hypolipidemic Effects of Ethanolic Extract from Alpinia Officinarum Hance (Zingiberaceae) in Rats Fed High-Fat Diet. J. Med. Food 2010, 13, 785–791. [Google Scholar] [CrossRef] [PubMed]
  91. Kim, I.; Kim, H.-R.; Kim, J.-H.; Om, A.-S. Beneficial Effects of Allium sativum L. Stem Extract on Lipid Metabolism and Antioxidant Status in Obese Mice Fed a High-Fat Diet: Beneficial Effects of Allium sativum L. Stem Extract in Obese Mice. J. Sci. Food Agric. 2013, 93, 2749–2757. [Google Scholar] [CrossRef] [PubMed]
  92. Wang, J.; Li, D.; Wang, P.; Hu, X.; Chen, F. Ginger Prevents Obesity through Regulation of Energy Metabolism and Activation of Browning in High-Fat Diet-Induced Obese Mice. J. Nutr. Biochem. 2019, 70, 105–115. [Google Scholar] [CrossRef]
  93. Karu, N.; Reifen, R.; Kerem, Z. Weight Gain Reduction in Mice Fed Panax Ginseng Saponin, a Pancreatic Lipase Inhibitor. J. Agric. Food Chem. 2007, 55, 2824–2828. [Google Scholar] [CrossRef]
  94. Li, X.; Luo, J.; Anandh Babu, P.V.; Zhang, W.; Gilbert, E.; Cline, M.; McMillan, R.; Hulver, M.; Alkhalidy, H.; Zhen, W.; et al. Dietary Supplementation of Chinese Ginseng Prevents Obesity and Metabolic Syndrome in High-Fat Diet-Fed Mice. J. Med. Food 2014, 17, 1287–1297. [Google Scholar] [CrossRef] [Green Version]
  95. Sheng, Y.; Zhao, C.; Zheng, S.; Mei, X.; Huang, K.; Wang, G.; He, X. Anti-obesity and Hypolipidemic Effect of Water Extract from Pleurotus Citrinopileatus in C57 BL /6J Mice. Food Sci. Nutr. 2019, 7, 1295–1301. [Google Scholar] [CrossRef] [Green Version]
  96. Hogan, S.; Canning, C.; Sun, S.; Sun, X.; Kadouh, H.; Zhou, K. Dietary Supplementation of Grape Skin Extract Improves Glycemia and Inflammation in Diet-Induced Obese Mice Fed a Western High Fat Diet. J. Agric. Food Chem. 2011, 59, 3035–3041. [Google Scholar] [CrossRef]
  97. Sharma, B.R.; Kim, H.J.; Kim, M.S.; Park, C.M.; Rhyu, D.Y. Caulerpa Okamurae Extract Inhibits Adipogenesis in 3T3-L1 Adipocytes and Prevents High-Fat Diet–Induced Obesity in C57BL/6 Mice. Nutr. Res. 2017, 47, 44–52. [Google Scholar] [CrossRef]
  98. Kim, J.; Choi, J.H.; Oh, T.; Ahn, B.; Unno, T. Codium Fragile Ameliorates High-Fat Diet-Induced Metabolism by Modulating the Gut Microbiota in Mice. Nutrients 2020, 12, 1848. [Google Scholar] [CrossRef]
  99. Kim, H.-J.; Hong, S.-H.; Chang, S.-H.; Kim, S.; Lee, A.Y.; Jang, Y.; Davaadamdin, O.; Yu, K.-N.; Kim, J.-E.; Cho, M.-H. Effects of Feeding a Diet Containing Gymnema Sylvestre Extract: Attenuating Progression of Obesity in C57BL/6J Mice. Asian Pac. J. Trop. Med. 2016, 9, 437–444. [Google Scholar] [CrossRef] [PubMed]
  100. Zar Kalai, F.; Han, J.; Ksouri, R.; El Omri, A.; Abdelly, C.; Isoda, H. Antiobesity Effects of an Edible Halophyte Nitraria Retusa Forssk in 3T3-L1 Preadipocyte Differentiation and in C57B6J/L Mice Fed a High Fat Diet-Induced Obesity. Evid. Based Complement. Alternat. Med. 2013, 2013, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Ono, Y.; Hattori, E.; Fukaya, Y.; Imai, S.; Ohizumi, Y. Anti-Obesity Effect of Nelumbo Nucifera Leaves Extract in Mice and Rats. J. Ethnopharmacol. 2006, 106, 238–244. [Google Scholar] [CrossRef] [PubMed]
  102. Song, J.; Kim, J.; Park, H.J.; Kim, H. Anti-Obesity Effects of a Prunus Persica and Nelumbo Nucifera Mixture in Mice Fed a High-Fat Diet. Nutrients 2020, 12, 3392. [Google Scholar] [CrossRef] [PubMed]
  103. Hu, J.-N.; Zhu, X.-M.; Han, L.-K.; Saito, M.; Sun, Y.-S.; Yoshikawa, M.; Kimura, Y.; Zheng, Y.-N. Anti-Obesity Effects of Escins Extracted from the Seeds of Aesculus Turbinata BLUME (Hippocastanaceae). Chem. Pharm. Bull. 2008, 56, 12–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Kimura, H.; Ogawa, S.; Katsube, T.; Jisaka, M.; Yokota, K. Antiobese Effects of Novel Saponins from Edible Seeds of Japanese Horse Chestnut (Aesculus Turbinata BLUME) after Treatment with Wood Ashes. J. Agric. Food Chem. 2008, 56, 4783–4788. [Google Scholar] [CrossRef]
  105. Aoe, S.; Kudo, H.; Sakurai, S. Effects of Liquid Konjac on Parameters Related to Obesity in Diet-Induced Obese Mice. Biosci. Biotechnol. Biochem. 2015, 79, 1141–1146. [Google Scholar] [CrossRef]
  106. Kim, J.H.; Hahm, D.H.; Yang, D.C.; Kim, J.H.; Lee, H.J.; Shim, I. Effect of Crude Saponin of Korean Red Ginseng on High-Fat Diet-Induced Obesity in the Rat. J. Pharmacol. Sci. 2005, 97, 124–131. [Google Scholar] [CrossRef] [Green Version]
  107. Suruga, K.; Tomita, T.; Kadokura, K.; Arai, T. Rhus Verniciflua Leaf Extract Suppresses Obesity in High-Fat Diet-Induced Obese Mice. Food Nutr. Res. 2019, 63. [Google Scholar] [CrossRef] [Green Version]
