Next Article in Journal
Gait Phase Estimation Based on User–Walker Interaction Force
Previous Article in Journal
A Data-Driven Forecasting Strategy to Predict Continuous Hourly Energy Demand in Smart Buildings
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Natural Dietary and Medicinal Plants with Anti-Obesity Therapeutics Activities for Treatment and Prevention of Obesity during Lock Down and in Post-COVID-19 Era

by
Wenli Sun
1,*,†,
Mohamad Hesam Shahrajabian
1,*,† and
Qi Cheng
2,3
1
Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
2
State Key Laboratory of North China Crop Improvement and Regulation, Hebei Agricultural University, Baoding 071000, China
3
Global Alliance of HeBAU-CLS&HeQiS for BioAl-Manufacturing, College of Life Sciences, Hebei Agricultural University, Baoding 071000, China
*
Authors to whom correspondence should be addressed.
Authors contributed equally to this research.
Appl. Sci. 2021, 11(17), 7889; https://doi.org/10.3390/app11177889
Submission received: 20 July 2021 / Revised: 14 August 2021 / Accepted: 17 August 2021 / Published: 26 August 2021

Abstract

:
Overweight and obesity have become global epidemics, especially during the lockdown due to the COVID-19 pandemic. The potential of medicinal plants as a better and safe option in treating obesity and overweight has gained attention in recent years. Obesity and overweight has become a major public health concern, and its incidence rising at an alarming rate. Obesity is one of the major types of metabolic syndrome, resulting in various types of problems such as hypertension, diabetes, dyslipidemia, and excess fat accumulation. The current searching was done by the keywords in main indexing systems including Scopus, PubMed/MEDLINE, the search engine of Google Scholar, and Institute for Scientific Web of Science. The keywords were traditional medicine, health benefits, pharmaceutical science, pomegranate, punicalin, punicalagin, and ellagitannins. Google Scholar was searched manually for possible missing manuscripts, and there was no language restriction in the search. This review was carried out to highlight the importance of medicinal plants which are common in traditional medicinal sciences of different countries, especially Asia to prevent and treatment of obesity and overweight during the global pandemic and the post-COVID-19 era.

1. Introduction

Traditional medicine is used not only for treatment but also for prevention [1,2,3,4,5,6], which is as old as mankind itself [7]. Medicinal plants which have been used in traditional medicinal science have relatively minimum or no negative impacts and toxicity, and their applications have been increased because of accessibility, affordability, easy availability, and acceptable efficacy [8,9,10]. Obesity is the main and primary reason of various metabolic ailments according to the epidemiological evidence, such as hypertension, diabetes, cardiovascular complications [11,12], asthma, arthritis, non-alcoholic fatty liver, degenerative disease, etc. [13]. Obesity is a serious health issue causing numerous health impairments, such as musculoskeletal disorders, cardiovascular diseases, type-2 diabetes, and different types of cancer [14]. Vulnerability to COVID-19 increases due to obesity, because both diseases share common inflammatory/metabolic pathways [15]. Notable changes in diet, sleep, physical activity, and mental health were reported by participants during the lockdown [16]. The Centers for Disease Control and Prevention list severe obesity (body mass index of ≥40 kg/m2) as a risk parameter for severe illness from COVID-19 [17]. During the pandemic, promotion of physical activity and prevention of obesity are as important as physical isolation of severely obese individuals [18]. Physical activity, maternal prenatal diet, psychological stress, sleep quality, and oxidative stress promoting disease conditions are important modulators of obesity and oxidative stress [19,20]. Obesity in individuals with COVID-19 independently has a relationship with increased risk for ICU admission and intubation [21,22]. Fat accumulation is caused by low levels of daily exercise and excessive food intake, which may influence fatty acid synthesis and betaoxidation. A linear dose–response link between body mass index and COVID-19 severity and mortality has been suggested, and obesity (body mass index ≥ 30 kg/m2) may cause a significantly increase in risk of critical COVID-19 and in-hospital mortality of COVID-19 [23,24]. Natural sources provide an advantageous basis for development of novel anti-obesity products [25]. Food supplements and functional foods containing fruit extracts, vegetables, and herbal products are also gaining increased attention [26] as an important natural alternative to prevent metabolic syndrome and obesity [27,28]. This review article is aimed to introduce and survey the most important medicinal plants on the basis of different traditional medicinal sciences containing chemical constituents with anti-obesity activities for treatment and prevention of obesity and related disease during the pandemic, quarantine, and the post-COVID-19 era.

2. Materials and Methods

The manuscript covers review articles, randomized control experiments, analytical studies and observations, which have been gathered from different sources such as Google Scholar, Scopus, Science Direct and PubMed. A review of the literature was carried out using the keywords COVID-19, obesity, natural products, medicinal plants, overweight, body mass index, and global pandemic.

3. Obesity

It has been reported that nutritional management may have obvious effects on the risk for chronic and obesity diseases [29,30]. The measurement tool which is mainly used in the clinical studies to identify obesity and overweight is called body mass index (BMI), which classifies people as normal weight (18.5–24.9 kg/m2), overweight (25–29.9 kg/m2), and obese (≥30 kg/m2) [31,32,33]. Obesity affects mortality, morbidity, and quality of life [34]; moreover, both overweight and obesity have increased attention in different societies, especially in urbanized populations [35,36]. Obesity shows harmful impacts and is believed to contribute to the increasing cardiovascular mortality [37]. Being overweight or obese is a risk factor for various costly comorbidities, such as type 2 diabetes (T2D), sleep apnea, chronic pain, cardiovascular disease, and certain cancers [38,39]. The frequency of obesity and overweight among children is increasing in the European region [40] and Canada [41], and women are presently more likely than before to suffer from obesity problems [42]. Impaired glucose tolerance of obese children is believed to exhibiting emerging oxidative stress levels [43]; obesity of childhood is a big concern as it is a principal predictor of obesity in adulthood [44]. The link between the presence of risk factors for different diseases and obesity in childhood with the early development of those diseases has been proven [45]. Prevention as well as treatment of obesity is important for children [46]. Obesity is linked with consequences of pneumonia, a protective impact coined the obesity paradox [47]. Obesity rates have dramatically risen among patients needing rehabilitation following join arthroplasty, injury, stroke, or an acute medical event [48]. Obesity may increase the risk of cardiovascular disease [49], and it is a risk factor for asthma [50].Obesity influences female reproduction by disturbing the hormone metabolism, the follicular environment, and body metabolism [51]. There is also a connection between obesity and cognitive reactivity, as well as depression [52]. An important risk factor for obesity is sleep duration [53]. The relationship between periodontal risk indicators and obesity in adolescents which may lead to oral morbidity has been found [54]. The important symptoms of metabolically healthy or unhealthy parameters are waist circumference, low-grade inflammation, HDL cholesterol, blood pressure, and diabetes [55].