  108. Weidner, C.; Wowro, S.J.; Freiwald, A.; Kodelja, V.; Abdel-Aziz, H.; Kelber, O.; Sauer, S. Lemon Balm Extract Causes Potent Antihyperglycemic and Antihyperlipidemic Effects in Insulin-Resistant Obese Mice. Mol. Nutr. Food Res. 2014, 58, 903–907. [Google Scholar] [CrossRef]
  109. Alsaggar, M.; Bdour, S.; Ababneh, Q.; El-Elimat, T.; Qinna, N.; Alzoubi, K.H. Silibinin Attenuates Adipose Tissue Inflammation and Reverses Obesity and Its Complications in Diet-Induced Obesity Model in Mice. BMC Pharmacol. Toxicol. 2020, 21, 8. [Google Scholar] [CrossRef] [PubMed]
  110. Joung, H.; Kim, B.; Park, H.; Lee, K.; Kim, H.-H.; Sim, H.-C.; Do, H.-J.; Hyun, C.-K.; Do, M.-S. Fermented Moringa Oleifera Decreases Hepatic Adiposity and Ameliorates Glucose Intolerance in High-Fat Diet-Induced Obese Mice. J. Med. Food 2017, 20, 439–447. [Google Scholar] [CrossRef] [PubMed]
  111. Metwally, F.M.; Rashad, H.M.; Ahmed, H.H.; Mahmoud, A.A.; Abdol Raouf, E.R.; Abdalla, A.M. Molecular Mechanisms of the Anti-Obesity Potential Effect of Moringa Oleifera in the Experimental Model. Asian Pac. J. Trop. Biomed. 2017, 7, 214–221. [Google Scholar] [CrossRef]
  112. Gourineni, V.; Shay, N.F.; Chung, S.; Sandhu, A.K.; Gu, L. Muscadine Grape (Vitis Rotundifolia) and Wine Phytochemicals Prevented Obesity-Associated Metabolic Complications in C57BL/6J Mice. J. Agric. Food Chem. 2012, 60, 7674–7681. [Google Scholar] [CrossRef] [Green Version]
  113. Lee, H.A.; Cho, J.-H.; Afinanisa, Q.; An, G.-H.; Han, J.-G.; Kang, H.J.; Choi, S.H.; Seong, H.-A. Ganoderma Lucidum Extract Reduces Insulin Resistance by Enhancing AMPK Activation in High-Fat Diet-Induced Obese Mice. Nutrients 2020, 12, 3338. [Google Scholar] [CrossRef] [PubMed]
  114. Chang, C.-J.; Lin, C.-S.; Lu, C.-C.; Martel, J.; Ko, Y.-F.; Ojcius, D.M.; Tseng, S.-F.; Wu, T.-R.; Chen, Y.-Y.M.; Young, J.D.; et al. Ganoderma Lucidum Reduces Obesity in Mice by Modulating the Composition of the Gut Microbiota. Nat. Commun. 2015, 6, 7489. [Google Scholar] [CrossRef] [Green Version]
  115. Lee, M.R.; Begum, S.; Oh, D.S.; Wee, A.J.; Yun, B.S.; Sung, C.K. Ameliorating Effect of Mycoleptodonoides Aitchisonii on High-Fat Diet-Induced Obese Mice. Prev. Nutr. Food Sci. 2014, 19, 69–74. [Google Scholar] [CrossRef] [Green Version]
  116. Chi, Q.; Wang, G.; Sheng, Y.; Xu, W.; Shi, P.; Zhao, C.; Huang, K. Ethanolic Extract of the Golden Oyster Mushroom, Pleurotus Citrinopileatus (Agaricomycetes), Alleviates Metabolic Syndrome in Diet-Induced Obese Mice. Int. J. Med. Mushrooms 2017, 19, 1001–1008. [Google Scholar] [CrossRef]
  117. Chen, Q.; Wu, X.; Liu, L.; Shen, J. Polyphenol-Rich Extracts from Oiltea Camellia Prevent Weight Gain in Obese Mice Fed a High-Fat Diet and Slowed the Accumulation of Triacylglycerols in 3T3-L1 Adipocytes. J. Funct. Foods 2014, 9, 148–155. [Google Scholar] [CrossRef]
  118. Shen, Y.; Song, S.J.; Keum, N.; Park, T. Olive Leaf Extract Attenuates Obesity in High-Fat Diet-Fed Mice by Modulating the Expression of Molecules Involved in Adipogenesis and Thermogenesis. Evid. Based Complement. Alternat. Med. 2014, 2014, 1–12. [Google Scholar] [CrossRef] [Green Version]
  119. Han, L.-K.; Takaku, T.; Li, J.; Kimura, Y.; Okuda, H. Anti-Obesity Action of Oolong Tea. Int. J. Obes. 1999, 23, 98–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Han, L.-K.; Zheng, Y.-N.; Yoshikawa, M.; Okuda, H.; Kimura, Y. Anti-Obesity Effects of Chikusetsusaponins Isolated from Panax Japonicus Rhizomes. BMC Complement. Altern. Med. 2005, 5, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Kim, M.-J.; Kim, H.K. Perilla Leaf Extract Ameliorates Obesity and Dyslipidemia Induced by High-Fat Diet: ANTIOBESITY EFFECT OF PERILLA LEAF EXTRACT. Phytother. Res. 2009, 23, 1685–1690. [Google Scholar] [CrossRef] [PubMed]
  122. Han, L.-K.; Xu, B.-J.; Kimura, Y.; Zheng, Y.; Okuda, H. Platycodi Radix Affects Lipid Metabolism in Mice with High Fat Diet–Induced Obesity. J. Nutr. 2000, 130, 2760–2764. [Google Scholar] [CrossRef] [Green Version]
  123. Lei, F.; Zhang, X.N.; Wang, W.; Xing, D.M.; Xie, W.D.; Su, H.; Du, L.J. Evidence of Anti-Obesity Effects of the Pomegranate Leaf Extract in High-Fat Diet Induced Obese Mice. Int. J. Obes. 2007, 31, 1023–1029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Park, Y.H.; An, M.; Kim, J.-K.; Lim, Y.-H. Antiobesity Effect of Ethanolic Extract of Ramulus Mori in Differentiated 3T3-L1 Adipocytes and High-Fat Diet-Induced Obese Mice. J. Ethnopharmacol. 2020, 251, 112542. [Google Scholar] [CrossRef] [PubMed]
  125. Kang, M.-C.; Kang, N.; Kim, S.-Y.; Lima, I.S.; Ko, S.-C.; Kim, Y.-T.; Kim, Y.-B.; Jeung, H.-D.; Choi, K.-S.; Jeon, Y.-J. Popular Edible Seaweed, Gelidium Amansii Prevents against Diet-Induced Obesity. Food Chem. Toxicol. 2016, 90, 181–187. [Google Scholar] [CrossRef] [PubMed]