4. Obesity and COVID-19

The COVID-19 pandemic, caused by the SARS-CoV-2 virus, has spread quickly from Wuhan, China to all parts of the world. A significant increase rate of obesity was found during the era of the COVID-19 epidemic [56,57], and also among obese patients with obstructive sleep apnea (OSA), an increased prevalence of COVID-19 was reported [58]. COVID-19 may lead to economic shutdown, confinement, and direct impacts, and consequently obesity development and eventually increased susceptibility to COVID-19 [59,60,61]. The worse outcomes of obese patients with COVID-19 consist of need for mechanical ventilation which is the consequence of respiratory failure and increase number of mortalities [62]. Body mass index, weight, and overweight escalated among children during the COVID-19 lockdown, influencing disadvantaged subpopulations, and related methods are required to decrease the impacts of the COVID-19 lockdown on childhood obesity and unhealthy weight gain [63]. The higher body mass index (BMI) in COVID-19 patients was reported, which is an important parameter that should always be evaluated for treatment and management of COVID-19 patients [64]. Obesity significantly increases the risk for hospitalization, demanding intensive care unit (ICU) admission, needing invasive mechanical ventilation (IMV) support, and risk for death among patients with COVID-19 [65,66,67]. Reports from the USA and Europe revealed remarkable morbidity and mortality in obese individuals with COVID-19 [68]. Obesity may have a noticeable role in the high incidence of mortality in black and Hispanic populations [69]. Recognition of populations with high fatality in COVID-19 gives clear insight to manage the pandemic by health services [70]. A need for policies is suggested which may support healthy lifestyle behaviors among low-income children during the pandemic [71]. Obesity may contribute to the increased risk and poorer prognosis of COVID-19 [72]. Obesity has a relationship with an immune state favorable to a cytokine storm, and serum concentrations of tumor necrosis factor alpha (TNF-α) and interleukin (IL)-17A are more increased in patients with obesity and COVID-19, and finally, they have a greater possibility of developing acute respiratory distress syndrome (ARDS) and death [73]. Biopsychosocial parameters which may influence COVID-19 pandemic-related obesity in children are (a) biological parameters including acute stress effects, altered leptin and adiponectin, chronic inflammation, intestinal dysbiosis, dysregulated immunological response, and stress increased inflammation and immune response; (b) social parameters such as household size, food insecurity, increased caloric consumption, increased screen time, lower educational achievement, parental factors, social distancing, sedentary lifestyle, socioeconomic status, and unhealthy diet; (c) psychological parameters like boredom, anxiety, food attitudes, food behavior, irritability, lack of self-regulation, maternal mental health loneliness, sleep disorders, and mental distress [74]. Reduced antithrombin is connected to mortality in COVID-19, and it has a relationship with obesity in COVID-19 [75,76,77]. Obesity and type 2 diabetes may have a role in the severity of disease and even mortality [78]. Subjects with obesity influenced by COVID-19 need longer hospitalization and much more time to clear from COVID-19 shedding compared to patients without obesity [79]. Inappropriate immune response is generated by obesity-induced chronic inflammatory status [80]. Isolation and quarantine can induce anger, anxiety, depression, and stress which are associated with unhealthy lifestyle leading to obesity. Higher BMI is connected with lower diet quality and levels of physical activity and a greater reported frequency of overeating during the lockdown [81]. Obesity could alter severe COVID-19 disease to younger ages [82]. Higher body mass index (BMI) and obesity are the major leading comorbidities to boost the risk of COVID-19 severity [83,84]. Obesity (BMI > 30 kg/m2) was connected with a significantly increased risk of critical COVID-19 and mortality [85]. Excess body weight multipliesthe risk of serious events connected to COVID-19 by 5.6 [86]. Dose–response meta-analysis indicates an increase of 5% risk for poor outcomes for every 5 kg/mg2 increase in body mass index [87]. There is a linear increase in risk of severe COVID-19 at a BMI of more than 23 kg/m2, leading to admission to hospital and death, and also a linear increase in admission to an ICU across the whole BMI range was reported [88]. Obese patients needed longer hospital stay and more days of mechanical ventilation compared to non-obese patients [89].