  126. Kang, J.-H.; Lee, H.-A.; Kim, H.-J.; Han, J.-S. Gelidium Amansii Extract Ameliorates Obesity by Down-Regulating Adipogenic Transcription Factors in Diet-Induced Obese Mice. Nutr. Res. Pract. 2017, 11, 17. [Google Scholar] [CrossRef] [Green Version]
  127. Bargallo, M.; Grau, A.; Fernandezlarrea, J.; Anguiano, G.; Segarra, M.; Rovira, M.; Ferre, L.; Olive, M. Moderate Red-Wine Consumption Partially Prevents Body Weight Gain in Rats Fed a Hyperlipidic Diet. J. Nutr. Biochem. 2006, 17, 139–142. [Google Scholar] [CrossRef]
  128. Villalpando-Arteaga, E.V.; Mendieta-Condado, E.; Esquivel-Solís, H.; Canales-Aguirre, A.A.; Gálvez-Gastélum, F.J.; Mateos-Díaz, J.C.; Rodríguez-González, J.A.; Márquez-Aguirre, A.L. Hibiscus sabdariffa L. Aqueous Extract Attenuates Hepatic Steatosis through down-Regulation of PPAR-γ and SREBP-1c in Diet-Induced Obese Mice. Food Funct. 2013, 4, 618. [Google Scholar] [CrossRef]
  129. Yan, K.; Wang, X.; Pan, H.; Wang, L.; Yang, H.; Liu, M.; Zhu, H.; Gong, F. Safflower Yellow and Its Main Component HSYA Alleviate Diet-Induced Obesity in Mice: Possible Involvement of the Increased Antioxidant Enzymes in Liver and Adipose Tissue. Front. Pharmacol. 2020, 11, 482. [Google Scholar] [CrossRef] [PubMed]
  130. Choi, S.-I.; Cho, I.-H.; Han, S.H.; Jeon, Y.-J.; Choi, J.-G.; Kim, J.-S.; Lee, J.-H. Antiobesity Effects of Salvia Plebeia R. Br. Extract in High-Fat Diet-Induced Obese Mice. J. Med. Food 2016, 19, 1048–1056. [Google Scholar] [CrossRef] [PubMed]
  131. Kwon, D.; Kim, S.H.; Son, S.W.; Seo, J.; Jeong, T.B.; Kim, K.-M.; Jung, J.-C.; Jung, M.S.; Lee, Y.-H.; Jung, Y.-S. Germinated Soybean Embryo Extract Ameliorates Fatty Liver Injury in High-Fat Diet-Fed Obese Mice. Pharmaceuticals 2020, 13, 380. [Google Scholar] [CrossRef]
  132. Yamasaki, M.; Ogawa, T.; Wang, L.; Katsube, T.; Yamasaki, Y.; Sun, X.; Shiwaku, K. Anti-Obesity Effects of Hot Water Extract from Wasabi (Wasabia Japonica Matsum.) Leaves in Mice Fed High-Fat Diets. Nutr. Res. Pract. 2013, 7, 267. [Google Scholar] [CrossRef] [Green Version]
  133. Neyrinck, A.M.; Bindels, L.B.; De Backer, F.; Pachikian, B.D.; Cani, P.D.; Delzenne, N.M. Dietary Supplementation with Chitosan Derived from Mushrooms Changes Adipocytokine Profile in Diet-Induced Obese Mice, a Phenomenon Linked to Its Lipid-Lowering Action. Int. Immunopharmacol. 2009, 9, 767–773. [Google Scholar] [CrossRef] [PubMed]
  134. Jung, Y.-M.; Lee, S.-H.; Lee, D.-S.; You, M.-J.; Chung, I.K.; Cheon, W.H.; Kwon, Y.-S.; Lee, Y.-J.; Ku, S.-K. Fermented Garlic Protects Diabetic, Obese Mice When Fed a High-Fat Diet by Antioxidant Effects. Nutr. Res. 2011, 31, 387–396. [Google Scholar] [CrossRef]
  135. Arçari, D.P.; Bartchewsky, W.; dos Santos, T.W.; Oliveira, K.A.; Funck, A.; Pedrazzoli, J.; de Souza, M.F.F.; Saad, M.J.; Bastos, D.H.M.; Gambero, A.; et al. Antiobesity Effects of Yerba Maté Extract (Ilex Paraguariensis) in High-Fat Diet-Induced Obese Mice. Obesity 2009, 17, 2127–2133. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Risk factors and diseases associated to obesity.
Figure 1. Risk factors and diseases associated to obesity.
Obesities 02 00015 g001
Figure 2. Rodent models of obesity that can be applied to natural compounds research.
Figure 2. Rodent models of obesity that can be applied to natural compounds research.
Obesities 02 00015 g002
Table 2. Effects of natural products on HFD-induced obesity models.
Table 2. Effects of natural products on HFD-induced obesity models.
Food Product/Plant Bioactive Compounds/Extraction MethodStrainDose and TreatmentObserved EffectsReference
Abeliophyllum distichumN.A. C57BL/6 mice ♂ 100 and 300 mg/kg/day (oral administration), for 8 weeks ↓ B.w.,[57]
↓ Lipid accumulation (liver and BAT)
↓ Expression of lipogenic genes (PPARγ, C/EBPα, ACC, and FAS) (at 300 mg/kg).
↑ Expression of p-AMPK and p-ACC (at 300 mg/kg)
Acanthopanax senticosusN.A.C57BL/6J mice ♂ 0.5 g/kg (oral administration), for 12 weeks↓ B.w. gain [58]
↓ Abdominal fat accumulation
↓ Liver TG accumulation
↓ LDL-cholesterol serum levels
Acanthopanax sessiliflorusSaponinsICR mice ♀1% and 0.5% (w/w; mixed with the diet), for 4 weeks↓ B.w. [59]
American ginseng
(Panax quinquefolium), stems and leaves
Saponins ICR mice ♀ 1% and 3% (w/w; mixed with the diet), for 8 weeks↓ Adipose tissue weight[60]
↓TG plasma levels
Balloon flower
(Platycodon grandiflorum)
Water extractSprague Dawley rats ♂ 150 mg/kg/day (oral administration), for 7 weeks↓ B.w. gain[61]
↓ Food intake
↓ Subcutaneous adipose tissue weight
↓ Subcutaneous adipocytes size
↓ TC and TG plasma levels
Bitter melon
(Momordica charantia L.)