5. Medicinal Plants and Obesity

The increasing risk of obesity is associated with unhealthy plant foods [90,91], and consumption of healthy plant foods may be appropriate to decrease inflammation factors like TGF-β and hs-CRP [92], and plants which may show great potency to prevent obesity may have significant potential to inhibit pancreatice lipase [93]. The negative relationship between water hardness in water purification plants and the percentage of obese people has been reported [94], while the effective intervention within a complex system approach to manage obesity may be formed by plant secondary metabolites [95]. Phytochemicals with anti-obesity impacts are phenolic acids (o-Coumaric acid, Caffeic acid, Chlorogenic acid), Curcuminoids (Curcumin), Lignans (Podophyllotoxin), Flavonols (Quercetin), Isoflavonoids (Genistein, Daidzein, Glycitein), Flavones (Apigenin, Luteolin, Tangeritin), Flavans-3-ol (Catechin), Phytosterols (Diosgenin, Brassicasterol, β-Sitosterol, Campesterol), Anthocyanins (Cyanidin, Delphinidin, Malvidin, Pelargonidin, Peonidin), and Alkaloids (Caffeine, Capsaicin) [96]. Wheat (Triticum aestivum) sprout (TAEE) meaningfully reduced serum total cholesterol (TC), body weight, and low-density lipoprotein cholesterol levels in highfatdiet (HFD)-fed mice, and decreased lipid accumulation in epididymal white adipose tissue (EWAT) and liver. TAEE-treated mice indicated significant reduction in peroxisome proliferator-activated receptor γ (PPARγ) and fatty acid synthase expression in EWAT, which proved its efficacy for therapy for obesity and related diseases [97]. It has been found that luteolin, which is a Chinese herb, may be putative Furin inhibitor with tremendous benefits against Dengue Fever, and application of Chinese herbals and ion channel inhibitors can restrict the endothelial penetration of SARS-CoV-2 [98]. Natural phenolic compounds can be beneficially applied as food or fortified foods to control obesity [99].
Wild edible species from Southern Italy such as Daucus carota L., followed by Bellis perennis L. and Asparagusofficinalis L. showed inhibitory activity on NO production and protein denaturation, and D. carota indicated the best lipase inhibitory potential, which represented them as the appropriate candidates and the potential therapeutic agents in the treatment of obesity and inflammatory disorders [100]. Rosmarinus officinalis and Prunus avium contain the highest amounts of gallic acid, quercetin, ferulic acid, and epigallocatechin gallate which can be utilized against obesity in Turkish folk medicine [101]. The impacts of gut microbiota on obesity have been discovered in most animal and some human trials, and certain strains of Bacteroidetes, Firmicutes, and Lactobacillus have anti-obesity activities [102]. Apple, berries, chili, grapes, turmeric, sorghum, soy, and barley show anti-obesity efficacy via improving the diversity of gut microbiota, down-regulating obesogenic gut microbiota, and up-regulating anti-obesity gut microbiota [102]. Plant-based bio-active and fiber-enriched diets help in the alleviation of obesity and related diseases [103]. The most famous traditional medicinal plants in India for treatment of diabetes mellitus ailments, and potent candidates and substitutes to synthetic drugs in obese models are Acalypha indica, Pergulari ademia, and Tinospora cardifolia [104]. Folk herbal medicines for obesity are Zingiber officinale Roscoe, Carica papaya L., Hibiscussabdariffa L., Bauhinia variegata L., Foeniculum vulgare Mill., Caralluma tuberculata N.E.BR., Prunus avium (L.), Citrus medica L., Senna alexandrina Mill., Rosmarinus officinalis L. [105,106], Fucus vesiculosus, and Citrus aurantium [107]. In South Africa, three medicinal plants, namely Curtisia dentate, Cissaempelos capensis, and Schotia latifolia were repeatedly highlighted by the traditional healers and the local dwellers to have weight-reducing properties [108]. In Brazil, some of the most important species with great effective anti-obesity properties are Annona muricata L. and Hancorniaspeciosa Gomes, Baccharis trimera (Less.) DC, Camellia sinensis (L.) Kuntze, and Hibiscus sabdariffa L., showing some beneficial activity against obesity [109]. A natural pigment that mainly exists in the mature fruit of tomatoes is called lycopene, which effectively contributes to protecting against obesity and diabetes in animal studies [110]. Wines and grape berries are reported as the principal sources of stilbenes in human nutrition, which can alleviate the adverse effect of obesity by regulating various pathways [111]. By-products of guava and acerola can be applied as a sustainable alternative in the treatment of obesity [112]. Rosmarinic acid prevents excessive lipid accumulation and inflammation in human adipocytes, which shows its great potency for treatment of obesity-related inflammation [113]. Garlic extract in combination with ginger can prevent obesity in rats, and improved antioxidant enzyme’s activity [114]. A bioactive compound in celery (Apium graveolens) is 3-N-butylphthalide (NBP), which blocks the inflammatory response and regulates fat browning in HFD-induced obese mice, which indicates its effectiveness in combating obesity and its related metabolic disorders [115]. Various natural agents have been discovered for their obesity treatment potential such as Garcinia cambogia, apple cider, green tea, Panax ginseng, chia seeds, etc. [116]. Withania somnifera Dunal presented blood glucose-lowering and diuretic impacts in humans, comparable to daonil [117]. Acia (Euterpe oleracea Mart.) fruit anthocyanins decreased high-fat diet-induced obesity and hepatic steatosis, and increased high-fat diet-induced insulin resistance [118]. One of the promising anti-obesity agents is coffee fruit and oneof the traditional Chinese medicines is Caffeic acid [119]. Berberine is found to have an effective influence on gene regulation for the absorption of cholesterol at a daily dose of 300 mg in humans [120]. A classic traditional Chinese herb medicine which is called Wu-Mei-Wan can prevent obesity and its underlying mechanisms through attenuating white adipose tissue and increasing brown adipose tissue function [121]. Isolated Tomatidine from the green fruits and leaves of some plants in the Solanaceae family may significantly suppress the expression of fatty acid synthase and transcription factors are involved in lipogenesis and improve the expression of adipose triglyceride lipase, promoting the sirtuin 1 (sirt1)/AMPK signaling pathway to boost lipolysis and β-oxidation in fatty liver cells [122]. Hibiscus (Hibiscus shizopetalous; Hibiscus subdariffa), Argel (Solenostemma argel), and Caralluma (Caralluma quadrangular; Caralluma tuberculata) are famous traditional medicinal plants, and Argel indicated the highest inhibitory activity against lipase, α-amylase, and α-glucosidase enzymes, and the strongest lipase inhibitory activity was reported for the pregnane glycoside, and stemmoside C [123]. Roselle (Hibiscus sabdariffa) showed adipogenesis, reduced oxidative stress and systemic inflammation, and increased obesity-induced insulin resistance and insulin sensitivity [124]. EGCG, kaempferol, and quercetin have shown significant potent capability of anti-obesity activities [125,126]. Grape seed flour (GSF) prevented diet-induced obesity in C57BL/6J mice [127]. Procyanidins are a group of flavonoids, showing anti-obesity effects and increased metabolic flexibility and energy expenditure [128]. Crataegus pubescens and Ocimum sanctum ameliorated obesity and hyperglycemia in obese rats, reduced adipocyte hypertrophy, and can be introduced as ingredients for the elaboration of functional beverages [129]. The saprophytic fungus belongs to the family polyporaceae, Poria cocos, and synthesized Poria cocos gold nanoparticles caused the distinctions of influential nanoparticles which proved its ability as the potent anti-obesity drug [130]. Hussein et al. [131] reported that green coffee bean aqueous extracts and Spirulina platensis reduced liver weight, the final body weight, and the serum levels of alanine aminotransferase, aspartate aminotransferase, and alkaline phosphate, and they can be potentially considered as anti-obesity substances. Cercato et al. [132] also reported that the flavonoids quercetin, naringenin, genistein, epigallocatechin gallate, apigenin, and cyaniding 3-glucoside have potential in complementary therapy against obesity. Some of the most important plants, herbs, and fruits with anti-obesity characteristics are shown in Table 1.
Two famous traditional Chinese medicines, namely Rheum palmatum L. (Chinese rhubarb) (Polygonaceae), and Prunella vulgaris L. (the common self-heal) (Labiatae) are rich sources of anti-lipase compounds which can be considered as natural sources for crude anti-obesity drugs [191]. Consumption of bitter almond gum may decrease body mass index and body weight, alleviate hyperinsulinemia in hyper lipidemic subjects, reduce serum triglycerides, and can be applied for the management of body weight in fruit juices [192]. Saponins are the best candidate as appetite suppressants and pancreatice lipase, and may manage fattlier liver formation and body weight [193]. Acacia arabica, Agrimonia eupatoria, Aegle marmelose, Allium sativum, Allium cepa, Azadirachta indica, Aloe vera, Beta vulgaris, Benincasa hispida, Caesalpinia bonducella, Coccinia indica, Citrullus colocynthis, Eucalyptus globules, Ficus bengalenesis, Gymnema sylvestre, Hibiscus rosasinesis, Ipomoea batatas, Jatropha curcus, Mangifera indica, Morus alba, Momordica charantia, Mucuna pruriens, Ocimum sanctum, Punica granatum, Pterocarpus marsupium, Syzigium cumini, Tinospora cordifolia, and Trigonella foenum graecum are the most notable plants with antidiabetic potential in Indian traditional medicine with anti-obesity characteristics [194]. Dietary fruit intake was positively associated with P53 and PTEN gene expression in visceral and subcutaneous adipose tissues (SAT) obese participants [195]. Dandelion (Taraxacum officinale (L.)) and luteolin may improve HDLcholesterol in obese rats fed a normal-fat diet [196]. The natural compound celastrol, a pentacyclic triterpene isolated from the roots of Tripterygium wilfordi (thunder god vine) plant shows different bioactivities including and antidiabetic and anti-obese impacts [197]. The impacts of taking the combination of Zataria multiflora Boiss. (Zm) and oxymel may reduce insulin resistance and hip and wasit circumferences in overweight patients [198]. Dietary ginger controls body weight gain by inducing browning of white adipose tissue (WAT) and remodeling whole-body