Ethanol extractC57BL/6 mice ♂ 250 and 500 mg/kg/day (mixed with the diet), for 12 weeks↓ B.w. [62]
↓ WAT weight
↑ Expression of SIRT-1
Suppressed PPARγ and SREBP-1 expressions of WAT
Blackcurrant
(Ribes nigrum)
25% anthocyanins and 40% polyphenolsC57BL/6J mice ♂0.1% (w/w; mixed with the diet), for 12 weeks↓ B.w., [63]
↓ Adipocyte size of the epididymal fat
↓ Inflammatory gene expression in the splenocytes (TNFα, IL-6, IL-1β)
↑ Gene expression in skeletal muscle (ACOX-1, PPARα, PPARδ, UCP-2, UCP-3, PGC-1α, TFAM)
Black tea extractPolyphenol fractionC57BL/6N mice ♀ 5% of extract (w/w; mixed with the diet), for 8 weeks↓ B.w., [64]
↓ Parametrial adipose tissue
↓ Liver lipid content
Black wattle tree
(Acacia meansii)
Acacia polyphenolKKAy mice ♂ 2.5% and 5% (w/w; mixed with the diet), for 7 weeks↓ B.w. [65]
↓ Glucose and insulin plasma levels
↓ mRNA expression of fat acid synthesis-related genes (SREBP-1c, ACC and FAS) in the liver
↓ mRNA expression of TNF-α in WAT
↑ mRNA expression of adiponectin in WAT
↑ mRNA expression of energy expenditure-related genes (PPARα, PPARδ, CPT1, ACOX and UCP-3) in skeletal muscle
↑ Protein expression of CPT1, ACOX and UCP-3
Broccoli florets and stalks
(Brassica oleracea L. var. italica)
Aqueous extractsAlbino rats ♂200 and 400 mg/kg/day (oral administration), for 1 month↓ B.w. gain[66]
↓ Adipose tissue index
↓ Serum levels of insulin, glucose, leptin, resistin and HOMA-IR index
↑ Adiponectin serum levels
↓ Serum levels of TC, TG and LDL-cholesterol
↑ HDL-cholesterol serum levels
N.A.C57BL/6J mice ♂10% florets or 10% stalks (w/w; mixed with the diet), for 17 weeks↓ Serum insulin levels and HOMA-IR index (only florets)[67]
↑ Adiponectin receptors 1 and 2 mRNA expression (only florets)
Broccoli microgreens juice
(Brassica oleracea L. var. italica)
N.A.C57BL/6J mice ♂20 g/kg/day (oral gavage), for 10 weeks↓ B.w. [68]
↓ WAT mass
↓ Liver fat
↓ Adipocyte size
↑ Water intake
↑ Glucose tolerance and insulin sensitivity
↓ Serum insulin levels, HOMA-IR index
↓ Serum levels of TG and LDL-cholesterol
↓ Serum levels of inflammatory cytokines (IL-1β, IL-6 and TNFα)
Broccoli sprouts extract
(Brassica oleracea L. var. italica)
Glucoraphanin-rich extractC57BL/6JSlc mice ♂0.3% (w/w; 2.2% extract powder mixed with the diet), for 14 weeks↓ B.w. gain[69]
↓ Fat mass
↓ Liver weight
↓ Hepatic steatosis
↓ Hepatic oxidative stress
↑ Energy expenditure and core body temperature
↑ Glucose tolerance and insulin sensitivity
↓ Plasma insulin levels and HOMA-IR index
↑ Insulin stimulated Akt phosphorylation on Ser473 in the liver, quadriceps muscle and epididymal WAT
↑ UCP-1 protein levels in epididymal and inguinal WAT
Brown alga
(Sargassum thunbergii)
Ethanol ExtractC57BL/6 mice ♂ 100 and 300 mg/kg/day (oral administration), for 7 weeks↓ B.w. [70]
↓ WAT mass
↓ Occurrence of fatty liver
↓ Insulin, TG and TC serum levels
↓ Gene expression of PPARγ in WAT
↑ Expression of thermogenic genes (UCP-1 and UCP-3) in BAT
Brown alga
(Ecklonia cava)
N.A.C57BL/6 mice ♂ 5, 25 or 150 mg/kg/day (mixed with the diet), for 10 weeks↓ B.w. [71]
↓ Liver weight,
↓ Epididymal, perirenal and mesenteric WAT
↓ Insulin, leptin and glutamate pyruvate transaminase serum levels
↓ Serum and hepatic levels of TG
↓ Hepatic protein expression levels of C/ERPα, PPARy, SREBP-1c, A-FABP, FAS and leptin
↑ Hepatic protein expression levels of GLUT4
Brown alga Wakame
(Undaria pinnatifida)
Fucoxanthin C57BL/6J mice ♂ 1.06% and 2.22% % (w/w; mixed with the diet), for 5 weeks↓ B.w. [72]
↓ WAT
↓ Plasma levels of insulin, glucose, leptin and LDL-cholesterol
↑ Plasma levels of TC
↓ mRNA expression of MCP-1 and leptin in WAT
↑ mRNA expression of Adrb3 in WAT
↑ mRNA expression of GLUT4 in skeletal muscle
Brown alga:
Undaria Pinnatifida (UP),
Laminaria Japonica (LJ),
Sargassum Fulvellum (SF),
Hizikia Fusiforme (HF)
N.A.C57BL/6N mice ♂ 5% of freeze-dried UP, LJ, SF, or HF (w/w; mixed with the diet), for 16 weeks↓ Plasma levels of leptin[73]
↓ Plasma levels of adiponectin (for UP supplementation)
↓ Formation of CLS in gonadal adipose tissue
↓ Insulin resistance (for LJ supplementation)
Cannabis sativaCannabinoid
Δ9-tetrahydrocannabivarin
C57BL/6 mice ♀0.3, 1, 2.5, 5 and 12.5 mg/kg twice daily (oral gavage), for 30 days↓ Glucose and insulin plasma levels for highest doses[27]
Improvement of insulin sensitivity index for highest doses.
Cauliflower mushroom
(Sparassis crispa)
Lupane-type saponins C57BL/6 mice ♂ 1%, 3% and 5% (w/w; mixed with the diet), for 12 weeks↓ B.w. gain[74]
↓ Food intake
↓ FER
↓ Serum levels of TC and TG
↓ Liver lipids
↓ Occurrence of fatty liver deposits and steatosis
Chinese willow dry leaves
(Salix matsudana)
Polyphenol fractionICR mice ♂ 2% and 5% (w/w; mixed with the diet), for 9 weeks↓ B.w.[75]
↓ Parametrial adipose tissue
↓ Adipocyte size
↓ Hepatic TC (at 5% concentration)
Chitooligosaccharide N.A.C57BL/6N mice ♂ 1% or 3% (w/w; mixed with the diet), for 5 months↓ B.w. gain (at 3% concentration)[76]
↓ Epididymal adipocyte size (at 3% concentration)
↓ Serum levels of TG and TC
↓ Hepatic levels of total lipid and TG (at 3% concentration)
↓ Serum levels of AST and ALT (at 3% concentration)
Chitosan and water-soluble derivatives chitosan oligosaccharidesN.A.Sprague Dawley rat ♂ 250, 500 and 1000 mg/kg × d (w/w; mixed with the diet), for 6 weeks↓ B.w. gain[77]
↓ Serum levels of LDL-cholesterol, TC and TG
↓ Hepatic levels of TC, TG and TBA
↑ Fecal levels of TC, TG and TBA
↓ ALT and AST levels in serum and liver
↑ SOD levels in serum and liver
↓ Growth inhibition of subcutaneous and mesenteric WAT
Relieved fatty liver
↑ Liver mRNA expression of PPARα and HL
Chitosan (CTS) and water-soluble chitosan microspheresN.A.Sprague Dawley ♂ 225 and 450 mg/kg/day (oral administration), for 4 weeks↓ B.w. gain[78]
↓ Blood lipids and plasma viscosity
↑ Serum levels of SOD
Clusia nemorosa L.Betulinic acidSwiss mice ♂ 50 mg/L (in drinking water), for 15 weeks↓ B.w. [79]
↓ Abdominal fat accumulation
↓ Plasma levels of glucose, TG and TC
↑ Plasma levels of insulin and leptin
↓ Plasma levels of amylase and ghrelin
Cocoa powder
(Theobroma cacao L.)
Cocoa polyphenol extract C57BL/6N mice ♂ 40 and 200 mg/kg (mixed with the diet), for 5 weeks↓ B.w. gain [80]
↓ Fat accumulation
Common bean dried
(Phaseolus vulgaris L.)
N.A. C57BL/6J mice ♂ 30% and 46.5% (w/w; mixed with the diet), for 7 or 12 days, respectively↓ B.w. and Lee Index[81]
↓ Plasma levels of TG, LDL-cholesterol
Cornelian cherries
(Cornus mas)
AnthocyaninsC57BL/6 mice ♂1 g/kg (w/w; mixed with the diet), for 8 weeks↓ B.w.,[82]
Improvement of glucose tolerance
↓ Lipid accumulation in liver
↑ Plasma insulin levels
Ursolic acidC57BL/6 mice ♂500 mg/kg (w/w; mixed with the diet), for 8 weeksImprovement of glucose tolerance[82]
↓ Lipid accumulation in liver
↑ Plasma insulin levels
Cudrania tricuspidata fruits6,8-diprenylgenistein (DPG) (a major isoflavonoid)C57BL/6J mice ♂ 10 and 30 mg/mL (in distilled water) (oral administration of 10 mL/kg), for 6 weeks↓ B.w. gain[83]
↓ FER
↓ Epididymal fat weight
↓ Epididymal adipocyte size
↓ Liver fat accumulation and weight
↓ Serum levels of TC, TG, HDL-cholesterol, LDL-cholesterol, ALT and AST
↓ Protein levels of PPARγ, C/EBPα and leptin in adipose tissue
↑ Protein levels of adiponectin in adipose tissue
↑ Phosphorylation of AMPK and ACC
Curcumin
(Curcuma longa)
CurcuminoidsSprague Dawley rat ♂ 0.2% and 1% (w/w; mixed with the diet), for 2 weeks↓ Epididymal adipose tissue weight[84]
↓ Hepatic levels of TC and TG (at 1% concentration)
↑ Hepatic ACOX activity
N.A.C57BL/6 mice ♂ 500 mg/kg (w/w; mixed with the diet), for 12 weeks↓ B.w. gain and body fat[85]
↓ Liver weight and hepatic steatosis
↓ TC serum levels
↓ mRNA expression of VEGF and VEGFR-2 in adipose tissue
↓ Microvessel density in adipose tissue
↑ Phosphorylation of AMPK and ACC in adipose tissue
↑ mRNA expression of CPT-1 in adipose tissue
↓ mRNA expression of GPAT-1 in adipose tissue
↓ mRNA expression of PPARγ and C/EBPα in adipose tissue
Dioscorea nipponica MakinoMethanol extractSprague Dawley ♂2% or 5% (w/w; mixed with the diet), for 8 weeks↓ B.w. gain[86]
↓ Subcutaneous, perirenal and epididymal fat
↓ Plasma levels of TG, TC, VLDL-cholesterol and atherogenic index
↑ HDL-cholesterol plasma levels
↑ Fecal fat excretion
Dioscorea oppositifolia n-BuOH extract of D. oppositifoliaICR mice ♀ 100 mg/kg (oral administration), for 8 weeks↓ B.w. gain [87]
↓ FER
↓ Parametrial adipose tissue weight
↓ Liver weight
↓ Serum levels of TG, TC LDL-cholesterol and atherogenic index
↑ HDL (in serum)
↓ Hepatic levels of total lipids, TG, TC, AST and ALT
↑ Fecal excretion of TG, TC and total lipids
Fig fruit
(Ficus carica L.)
Aqueous-ethanolic extractWistar male ♂ 400 mg/kg (w/w; mixed with the diet), for 8 weeks↓ Plasma levels of TC, TG and LDL-cholesterol [88]
↑ HDL-cholesterol plasma levels
↓ TBARS levels in liver, kidney and heart
↑ Antioxidant enzymes (GPx, SOD and CAT) in liver, kidney and heart
Fraxinus excelsior L. seed extract (FraxiPureTM)1 phenolic compound, and 9 secoiridoid glucosides C57BL/6J mice ♂ 0.5% (w/w; mixed with the diet), for 16 weeks↓ B.w. gain[89]
↓ omental and retroperitoneal fat
↓ fasting blood glucose levels and plasma insulin levels
↓ Liver weight gain and incidence of fatty liver
Galangal
(Alpinia officinarum Hance)
Ethanolic extractSprague Dawley ♂ 3% and 5% (w/w; mixed with the diet), for 6 weeks↓ B.w. gain[90]
↓ FER
↓ Adipose tissue weight
↓ Serum levels of TC, TG, LDL-cholesterol, atherogenic index, leptin and ALT
↑ HDL-cholesterol serum levels
↓ Hepatic levels of TC and TG
Garlic
(Allium sativum L.)
Chlorophyll, carotenoids and vitamin C C57BL/6J mice ♂ 100, 250 and 500 mg/kg/day (oral administration), for 4 weeks↓ B.w. gain[91]
↓ FER
↓ WAT weight and adipocyte size
↓ Serum levels of TC, TG and leptin
↓ Serum levels of fasting glucose, insulin and HOMA-IR
↑ Serum levels of high-molecular-weight adiponectin
↓ Hepatic levels of TC and TG
↑ Fecal TG excretion
↓ Hepatic FAS levels
↑ Hepatic CPT-1A levels
↓ HMG-CoA reductase activity
↑ Hepatic antioxidant enzyme activities (SOD, GST, GSH, GPx and GR)
↓ Hepatic MDA activity
Ginger rhizomes
(Zingiber officinale Roscoe)
N.A.C57BL/6J mice ♂ 500 mg/kg/day (oral gavage), for 16 weeks↓ B.w. [92]
↓ Fat accumulation
↓ Serum levels of glucose, TG and TC
Enhancement of BAT function and activation of WAT browning
Ginseng
(Panax ginseng)
Saponins Balb/c ♂ mice3% (w/w; mixed with the diet), for 3 weeks↓ B.w.[93]
↓ FER
↓ TG plasma levels
N.A.C57BL/6J mice ♂ 0.5 g/kg (w/w; mixed with the diet), for 15 weeks↓ Body fat mass, [94]
↑ Glucose tolerance and insulin sensitivity
↓ Plasma levels of TG, HDL-cholesterol, insulin and leptin
↑ Body temperature
Prevented hypertension
↑ Fatty acid oxidation in liver
↑ mRNA expression of C/EBPa, PPARγ and FAS in adipose tissue
Golden mushroom (Pleurotus citrinopileatus)Water extractC57BL/6J mice ♂ 400 and 800 mg/kg/day (oral gavage), for 12 weeks ↓ B.w. gain[95]
↓ Food intake
↓ Fat accumulation
↑ Glucose tolerance
↓ Serum levels of TG, TC, LDL-cholesterol, AST, nonesterified fatty acid and creatinine
↑ HDL-cholesterol serum levels
Grape skin extract (Vitis aestivalis)Phenolic compoundsC57BLK/6J mice ♂ 250 mg/kg/day (mixed with the diet), for 12 weeks↑ B.w.[96]
↓ Fasting blood glucose and plasma CRP levels
Green alga
(Caulerpa okamurae)
Ethanolic extractC57BL/6 mice ♂ 250 mg/kg (oral gavage), for 10 weeks↓ B.w., [97]
↓ Fat weight
↓ Liver weight
↓ Plasma levels of FFA, TG, TC, glucose and insulin
↓ Hepatic levels of FFA, TG, TC, and total lipid
↓ PPARγ and C/EBPα protein levels in adipose tissue
↓ mRNA expression of FAS, SREBP-1c, ACC, and CD36 in adipose tissue
Green alga
(Codium fragile)
Ethanol ExtractC57BL/6 mice ♂ 600 mg/kg/day (intragastric administration), for 12 weeks ↓ B.w., [98]
↓ Size of adipocytes
↓ Serum levels of TC and glucose
↑ Abundance of Bacteroidetes species in the gut
↓ Abundance of Verrucomicrobia species in the gut
Gymnema sylvestreMethanol extractC57BL/6 mice ♂ 1 g/kg (w/w; mixed with the diet), for 4 weeks ↓ B.w. [99]
↓ Abdominal and epididymal fat weight
↓ Serum levels of TC, TG, LDL-cholesterol, VLDL-cholesterol, leptin, AST and ALT
↓ Occurrence of hepatic steatosis
Halophyte
(Nitraria retusa)
Ethanol extract C57BL/6 mice ♂ 50 and 100 mg/kg/day (oral administration), for 4 weeks↓ B.w. gain [100]
↓ FER
↓ Adipose tissue weight
↓ Serum levels of TG and glucose
↑ HDL-cholesterol
↑ Hepatic mRNA expression of PPARγ1, PPARα, ACC-1, CPT1 and LPL
↓ Hepatic mRNA expression of FAS
Indian lotus leaves extract
(Nelumbo nucifera Gaertn.)