energy metabolism [199]. Coleus forskohlii extract and Garcinia indica extract increase energy expenditure through promotion of fatty acid β-oxidation, and attenuating the Firmicutes/Bacteroidetes to attenuate obesity [200]. African walnuts reduced lipid accumulation in adipose and ectopic tissues in MSG-obese rats [201]. Red maple (Acer rubrum) leaves extract decreased diet-induced obesity without influencing energy intake, and its impacts are partially because of the modulation of gut flora [202]. Indian brown algae (Padina tetrastromatica) is the major component for obesity management [203]. Fagara tessmannii is a shrub of the African rainforests in the South-West, Center, South, and East provinces in Cameroon, regulating the loss of ectopic fat and other fatty tissues, the energy expenditure and the renovascular decompression, the sensitivity of the peripheric tissues to insulin, and promotes ion movement which prevents hypertension [204]. The ethanolic extracts of Actaea racemosa L. have been proven as the best candidate for treatment of obesity and related diseases for further studies [205]. Garcinia indica extract standardized for 20% Garcinol inhibited adipogenesisin vitro3T3-L1 cells, decreased endoplasmic reticulum stress in adipocytes, alleviated visceral fat weight, and regulated obesity by acting on the AMPK-ER stress axis [206]. Chrysin is a flavonoid found in plant extracts from Passiflora species, alleviated the body weight of rats, decreased calorie intake of rats, and the hypertrophy of adipocytes [207]. Application of licorice extract with a low-calorie diet can efficiently increase the lipid profile in overweight and obese subjects [208]. The anti-obesity activities of the essential oils of hedgenettles (Stachys inflata, S. lavandulifolia, and S. byzantina) have been reported [209]. Huang-Qi San (HQS), the traditional Chinese medicine, can ameliorate hyperlipidemia with obesity [210]. Meratrim formulation, yerba mate, spinach, brown beans, sorghum, psyllium, and rye showed evidence for suppressing appetite [211]. A combination of resistance training with 500 mL/g of glycyrrhizic acid supplement may be appropriate to decrease body weight and body fat percentage [212]. In Mexico, one of the common herbal products used for weightloss is based on soybean, green tea, and aceitilla [213]. Methylxanthines are nutraceuticals mainly present in tea, coffee, and chocolate, and the most well-known methylxanthines are theophylline, caffeine, and theophylline, which can contribute to weight loss, fat depletion, and inhibit adipogenesis and stimulate lipolysis [214]. Berberine consumption significantly decreased body mass index and alleviates waist circumference (WC) [215]. Cooked mung bean may prevent obesity in mice fed with a highfatdiet and regulates lipid metabolic disorders [216]. Garlic oil had a significant anti-obesity influence on obese rats by protecting the liver from damage and regulating the body weight [217]. A significant decrease of intracellular lipid accumulation in 3T#-L1 pre-adipocyres, improvement of plasma lipid profile in HFD-induced mice, and regulation of body weight gain has been reported due to application of bound phenolics isolated from lotus seeds [218]. The ethanolic extract of Cuscuta reflexa suppressed the development of obesity in HFD-induced obesity [219]. Hunteria umbellate (K. Schum.) Hallier f. indicated both antihyperlipidaemic and anti-obesity effects which may partly be mediated via inhibition of intestinal lipid absorption and de novo biosynthesis of cholesterol [220]. Lagenaria siceraria (fruit) and Commiphora mukul (gum resin) can regulate the high fat diet-induced obesity [221]. Erigeron annuus L., and Borago officinalis L. significantly attenuated improved in body weight gain, lipid accumulation, and adipocyte size, a unique adipokine known to promote the breakdown of fat/lipids [222]. Blueberry had potential health benefits in ameliorating the development of obesity and its related comorbidities, such as chronic inflammation and type 2 diabetes [223]. Morin (3,5,7,2,4-pentahydroxy flavones) is found in some natural products such as almond (Prunus dutcis), figs (chlorophora tinctoria), guava leaves (psidium guajava), and some other Maraceae family plants [224,225]; morin treatment produced dose-dependent improvement in lipid profile and vascular endothelium protection, thus rationalizing its medicinal use in cardiovascular-related endothelial disorders, and dyslipidemia diseases [226]. In Kampo, Japanese traditional medicine, two formulas, this bofutsushosan (the composition of scutellaria root, platycodon root, glycyrrhiza, atractylodes rhizome, gypsum, rhubarb, schizonepeta spike, peony root, gardenia fruit, Japanese angelica root, cnidium rhizome, menthe herb, saposhnikovia root, forsythia fruit, Ephedra herb, ginger) and boiogito (the composition of astragalus root, sinomenium stem, jujube, atractylodes lancea rhizome, glycyrrhiza, and ginger) are supported by the national health insurance in Japan for treatment of obesity [227]. In Bangladesh, the most important medicinal plants with anti-obesity characteristics are Achyranthes aspera Linn, Aegle marmelos Linn, Alliun sativum Linn, Acorus calamus Linn, Allium cepa Linn, Bombax ceiba L., Moringa oliefera, Hibiscus sabdariffa L., Impomoea batatas L., Punica granatum L., Citrus limon L., and Zingiber officinale Roscoe [228]. One of the screening methods applied in the discovery of anti-obesity drugs is to search for potent lipase inhibitors from plant extracts; the major plant extracts that inhibit porcine pancreatic lipase (PPL) are Platycodon grandiflorum A. De Candolle, Aconitumcarmichaeli Debeaux, Chaenomeles sinensis (Thouin) Koehne, Cannabis sativa Linne, Actinidachinensis, Tribulus terrestris, Luffa cylindrical Roemer, Lilium brownii var. viridulun Baker, Pueraria thunbergiana Bentham, Crataegus pinnatifidaBunge var. typical Schneider, Nardostachys chinensis Batal, Zizyphus jujuba Miller Var. inermis Rehder, Akebia quinata Decaisne, Quisqualis indica Linne, Rehmannia glutinosa, Loranthus parasticus Merr., Schizandra chinensis Baillon, Lonicera japonica Thunberg, Dipsacus asperoides C. Y. Cheng et T. M. Ai, Perilla sikokiana Nakai, Morinda officinalis How, Prunus nakaii Leveille, Melia azedarachLinne var. japonica Makino, Poria cocos wolf, Gastrodia Blume, Bletilla striata (Thunberg) Reichenbach fil., Oldenlandia diffusa (Willd.) Roxburgh, Gentiana scabra, Cuscuta chinensis Lamark, Tetrapanax papyriferus K. Koch, Fritillaria thunbergii Miquel, Astragalus membranaceus Bunge, Patrinia villosa Jussieu, Scutellaria baicalenis Georgi, Phellodendron amurense Ruprecht, Rubus coreanus Miquel, Drynaria fortunei Smith, Eriobotrya japonica Lindley, Amomum tsao-ko Crevost et Lemaire, Cornus officinalis Siebold et Zuccarini, Forsythia koreana Nakai, Ulmus darvidian for. Suberose, Polygonum aviculare Linne, and Geraniumthunbergia. Siebold et Zuccarini [229] and Bahmani et al. [230] reported that a combination of ephedrine and caffeine, capsicin from red pepper and chili, yohimbine, guar gum extracted from the plant Cyamposistetragonolobus, glucomannan extracted from the root of Amorphophalluskonjac, Garcinia cambogia, the active ingredient from Pausinystaliajohimbe, and Hoodia gordonni have functional roles in reducing weight, improving energy, and inhibiting fat absorption, with no negative impacts in the treatment groups. The most principal medicinal plants and herbs in traditional Chinese medicine in the treatment of obesity are celastrol (Tripterygium wilfordii), Berberine (Coptis chinensis), Capsaicin (Chili pepper), Resveratrol (Polygonum cuspidatum), Chrysophanic acid/Rhein (Rhubarb), Fenugreek (Trigonella foenumgraecum L.), Curcumin (Turmeric), Radix astragali, Tea catechins GTpolyphenols (Green tea), Xiexin decoction, white tiger plus ginseng decoction, and Chaihu Shugan powder through increased leptin sensitivity, blocking fat production and accumulation, improving lipid decomposition, regulating the level of adiponectin, etc. [231,232]. Polyphenol, quercetin, caffeic acid, hydroxyflavin, and hesperetin are active constituents of mulberry leaf with possible anti-obesity impacts [233]. Capsicoside G is the main component of pepper which can block adipogenesis through activation of adenosine monophosphate-activated protein [234]. The principal active component of cocoa with anti-obesity activity is polyphenols which decrease lipids in the liver; genes in lipid catabolism, primarily in fatty acid oxidation, was up-regulated, whereas genes in lipid synthesis pathways were down-regulated, regulating obesity-induced steatosis markers [235]. Coumaric acid and ferulic acid are the main anti-obesity components of barley which may inhibit adipocyte differentiation, dysregulate lipid profiles, and prevent body weight gain [236]. Black soybean contains anthocyanin, which can markedly alleviate fat accumulation, regulate the expression of lipogenesis genes (acetyl-CoA carboxylase), and improve the levels of lipolysis proteins (hormone-sensitive lipase, lipoprotein lipase, and adenosine monophosphate-activated protein kinase) in mesenteric fat [237]. The active anti-obesity of red chili pepper, capsinoids, can suppress diet-induced obesity via uncoupling protein 1-dependent mechanism [238]. The most notable active components of garlic are S-allyl-l-cysteine sulphoxide, S-allyl cysteine which can alleviate relative masses of liver and fat tissues, hepatic oxidative stress levels, serum triacylglyceride levels, and improve fecal lipid contents in high fat diet rats, up-regulated adenosine monophosphate-activated protein kinase, adipose triacyglyceride lipase, Sirtuin 1, hormone-sensitive lipase, palmitoyltransderase 1, Acyl-CoA oxidase, whereas it down-regulated cluster of differentiation 36 [239]. Crocin of saffron can significantly alleviate total cholesterol and plasma levels of triacylglycerol [240]. Tiliroside of raspberry and strawberry may inhibit obesity-induced hepatic inflammation and muscular triglyceride accumulation [241]. Polyphenols are the active anti-obesity constituent of coffee which can regulate postprandial hyperglycaemia and hyperlipidemia, suppress lipogenesis by down-regulating sterol regulatory element-binding proteins acetyl-CoA carboxylase-1 and -2, stearoyl-CoA desaturase-1 and pyruvate dehydrogenase kinase-4 in the liver [242]. Anthocyanidins of bilberry can block adipocyte differentiation through impacting the gene expressions of the insulin pathway, reduced PPAR, sterol regulatory element-binding protein 1c and tyrosine residues of insulin receptor substrate 1 phosphorylation [243].

6. Conclusions

Many chronic diseases, asthma, etc. are associated with obesity and overweight, and the dramatic rise in obesity prevalence worldwide has become a real health concern. As obesity is a risk factor for many diseases, obesity itself may worsen the outcomes of COVID-19, which may require intensive care. Obesity/overweight is linked with increase COVID-19 mortality, and the monotonic relationship between COVID-19 infection and body mass index and risks of hospitalization have been reported. The main reasons which may contribute to obesity are reduced physical activity, excess intake of calorie loaded food, depression, pharmaceutical concomitant, food obsession, personality traits, genital/hereditary predisposition, economic growth, and lifestyle modifications. Medicinal plants, particularly edible plants have long been used as traditional knowledge to treat and prevent obesity, especially in Asian countries, because various bioactive compounds from both herbs and fruits have been found useful for anti-obesity drug discovery and development processes. Obesity is an important problem for normal growth in children, with both primary and secondary health risks such as high blood pressure, insulin resistance, hypertension, cardiovascular diseases, and different cancers. The different mechanisms which medicinal plants may affect weight loss consist of increasing levels of leptin, hypolipidemic and hypoglycemic effects, reducing fat absorption, influencing fat metabolism, enhancing metabolism, decreasing appetite, and preventing carbohydrate intake. The most important medicinal plants which are common in traditional medicinal sciences of different countries due to their anti-obesity activities are Acosmium dasycarpum (Vog.) Yakovlev, Allium cepa L., Aloe barbadensis Miller, Amorphophallus konjac K. Koch., Artemisiasphaerocephala Krasch, Betula utilis, Bupleuri Radix, Butea monosperma, Caralluma fimbriata, Corchorus olitorius L., Cuminum cyminum L., Carum carvi L., Cyclopia spp., Cynara scolymus L., Cynometra cauliflora Linn., Cynomorium songaricum Rupr., Echium angustifolium Mill., Echium angustifolium Mill., Garcinia cambogia, Gnidia glauca (Fres.) Glig, Ganodermalucidum sensu strict, Gnidia glauca (Fres.) Glig., Hibiscus sabdariffa L., Ilex paraguariensis, Justicia carnea Lindl., Juniperus communis L., Ligustrum robustum Blume, Lobelia chinensis lour, Macrotyloma uniflorum (Lam.) Verdc., Mangifera indica Linn., Melissa officinalis L., Memecylon umbellatum Burm. f., Moringa oleifera Lam., Moringa peregrine (Forssk.) Fiori., Morus alba L., Nigella sativa L., Oroxylum indicum Kurz, Passiflora edulis Sims, Pilosocereusgounellei (F.A.C. Weber) Byles and G.D.Rowley, Piper nigrum L., Populus balsamifera L., Psidium guajava L., Raphanus sativus L., Salacia chinensis L., Salvia hispanica L., Salviaofficinalis L., Smilax china L., Smilax glabra Roxb., Solenostemma argel Hayne, TabebuiaavellanedaeLorentz ex Griseb, Tinospora cordifolia (Thunb.) Miers., Urtica dioica L., Vaccinium arctostaphylos L., and Withania somnifera (L.) Dunal.

Author Contributions

W.S.: writing—original draft preparation; M.H.S.: writing—original draft preparation; Q.C.: writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Natural Science Foundation of Beijing, China (Grant No.M21026). This research was also supported by the National Key R&D Program of China (Research grant 2019YFA0904700).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

T2D: type 2 diabetes; BMI: body mass index; ICU: intensive care unit; IMV: invasive mechanical ventilation; TNFα: tumor necrosis factor alpha; IL: interleukin; ARDS: acute respiratory distress syndrome; TC: total cholesterol; HFD: high-fatdiet; EWAT: epididymal white adipose tissue; PPARγ: peroxisome proliferator-activated receptor γ; NBP: 3-N-butylphthalide; Sirt1: Sirtuin 1; GSF: grape seed flour; BupE: Bupleuri Radix extract; WFA: Withaferin A; MAPK: mitogen-activated protein kinase; AMPK: AMP-activated protein kinase; HQS: Huang-Qi San.