Alcoholic extractICR mice ♀ 5% (w/w; mixed with the diet), for 5 weeks↓ B.w.[101]
↓ Parametrial adipose tissue weight
↓ Hepatic TG levels
Indian lotus (Nelumbo nucifera) and Peach tree (Prunus persica) mixtureN.A.C57BL/6 mice ♂ 0.1%, 0.2% and 0.4% (w/w; mixed with the diet), for 12 weeks↓ B.w. gain [102]
↓ Abdominal fat weight
↓ Liver weight
↓ Hepatic levels of TG and TC
↓ Serum levels of glucose, TC, ALT, AST and leptin
↑ Adiponectin serum levels
↑ AST/ALT and adiponectin/leptin ratios
↓ mRNA levels of FAS and SCD-1 in adipose tissue (at 0.4% concentration)
↑ mRNA levels of PGC-1a and PPARα in adipose tissue (at 0.4% concentration)
Japanese Horse Chestnut (Aesculus turbinata BLUME)Escins (saponin)ICR mice ♀ 0.35%, 1% and 2% (w/w; mixed with the diet), for 11 weeks↓ Parametrial adipose tissue (at 2% concentration) [103]
↓ Hepatic levels of TG
↑ TG fecal excretion (at 2% concentration)
SaponinsICR mice ♀ 0.1% and 0.5% (w/w; mixed with the diet), for 8 weeks↓ B.w.[104]
↓ Peritoneal adipose tissues
↓ TG plasma levels
↓ GOT activity
↑ TG fecal excretion
Konjac
(Amorphophallus konjac)
Liquid konjacC57BL/6J mice ♂ 2.5% and 5% (w/w; mixed with the diet), for 80 days↓ B.w. gain[105]
↓ FER
↓ Abdominal fat accumulation
↓ Liver weight
↓ Serum levels of TC, leptin, insulin and HOMA-IR
↓ Hepatic levels of TC and TG
↑ Fecal fat excretion
Korean red ginseng (Ginseng Radix Rubra) Crude saponin Sprague Dawley rats ♂ 200 mg/kg/day (intraperitoneal administration), for 3 weeks ↓ B.w. [106]
↓ Food intake
↓ Fat weight
↓ Serum leptin levels
↓ Expression of NPY neurons in the hypothalamus
Lacquer tree leaf extract
(Rhus verniciflua)
QuercetinC57BL/6 mice ♂ 1% and 2% (w/w; mixed with the diet), for 56 days ↓ B.w.[107]
↓ Intra-abdominal fat
↓ Plasma leptin levels
Lemon balm
(Melissa officinalis)
Ethanol extractC57BL/6J mice ♂ 200 mg/kg/day (in drinking water), for 6 weeks↓ Hyperglycemia and insulin resistance [108]
↓ Plasma levels of TG, nonesterified fatty acids and LDL/VLDL cholesterol ratio
↑ HDL/LDL cholesterol ratio
hyperglycemia
and insulin resistance,
hyperglycemia
and insulin resistance,
hyperglycemia
and insulin resistance
Milk thistle seeds extract
(Silybum marianum)
SilibininC57BL/6 mice ♂ 50 mg/kg (intraperitoneal injection), for 8 weeks↓ B.w. gain[109]
↓ Fat accumulation in liver
↓ Fat accumulation and adipose tissue hypertrophy
Reversed gene expression profile from pro-inflammatory to anti-inflammatory profile
Moringa oleifera L.N.A.C57BL/6J mice ♂ 250 mg/kg/day (oral administration), for 10 weeks↓ Liver weight and hepatic lipid accumulation [110]
↑ Glucose tolerance
↓ Oxidative stress, endoplasmic reticulum stress and lipotoxicity in quadriceps muscles
↓ Hepatic expression of genes involved in lipid synthesis (ACC, FAS, LPL and SREBP-1c)
↑ Hepatic genes involved in lipid oxidation (CD36 and ATGL)
↓ Proinflammatory cytokine mRNA expression (TNF-α, IL-1β, IL-6, IL-12 and MCP-1) in the liver, epididymal adipose tissue, and quadriceps
Ethanolic extract Wistar rats ♀ 600 mg/kg/day (oral administration), for 12 weeks↓ B.w.,[111]
Improvement of the atherogenic index and coronary artery index
↓ Serum glucose levels and insulin resistance
↓ mRNA expression of leptin and resistin in adipose tissue
↑ mRNA expression of adiponectin in adipose tissue
↓ Hepatic levels of AST and ALT
Muscadine wine phytochemical and muscadine grape phytochemical
(Vitis rotundifolia)
AnthocyaninsC57BL/6J mice ♂ 0.4% (w/w; mixed with the diet) of each phytochemical, for 15 weeks↓ B.w.[112]
↑ Glucose tolerance
↓ Plasma levels of FFA, TG, TC, CRP
Mushroom
(Ganoderma lucidum)
Ethanolic extractSpecific pathogen free (SPF) C57BL/6 mice ♂1%, 3% or 5% (w/w; mixed with the diet), for 12 weeks↓ B.w. gain[113]
↓ FER
↓ WAT weight
↓ Size of adipocytes in epidydimal WAT
↓ Liver weight
↓ Serum levels of TG, TC, HDL-cholesterol, LDL-cholesterol and FFA
↓ Serum levels of glucose, insulin and leptin
↑ Serum adiponectin levels
↑ Glucose tolerance and insulin sensitivity
↓ mRNA expression of lipogenic genes (FAS, SCD1 and SREBP-1c) in liver and WAT
Water extractC57BL/6NCrlBltw ♂ 2%, 4% and 8% (w/v; oral gavage of 100 µL, daily), for 8 weeks↓ B.w., [114]
↓ Epididymal and subcutaneous fat
↓ Liver weight
↓ mRNA expression levels pro-inflammatory cytokines (TNF-α, IL-1β, IL-6 and PAI-1) in the liver and adipose tissues
↑ mRNA expression levels of IL-10 in the liver and adipose tissues
Mushroom
(Mycoleptodonoides aitchisonii)
N.A.C57BL/6 mice ♂ 1%, 3% and 5% (w/w; mixed with the diet), for 12 weeks↓ B.w. gain[115]
↓ Food intake
↓ FER
↓ Adipose tissue weight
↓ Serum levels of TC and TG
↓ Hepatic lipid and TC levels
↓ Occurrence of fatty liver deposits and steatosis
↓ Epididymal adipocyte size
Mushroom
(Pleurotus citrinopileatus)
Ethanolic extract C57BL/6J mice ♂ 200 and 500 mg/kg, for 12 weeks ↓ B.w. gain[116]
↓ Fat accumulation
↑ Glucose tolerance
Oiltea camelliaEthanolic extractICR mice ♀ 100, 200 and 300 mg/kg (oral administration), for 4 weeks ↓ B.w.[117]
↓ Fat accumulation
↓ Serum levels of TC and TG
↑ HDL-cholesterol serum levels
↓ Hepatic FAS activity
Olive leaf extract
(Olea europaea L.)