References

  1. Shahrajabian, M.H.; Sun, W.; Cheng, Q. Exploring Artemisia annua L., artemisinin and its derivatives, from traditional Chinese wonder medicinal science. Not. Bot. Horti Agrobot. 2020, 48, 1719–1741. [Google Scholar] [CrossRef]
  2. Shahrajabian, M.H.; Sun, W.; Cheng, Q. Chemical components and pharmacological benefits of basil (Ocimum basilicum): A review. Int. J. Food Prop. 2020, 23, 1961–1970. [Google Scholar] [CrossRef]
  3. Shahrajabian, M.H.; Sun, W.; Cheng, Q. Traditional herbal medicine for the prevention and treatment of cold and flu in the autumn of 2020, overlapped with COVID-19. Nat. Prod. Commun. 2020, 15, 1–10. [Google Scholar] [CrossRef]
  4. Shahrajabian, M.H.; Sun, W.; Soleymani, A.; Cheng, Q. Traditional herbal medicines to overcome stress, anxiety and improve mental health in outbreaks of human coronaviruses. Phytother. Res. 2020, 2020, 1–11. [Google Scholar] [CrossRef]
  5. Shahrajabian, M.H.; Sun, W.; Shen, H.; Cheng, Q. Chinese herbal medicine for SARS and SARS-CoV-2 treatment and prevention, encouraging using herbal medicine for COVID-19 outbreak. Acta Agric. Scand. Sec. B Soil Plant. Sci. 2020, 70, 437–443. [Google Scholar] [CrossRef]
  6. Shahrajabian, M.H.; Sun, W.; Cheng, Q. Product of natural evolution (SARS, MERS, and SARS-CoV-2); deadly diseases from SARS to SARS-CoV-2. Hum. Vaccines Immunother. 2020, 17, 62–83. [Google Scholar] [CrossRef] [PubMed]
  7. Marmitt, D.J.; Shahrajabian, M.H. Plant species used in Brazil and Asia regions toxic properties. Phytother. Res. 2021, 2021, 1–24. [Google Scholar] [CrossRef]
  8. Shahrajabian, M.H.; Sun, W.; Cheng, Q. Asafoetida, a natural medicine for a future. Curr. Nutr. Food Sci. 2021, 17, 1–10. [Google Scholar] [CrossRef]
  9. Shahrajabian, M.H.; Sun, W.; Cheng, Q. Improving health benefits with considering traditional and modern health benefits of Peganum harmala. Clin. Phytosci. 2021, 7, 1–10. [Google Scholar] [CrossRef]
  10. Shahrajabian, M.H.; Sun, W.; Cheng, Q. Different methods for molecular and rapid detection of human novel coronavirus. Curr. Pharm. Des. 2021. [Google Scholar] [CrossRef]
  11. Lavie, C.J.; Milani, R.V. Obesity and cardiovascular disease: The Hippocrates paradox? J. Am. Coll. Cardiol. 2003, 42, 677–679. [Google Scholar] [CrossRef] [Green Version]
  12. Poirier, P.; Giles, T.D.; Bray, G.A.; Hong, Y.; Stern, J.S.; Pi-Sunyer, F.X.; Eckel, R.H. Obesity and cardiovascular disease pathophysiology, evaluation, and effect on weight loss. Arterioscl. Thromb. Vas Biol. 2006, 26, 968–976. [Google Scholar] [CrossRef] [PubMed]
  13. Tilg, H.; Moschen, A.R. Adipocytokines: Mediators linking adipose tissue, inflammation and immunity. Nat. Rev. Immunol. 2006, 6, 772–783. [Google Scholar] [CrossRef] [PubMed]
  14. Wlodarczyk, M.; Slizewska, K. Obesity as the 21st century’s major disease: The role of probiotics and prebiotics in prevention and treatment. Food Biosci. 2021, 42, 101115. [Google Scholar] [CrossRef]
  15. Michalakis, K.; Ilias, I. SARS-CoV-2 infection and obesity: Common inflammatory and metabolic aspects. Diabetes Metab. Syndr. Clin. Res. Rev. 2020, 14, 469–471. [Google Scholar] [CrossRef]
  16. Brown, A.; Flint, S.W.; Kalea, A.Z.; O’Kane, M.; Williams, S.; Batterham, R.L. Negative impact of the first COVID-19 lockdown upon health-related behaviours and psychological wellbeing in people living with severe and complex obesity in the UK. EClinicalMedicine 2021, 34, 100796. [Google Scholar] [CrossRef] [PubMed]
  17. Zhu, Z.; Hasegawa, K.; Ma, B.; Fujiogi, M.; Camargo, C.A.; Liang, L. Association of obesity and its genetic predisposition with the risk of severe COVID-19: Analysis of population-based cohort data. Metab. Clin. Exp. 2020, 112, 154345. [Google Scholar] [CrossRef]
  18. Katsoulis, M.; Pasea, L.; Lai, A.G.; Dobson, R.J.B.; Denaxas, S.; Hemingway, H.; Banerjee, A. Obesity during the COVID-19 pandemic: Both cause of high risk and potential effect of lockdown? A population-based electronic health record study. Public Health 2021, 191, 41–47. [Google Scholar] [CrossRef]
  19. Tobore, T.O. Towards a comprehensive theory of obesity and a healthy diet: The causal role of oxidative stress in food addiction and obesity. Behav. Brain Res. 2020, 384, 112560. [Google Scholar] [CrossRef] [PubMed]
  20. Al Heialy, S.; Hachim, M.Y.; Hachim, I.Y.; Naeem, K.B.; Hannawi, H.; Lakshmanan, J.; Al Salmi, I.; Hannawi, S. Combination of obesity and co-morbidities leads to unfavorable outcomes in COVID-19 patients. Saudi J. Biol. Sci. 2021, 28, 1445–1450. [Google Scholar] [CrossRef] [PubMed]
  21. Malik, V.S.; Ravindra, K.; Attri, S.V.; Bhadada, S.K.; Singh, M. Higher body mass index is an important risk factor in COVID-19 patients: A systematic review and meta-analysis. Environ. Sci. Pollut. Res. 2020, 27, 42115–42123. [Google Scholar] [CrossRef]
  22. Suresh, S.; Siddiqui, M.; Ghanimeh, M.A.; Jou, J.; Simmer, S.; Mendiratta, V.; Russell, S.; Al-Shammari, M.; Chatfield, A.; Alsheik, E.; et al. Association of obesity with illness severity in hospitalized patients with COVID-19: A retrospective cohort study. Obes. Res. Clin. Pract. 2021, 15, 172–176. [Google Scholar] [CrossRef] [PubMed]
  23. Cai, H.; Yang, L.; Lu, Y.; Zhang, S.; Ye, C.; Zhang, X.; Yu, G.; Gu, J.; Lian, J.; Hao, S.; et al. High body mass index is a significant risk factor for the progression and prognosis of imported COVID-19: A multicenter, retrospective cohort study. BMC Infect. Dis. 2021, 21, 147. [Google Scholar] [CrossRef] [PubMed]
  24. Du, Y.; Lv, Y.; Zha, W.; Zhou, N.; Hong, X. Association of body mass index (BMI) with critical COVID-19 and in-hospital mortality: A dose-response meta-analysis. Metabolism 2021, 117, 154373. [Google Scholar] [CrossRef] [PubMed]
  25. Karri, S.; Sharma, S.; Hatware, K.; Patil, K. Natural anti-obesity agents and their therapeutic role in management of obesity: A future trend perspective. Biomed. Pharmacother. 2019, 110, 224–238. [Google Scholar] [CrossRef] [PubMed]
  26. Sander, G.; Konig, A.; Wallner, M.; Weghuber, J. Functional foods-dietary or herbal products on obesity: Application of selected bioactive compounds to target lipid metabolism. Curr. Opin. Food Sci. 2020, 34, 9–20. [Google Scholar] [CrossRef]
  27. Engin, A.B.; Tsatsakis, A.M.; Tsoukalas, D.; Engin, A. Do flavanols-rich natural products relieve obesity-related insulin resistance? Food Chem. Toxicol. 2018, 112, 157–167. [Google Scholar] [CrossRef]
  28. Ngoc, L.P.; Man, H.-Y.; Besselink, H.; Cam, H.D.T.; Brouwer, A.; Burg, B.V.D. Identification of PPAR-activating compounds in herbal and edible plants and fungi from Vietnam. Ind. Crops Prod. 2019, 129, 195–200. [Google Scholar] [CrossRef]
  29. Tchang, B.G.; Tarazi, M.S.; Aras, M.; Shukla, A.P. An update on pharmacotherapeutic strategies for obesity. Expert Opin. Pharmacother. 2021, 22, 1305–1318. [Google Scholar] [CrossRef]
  30. Van der Merwe, M.-T. Obesity in women-a life cycle of medical risk. J. Endocrinol. Metab. Diabetes S. Afr. 2009, 14, 139–142. [Google Scholar] [CrossRef] [Green Version]
  31. Monteiro, M.P. Obesity vaccines. Hum. Vaccines Immunother. 2014, 10, 887–895. [Google Scholar] [CrossRef] [Green Version]
  32. Fruhbeck, G.; Catalan, V.; Rodriguez, A.; Gomez-Ambrosi, J. Adiponectin-leptin ratio: A promising index to estimate adipose tissue dysfunction. Relation with obesity-associated cardiometabolic risk. Adipocyte 2018, 7, 57–62. [Google Scholar] [CrossRef] [PubMed]
  33. Bishwajit, G.; Yaya, S. Overweight and obesity among under-five children in South Asia. Child. Adolesc. Obes. 2020, 3, 105–121. [Google Scholar] [CrossRef]
  34. Garvey, W.T. Phentermine and topiramate extended-release: A new treatment for obesity and its role in a complications-centric approach to obesity medical management. Expert Opin. Drug Saf. 2013, 12, 741–756. [Google Scholar] [CrossRef]
  35. Wang, Y.; Lobstein, T. Worldwide trends in childhood overweight and obesity. Int. J. Pediatr. Obes. 2006, 1, 11–25. [Google Scholar] [CrossRef] [PubMed]
  36. Bell, J.A.; Hamer, M. Healthy obesity as an intermediate state of risk: A critical review. Expert Rev. Endocrinol. Metab. 2016, 11, 403–413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Papagianni, M.; Tziomalos, K. Effect of obesity on the outcome of pneumonia. Expert Rev. Endocinol. Metab. 2017, 12, 315–320. [Google Scholar] [CrossRef] [PubMed]
  38. Wang, Y.; Lim, H. The global childhood obesity epidemic and the association between socio-economic status and childhood obesity. Int. Rev. Psychiatr. 2012, 24, 176–188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Li, Q.; Blume, S.W.; Huang, J.C.; Hammer, M.; Ganz, M.L. Prevalence and healthcare costs of obesity-related comorbidities: Evidence from an electronic medical records system in the United States. J. Med. Econ. 2015, 18, 1020–1028. [Google Scholar] [CrossRef]
  40. Jackson-Leach, R.; Lobstein, T. Estimated burden of paediatric obesity and co-morbidities in Europe. Part 1. The increase in the prevalence of child obesity in Europe is itself increasing. Int. J. Pediatr. Obes. 2006, 1, 26–32. [Google Scholar] [CrossRef] [PubMed]
  41. Shields, M.; Tremblay, M.S. Canadian childhood obesity estimates based on WHO, IOTF and CDC cut-points. Int. J. Pediatr. Obes. 2010, 5, 265–273. [Google Scholar] [CrossRef]
  42. Galliano, D.; Bellver, J. Female obesity: Short-and long-term consequences on the offspring. J. Gynaecol. Endocrinol. 2013, 29, 626–631. [Google Scholar] [CrossRef] [PubMed]
  43. Kloppenborg, J.T.; Fonvig, C.E.; Johannesen, J.; Bjerrum, P.J.; Poulsen, H.E.; Holm, J.-C. Urinary markers of nucleic acid oxidation in Danish overweight/obese children and youths. Free Radic. Res. 2016, 50, 691–697. [Google Scholar] [CrossRef] [PubMed]
  44. Chu, N.-F. Strategies for prevention and treatment of obesity among children in Taiwan. Res. Sports Med. 2010, 18, 37–48. [Google Scholar] [CrossRef] [PubMed]
  45. Lobstein, T.; Jackson-Leach, R. Estimated burden of paediatric obesity and co-morbidities in Europe. Part 2. Numbers of children with indicators of obesity-related disease. Int. J. Pediatr. Obes. 2006, 1, 33–41. [Google Scholar] [CrossRef]
  46. Nowicka, P.; Flodmark, C.-E. Family in pediatric obesity management: A literature review. Int. J. Pediatr. Obes. 2008, 3, 44–50. [Google Scholar] [CrossRef]
  47. Papagianni, M.; Tziomalos, K. Obesity in patients with HIV infection: Epidemiology, consequences and treatment options. Expert Rev. Endocinol. Metab. 2016, 11, 395–402. [Google Scholar] [CrossRef]
  48. Seida, J.C.; Sharma, A.M.; Johnson, J.A.; Forhan, M. Hospital rehabilitation for patients with obesity: A scoping review. Disabil. Rehabil. 2018, 40, 125–134. [Google Scholar] [CrossRef]
  49. Khan, U.I.; Rastogi, D.; Isasi, C.R.; Coupey, S.M. Independent and synergistic associations of asthma and obesity with systemic inflammation in adolescents. J. Asthma 2012, 49, 1044–1050. [Google Scholar] [CrossRef]
  50. Dixon, A.E.; Clerisme-Beaty, E.M.; Sugar, E.A.; Cohen, R.; Lang, J.E.; Brown, E.D.; Richter, J.E.; Irvin, C.G.; Mastronarde, J.G. Effects of obstructive sleep apnea and gastroesophageal reflux disease on asthma control in obesity. J. Asthma 2012, 48, 707–713. [Google Scholar] [CrossRef]
  51. Pantasri, T.; Norman, R.J. The effects of being overweight and obese on female reproduction: A review. Gynecol. Endocrinol. 2014, 30, 90–94. [Google Scholar] [CrossRef] [PubMed]
  52. Minkwitz, J.; Scheipl, F.; Carwright, L.; Campbell, I.C.; Chittka, T.; Thormann, J.; Hegerl, U.; Sander, C.; Himmerich, H. Why some obese people become depressed whilst others do not: Exploring links between cognitive reactivity, depression and obesity. Psychol. Health Med. 2019, 24, 362–373. [Google Scholar] [CrossRef] [PubMed]
  53. Teodorescu, M.; Polomis, D.A.; Gangnon, R.E.; Consens, F.B.; Chervin, R.D.; Teodorescu, M.C. Sleep duration, asthma and obesity. J. Asthma 2013, 50, 945–953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Modeer, T.; Blomberg, C.; Wondimu, B.; Lindberg, T.Y.; Marcus, C. Association between obesity and periodontal risk indicators in adolescents. Int. J. Pediatr. Obes. 2010, 6, e264–e270. [Google Scholar] [CrossRef]
  55. Berstein, L.M. Dark and light side of obesity: Mortality of metabolically healthy obese people. Expert Rev. Endocrinol. Metab. 2012, 7, 629–632. [Google Scholar] [CrossRef]
  56. Abbas, A.M.; Fathy, S.K.; Fawzy, A.T.; Salem, A.S.; Shawky, M.S. The mutual effects of COVID-19 and obesity. Obes. Med. 2020, 19, 100250. [Google Scholar] [CrossRef]
  57. McNeill, J.N.; Lau, E.S.; Paniagua, S.M.; Liu, E.E.; Wang, J.K.; Bassett, I.V.; Selvaggi, C.A.; Lubitz, S.A.; Foulkes, A.S.; Ho, J.E. The role of obesity in inflammatory markers in COVID-19 patients. Obes. Res. Clin. Pract. 2021, 15, 96–99. [Google Scholar] [CrossRef]
  58. Vaishali, K.; Gatty, A.; Srivastav, P.; Amin, R.R. Coping strategies for obese individuals with obstructive sleep apnea during COVID-19 pandemic: A narrative review. Obes. Med. 2021, 22, 100324. [Google Scholar] [CrossRef]
  59. Ghanemi, A.; Yoshioka, M.; St-Amand, J. Will an obesity pandemic replace the coronavirus disease-2019 (COVID-19) pandemic? Med. Hypotheses 2020, 144, 110042. [Google Scholar] [CrossRef]
  60. Lui, B.; Samuels, J.D.; White, R.S. Potential pathophysiology of COVID-19 in patients with obesity. Br. J. Anaesth. 2020, 125, e283–e284. [Google Scholar] [CrossRef]
  61. Pontiroli, A.; Sala, L.L.; Chiumello, D.; Folli, F. Is blood glucose or obesity responsible for the bad prognosis of COVID-19 in obesity-diabetes? Diabetes Res. Clin. Pract. 2020, 167, 108342. [Google Scholar] [CrossRef]
  62. Sanchis-Gomar, F.; Lavie, C.J.; Mehra, M.R.; Henry, B.M.; Lippi, G. Obesity and outcomes in COVID-19: When an epidemic and pandemic collide. Mayo Clin. Proc. 2020, 95, 1445–1453. [Google Scholar] [CrossRef]
  63. Mulugeta, W.; Hoque, L. Impact of the COVID-19 lockdown on weight status and associated factors for obesity among children in Massachusetts. Obes. Med. 2021, 22, 100325. [Google Scholar] [CrossRef]
  64. Soeroto, A.Y.; Soetedjo, N.N.; Purwiga, A.; Santoso, P.; Kulsum, I.D.; Suryadinata, H.; Ferdian, F. Effect of increased BMI and obesity on the outcome of COVID-19 adult patients: A systematic review and meta-analysis. Diabetes Metab. Syndr. Clin. Res. Rev. 2020, 14, 1897–1904. [Google Scholar] [CrossRef] [PubMed]