Ethanolic extract C57BL/6N mice ♂ 0.15% (w/w; mixed with the diet), for 8 weeks↓ B.w. gain [118]
↓ Food intake
↓ FER
↓ Visceral fat-pad weights
↓ Plasma levels of glucose and leptin
↓ Plasma levels of TG, TC, LDL + VLDL cholesterol and FFA
↓ Gene expression of PPARγ, C/EBPα, CD36, FAS, and leptin in the epididymal adipose tissue
Oolong tea dry leaf
(Thea sinensis L.)
CaffeineICR mice ♀ 5% (w/w; mixed with the diet), for 10 weeks↓ B.w. [119]
↓ Parametrial adipose tissue
↓ Accumulation of liver TG
Panax japonicus rhizomesChikusetsusaponinsICR mice ♀ 1% and 3% (w/w; mixed with the diet), for 9 weeks↓ B.w. [120]
↓ Parametrial adipose tissue weight
↓ Liver weight
↓ Hepatic TG levels
↑ Feces weight and TG fecal excretion
Perilla leaf extract
(Perilla frutescens L.)
Ethanolic extractC57BL/6 mice ♀ 1% and 3% (w/w; mixed with the diet), for 4 weeks↓ B.w. gain[121]
↓ FER
↓ Epididymal fat mass
↓ Liver weight
↓ Occurrence of hepatic steatosis
↓ Plasma levels of TG, TC and LDL-cholesterol
↑ HDL-cholesterol plasma levels
↓ Gene expression of ACC, GPDH and PPARγ in epididymal adipose tissue
Platycodi radix
(Platycodon grandiflorum)
SaponinsICR mice ♀ 5% (w/w; mixed with the diet), for 8 weeks↓ B.w., [122]
↓ Parametrial adipose tissue weight
↓ Hepatic TG levels
Pomegranate leaf extract
(Punica granatum)
10.6% ellagic acidICR mice ♀ and ♂ 400 and 800 mg/kg/day (oral gavage), for 5 weeks ↓ B.w.[123]
↓ Energy intake
↓ Adipose pad weight percents and Lee index
↓ Serum levels of glucose, TC, TG and TC/HDL-cholesterol ratio
Ramulus mori
(the twig of Morus alba L.)
Ethanolic extractC57BL/6 mice ♂ 20, 50 and 100 mg/kg/day (oral administration), for 7 weeks↓ B.w.[124]
↓ Epididymal adipose tissue weight
↓ Liver weight
↓ Lipid accumulation in the liver
↓ Serum levels of TC and TG
↓ mRNA expression and protein levels of PPARγ, C/EBPα, SREBP-1, ACC, FAS and SCD-1
↑ mRNA expression and protein levels of lipolytic genes (ATGL and HSL)
Red alga
(Gelidium amansii)
70% ethanol extractC57BL/6 mice ♂ 1% and 3% (w/w; mixed with the diet), for 12 weeks↓ B.w. [125]
↑ Food and water intake
↓ Epididymal fat weight
↓ Serum levels of TC, TG, glucose and insulin
Ethanolic extractC57BL/6J mice ♂ 0.5%, 1% and 2% (w/w; mixed with the diet), for 8 weeks ↓ B.w.[126]
↓ Epididymal and mesenteric adipose tissue weight
↓ Liver weight
↓ Plasma levels of TC, TG, LDL-cholesterol, FFA, and leptin
↑ Plasma levels of HDL-cholesterol and adiponectin
↓ Hepatic TC and TG levels
↓ Protein expression of FAS, SREBP-1c, PPARγ, and C/EBPα
↑ Protein expression of HSL and p-AMPK
Red wine2.09 g/L total polyphenolZucker lean rats ♂ Free access to red table wine (average consumption of 1.70 ± 0.38 mL/day per animal, corresponding to a dose of 3.4 ± 0.79 mg/day per animal of total polyphenols)↓ B.w. gain[127]
↓ Energy intake
↓ Epididymal fat weight
Roselle
(Hibiscus sabdariffa L.)
Anthocyanins (delphinidin-3-sambubioside and cyanidin-3-sambubioside)C57BL/6NHsd mice ♂ 33 mg/kg (oral gavage) three times a week, for 8 weeks↓ B.w. gain[128]
↓ WAT accumulation
↓ Serum levels of TG, LDL-cholesterol and glucose
↑ HDL-cholesterol serum levels
↓ Hepatic steatosis
↓ Hepatic mRNA levels of SREBP-1c, PPARγ, TNF-α and IL-1
↑ Hepatic CAT mRNA expression
Safflower
(Carthamus tinctorius L.)
Safflower yellow (SY) and hydroxysafflor yellow A (HSYA)C57BL/6 mice ♂ 200 mg/kg/day SY or HSYA (intraperitoneal injection), for 10 weeks↓ B.w.[129]
↓ WAT mass
↓ Blood glucose levels and HOMA-IR
↓ Serum ALT levels
↑ Hepatic SOD activity
↑ mRNA levels of antioxidant enzymes in liver and epididymal adipose tissues
Salvia plebeianEthanolic extractC57BL/6 mice ♂ 200 and 400 mg/kg/day (oral administration), for 8 weeks↓ B.w.[130]
↓ Adipose tissue weight
↓ Liver weight
↓ Hepatic steatosis, TG accumulation and inflammatory cells infiltration
↓ Serum levels of TG, HDL-cholesterol, leptin, adiponectin and glucose
↓ Adipocytes size in adipose tissue
↓ Expression of adipogenesis transcription factors and lipogenesis-related target genes in adipose tissue
Soybean
(Glycine max (L.) Merrill)
Ethanolic extract (soyasaponin Ab)C57BL/6 mice ♂ 15 and 45 mg/10 mL/kg/day (oral administration), for 10 weeks↓ B.w.[131]
↓ Adipose tissue weight
↓ Liver weight and steatosis
↓ Serum levels of TC, TG, FFA, LDL-cholesterol, AST and ALT
↓ Hepatic lipid synthesis (SREBP1c)
↑ Hepatic fatty acid oxidation (p-AMPKα, PPARα, PGC1α, and ACOX) and lipid export (MTTP and ApoB)
↓ Expression of inflammatory genes (TNFα, IL-6, IL-1β, iNOS, COX2, CD14 and F4/80) in liver
↓ WAT differentiation and lipogenesis (PPARγ, C/EBPα, and FAS)
↑ Browning genes (PGC1α, PRDM16, CIDEA, and UCP1) in adipose tissue
Wasabi leaf
(Wasabia japonica Matsum.)
Water extract C57J/BL mice ♂ 5% (w/w; mixed with the diet), for 163 days↓ B.w. gain[132]
↓ Liver weight
↓ Epididymal WAT
↓ Plasma levels of TC, leptin and γ-GTP
↓ Gene expression of PPARγ, leptin and C/EBPα in WAT
↑ Gene expression of adiponectin and ACOX1 in WAT
↓ Gene expression of PPARγ, SREBP-1c, ACC1, FAS and HMG-CoA reductase in liver
↑ PPARα gene expression in liver
White mushroom exoskeleton (Agaricus bisporus)Chitosan C57BL/6J mice ♂ 5% (w/w; mixed with the diet), for 10 weeks↓ B.w. gain[133]
↓ FER
↓ Adipose tissue weight
↓ Fat accumulation in liver and muscle
↓ TG levels in serum, liver and muscle
↓ TC levels in serum and muscle
↓ Serum levels of IL-6, leptin, resistin, insulin
↓ FIAF mRNA expression in visceral adipose tissue
↑ Caecal tissue weight and content weight
↑ Caecal total lipids and nonesterified fatty acid levels
↑ β-hydroxybutyrate plasma levels in postprandial state
Yeast (Saccharomyces cerevisiae)-fermented aged black garlic N.A.ICR mice ♀ 100, 200 and 400 mg/kg (10 mL/kg; oral administration), for 63 days↓ B.w.[134]
↓ Adipose tissue accumulation and adipocytes diameter
↓ Serum levels of TC, TG, LDL-cholesterol, AST, ALT, BUN and creatinine
↑ HDL-cholesterol serum levels
↓ Hepatic steatosis and hepatocyte hypertrophy
↓ Number of abnormal kidney tubules
Yerba maté extract (Ilex paraguariensis) Water extractSwiss mice ♂ 1 g/kg (oral gavage), for 8 weeks↓ B.w.[135]
↓ Epididymal fat weight
↓ Serum levels of TC, TG, LDL-cholesterol and glucose
↓ Expression levels of cytokines (TNF-α, IL-6 and leptin) and chemoattractant proteins (CCR2 and CCL2) in WAT
↓ Expression of genes involved in the regulation of blood pressure, vascular homeostasis or angiogenesis (angiotensinogen and PAI-1) in WAT
↑ Expression of genes involved in adipogenesis (PPARγ) and glucose and lipid metabolism (adiponectin) in WAT
↑ Expression of genes implicated in thermogenesis (PGC-1α and UCP-1) in BAT
↓ Macrophage infiltration marker (F4/80) in epididymal fat
♂, male; ♀, female; ↓, decrease; ↑, increase; ACC, acetyl-CoA carboxylase; ACOX-1, acyl-CoA oxidase 1; Adrb3, β3-adrenergic receptor; A-FABP, adipose fatty acid-binding protein; ALT, alanine aminotransferase; AST, aspartate aminotransferase; aP2, adipocyte fatty acid-binding protein; ApoB, apolipoprotein B; ATGL, adipose triglyceride lipase; BAT, brown adipose tissue; B.w., body weight; BUN, blood urea nitrogen; CAT, catalase; CCL2, C-C motif chemokine ligand2; CCR2, CCL receptor 2; CD14, cluster of differentiation 14; CD36, cluster of differentiation 36; C/EBPα, CCAAT/enhancer binding protein alpha; CIDEA, cell death-inducing DNA fragmentation factor-like effector A; CLS, crown-like structures; CPT1, carnitine palmitoyl-transferase 1; CRP, C-reactive protein; eNOS, endothelial nitric oxide-synthase; FAS, fatty acid synthase; FER, food efficiency ratio; FFA, free fatty acids; FIAF, fasting-induced adipose factor; γ-GTP, gamma-glutamyltranspeptidase; GLUT4, glucose transporter type 4; GOT, glutamic oxaloacetic transaminase; GPAT-1, glycerol-3-phosphate acyl transferase 1; GPDH, glycerol-3-phosphate dehydrogenase; GPx, glutathione peroxidase; GR, glutathione reductase; GSH, glutathione; GST, glutathione S-transferase; HDL, high-density lipoprotein; HL, hepatic lipase; HMG-CoA reductase, 3-hydroxy-3-methylglutaryl-coenzyme A reductase; HOMA-IR, homeostatic model assessment for insulin resistance; HSL, hormone-sensitive lipase; IL-1β, interleukin 1 beta; IL-6, interleukin 6; iNOS, inducible nitric oxide synthase; LDL, low density lipoprotein; LPL, lipoprotein lipase; MCP-1, monocyte chemoattractant protein 1; MDA, malondialdehyde; MSG, monosodium glutamate; MTTP, microsomal triglyceride transfer protein; N.A., not applicable; NO, nitric oxide; NPY, neuropeptide Y; p-ACC, phosphorylated ACC; PAI-1, plasminogen activator inhibitor 1; p-AMPK, phosphorylated adenosine monophosphate-activated protein kinase; PGC-1α, PPARγ coactivator 1 alpha; PPAR-α, peroxisome proliferator-activated receptor alpha; PPARδ, peroxisome proliferator-activated receptor delta; PPARγ, peroxisome proliferator-activated receptor gamma; PRDM16, PR domain containing 16; SCD-1, stearoyl-CoA desaturase-1; SIRT1, Sirtuin 1; SOD, superoxide dismutase; SREBP-1c, sterol regulatory element binding protein 1c; TBA, total bile acid; TBARS, thiobarbituric acid reacting substances; TC, total cholesterol; TFAM, mitochondrial transcription factor A; TG, triglycerides; TNFα, tumor necrosis factor alpha; UCP-1, uncoupling protein 1; UCP-2, uncoupling protein 2; UCP-3, uncoupling protein 3; VEGF, vascular endothelial growth factor; VEGFR-2, VEGF receptor 2; VLDL, very low density lipoprotein; WAT, white adipose tissue; weeks, weeks.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Martins, T.; Ferreira, T.; Nascimento-Gonçalves, E.; Castro-Ribeiro, C.; Lemos, S.; Rosa, E.; Antunes, L.M.; Oliveira, P.A. Obesity Rodent Models Applied to Research with Food Products and Natural Compounds. Obesities 2022, 2, 171-204. https://doi.org/10.3390/obesities2020015

AMA Style

Martins T, Ferreira T, Nascimento-Gonçalves E, Castro-Ribeiro C, Lemos S, Rosa E, Antunes LM, Oliveira PA. Obesity Rodent Models Applied to Research with Food Products and Natural Compounds. Obesities. 2022; 2(2):171-204. https://doi.org/10.3390/obesities2020015

Chicago/Turabian Style

Martins, Tânia, Tiago Ferreira, Elisabete Nascimento-Gonçalves, Catarina Castro-Ribeiro, Sílvia Lemos, Eduardo Rosa, Luís Miguel Antunes, and Paula Alexandra Oliveira. 2022. "Obesity Rodent Models Applied to Research with Food Products and Natural Compounds" Obesities 2, no. 2: 171-204. https://doi.org/10.3390/obesities2020015

Article Metrics

Back to TopTop