  65. Belancic, A.; Kresovic, A.; Racki, V. Potential pathophysiological mechanisms leading to increased COVID-19 susceptibility and severity in obesity. Obes. Med. 2020, 19, 100259. [Google Scholar] [CrossRef]
  66. Biscarini, S.; Colaneri, M.; Ludovisi, S.; Seminari, E.; Pieri, T.C.; Valsecchi, P.; Gallazzi, I.; Giusti, E.; Camma, C.; Zuccaro, V.; et al. The obesity paradox: Analysis from the Smatteo COVID-19 Registry (SMACORE) cohort. Nutr. Metab. Cardiovasc. Dis. 2020, 30, 1920–1925. [Google Scholar] [CrossRef]
  67. Huang, Y.; Lu, Y.; Huang, Y.-M.; Wang, M.; Ling, W.; Sui, Y.; Zhao, H.-L. Obesity in patients with COVID-19: A systematic review and meta-analysis. Metabolism 2020, 113, 154378. [Google Scholar] [CrossRef]
  68. Yadav, R.; Aggarwal, S.; Singh, A. SARS-CoV-2-host dynamics: Increased risk of adverse outcomes of COVID-19 in obesity. Diabetes Metab. Syndr. Clin. Res. Rev. 2020, 14, 1355–1360. [Google Scholar] [CrossRef] [PubMed]
  69. Chaar, M.E.; King, K.; Lima, A.G. Are black and Hispanic persons disproportionately affected by COVID-19 because of higher obesity rates? Surg. Obes. Relat. Dis. 2020, 16, 1096–1099. [Google Scholar] [CrossRef] [PubMed]
  70. Pena, J.E.-D.I.; Rascon-Pacheco, R.A.; Ascencio-Montiel, I.D.J.; Gonzalez-Figueroa, E.; Fernandez-Garate, J.E.; Medina-Gomez, O.S.; Borja-Bustamante, P.; Santillan-Oropeza, J.A.; Borja-Aburto, V.H. Hypertension, diabetes and obesity, major risk factors for death in patients with COVID-19 in Mexico. Arch. Med. Res. 2020, 52, 443–449. [Google Scholar] [CrossRef] [PubMed]
  71. Beck, A.L.; Huang, J.C.; Lendzion, L.; Fernandez, A.; Martinez, S. Impact of the COVID-19 pandemic on parents’ perception of health behaviors in children with overweight and obesity. Acad. Pediatr. 2021. [Google Scholar] [CrossRef]
  72. Tamara, A.; Tahapary, D.L. Obesity as a predictor for a poor prognosis of COVID-19: A systematic review. Diabetes Metab. Syndr. Clin. Res. Rev. 2020, 14, 655–659. [Google Scholar] [CrossRef]
  73. Leija-Martinez, J.J.; Huang, F.; Del-Rio-Navarro, B.E.; Sanchez-Munoz, F.; Munoz-Hernandez, O.; Giacoman-Martinez, A.; Hall-Mondragon, M.S.; Espinosa-Velazquez, D. IL-17A and TNF-α as potential biomarkers for acute respiratory distress syndrome and mortality in patients with obesity and COVID-19. Med. Hypotheses 2020, 144, 109935. [Google Scholar] [CrossRef] [PubMed]
  74. Tsenoli, M.; Smith, J.E.M.; Khan, M.A.B. A community perspective of COVID-19 and obesity in children: Causes and consequences. Obes. Med. 2021, 22, 100327. [Google Scholar] [CrossRef]
  75. Gazzaruso, C.; Paolozzi, E.; Valenti, C.; Brocchetta, M.; Naldani, D.; Grignani, C.; Salvucci, F.; Marino, F.; Coppola, A.; Gallotti, P. Association between antithrombin and mortality in patients with COVID-19. A possible link with obesity. Nutr. Metab. Cardiovasc. Dis. 2020, 30, 1914–1919. [Google Scholar] [CrossRef]
  76. Rahmati-Ahmadabad, S.; Hosseini, F. Exercise against SARS-CoV-2 (COVID-19): Does workout intensity matter? (A mini review of some indirect evidence related to obesity. Obes. Med. 2020, 19, 100245. [Google Scholar] [CrossRef] [PubMed]
  77. Browne, N.T.; Snethen, J.A.; Greenberg, C.S.; Frenn, M.; Kilanowski, J.F.; Gance-Cleveland, B.; Burke, P.J.; Lewandowski, L. When pandemics collide: The impact of COVID-19 on childhood obesity. J. Pediatr. Nurs. 2021, 56, 90–98. [Google Scholar] [CrossRef] [PubMed]
  78. Halvatsiotis, P.; Kotanidou, A.; Tzannis, K.; Jahaj, E.; Magira, E.; Theodorakopoulou, M.; Konstandopoulou, G.; Gkeka, E.; Pourzitaki, C.; Kapravelos, N.; et al. Demographic and clinical features of critically ill patients with COVID-19 in Greece: The burden of diabetes and obesity. Diabetes Res. Clin. Pract. 2020, 166, 108331. [Google Scholar] [CrossRef] [PubMed]
  79. Moriconi, D.; Masi, S.; Rebelos, E.; Virdis, A.; Manca, M.L.; Marco, S.D.; Taddei, S.; Nannipieri, M. Obesity prolongs the hospital stay in patients affected by COVID-19, and may impact on SARS-COV-2 shedding. Obes. Res. Clin. Pract. 2020, 14, 205–209. [Google Scholar] [CrossRef]
  80. Dugail, I.; Amri, E.-Z.; Vitale, N. High prevalence for obesity in severe COVID-19: Possible links and perspectives towards patient stratification. Biochimie 2020, 179, 257–265. [Google Scholar] [CrossRef] [PubMed]
  81. Robinson, E.; Boyland, E.; Chisholm, A.; Harrold, J.; Maloney, N.G.; Marty, L.; Mead, B.R.; Noonan, R.; Hardman, C.A. Obesity, eating behavior and physical activity during COVID-19 lockdown: A study of UK adults. Appetite 2021, 156, 104853. [Google Scholar] [CrossRef]
  82. Kass, D.A.; Duggal, P.; Cingolani, O. Obesity could shift severe COVID-19 disease to younger ages. Lancet 2020, 395, 1544–1545. [Google Scholar] [CrossRef]
  83. Chowdhury, A.I.; Alam, M.R.; Rabbi, M.F.; Rahman, T.; Reza, S. Does higher body mass index increase COVID-19 severity? A systematic review and meta-analysis. Obes. Med. 2021, 23, 100340. [Google Scholar] [CrossRef]
  84. Han, J.J.; Iyengar, A.; Helmers, M.R.; Smood, B.F.; Patrick, W.L.; Kelly, J.J.; Moss, N.; Najjar, S.S.; Houston, B.A.; Tedford, R.J.; et al. The effect of body mass index on outcomes among COVID-19 patients with left ventricular assist devices: A multi-institutional study. J. Heart Lung Transplant. 2021, 40, S101. [Google Scholar] [CrossRef]
  85. Do, M.H.; Lee, H.-B.; Oh, M.-J.; Jhun, H.; Choi, S.Y.; Park, H.-Y. Polysaccharide fraction from greens of Raphanus sativus alleviates high fat diet-induced obesity. Food Chem. 2021, 343, 128395. [Google Scholar] [CrossRef]
  86. Pietri, L.; Giorgi, R.; Begu, A.; Lojou, M.; Koubi, M.; Cauchois, R.; Grangeot, R.; Dubois, N.; Kaplanski, G.; Valero, R.; et al. Excess body weight is an independent risk factor for severe forms of COVID-19. Metabolism 2021, 117, 154703. [Google Scholar] [CrossRef]
  87. Pranata, R.; Lim, M.A.; Yonas, E.; Vania, R.; Lukito, A.A.; Siswanto, B.B.; Meyer, M. Body mass index and outcome in patients with COVID-19: A dose-response meta-analysis. Diabetes Metabol. 2021, 47, 101178. [Google Scholar] [CrossRef] [PubMed]
  88. Gao, M.; Piernas, C.; Astbury, N.M.; Hippisley-Cox, J.; O’Rahilly, S.; Aveyard, P.; Jebb, S.A. Associations between body-mass index and COVID-19 severity in 6.9 million people in England: A prospective, community-based cohort study. Lancet 2021, 9, 350–359. [Google Scholar] [CrossRef]
  89. Perez-Cruz, E.; Castanon-Gonzalez, J.A.; Ortiz-Gutierrez, S.; Garduno-Lopez, J.; Luna-Camacho, Y. Impact of obesity and diabetes mellitus in critically ill patients with SARS-CoV-2. Obes. Res. Clin. Pract. 2021, 15, 402–405. [Google Scholar] [CrossRef]
  90. Zamani, B.; Daneshzad, E.; Siassi, F.; Guilani, B.; Bellissimo, N.; Azadbakht, L. Association of plant-based dietary patterns with psychological profile and obesity in Iranian women. Clin. Nutr. 2020, 39, 1799–1808. [Google Scholar] [CrossRef]
  91. Tsigalou, C.; Paraschaki, A.; Karvelas, A.; Kantartzi, K.; Gagali, K.; Tsairidis, D.; Bezirtzoglou, E. Gut microbiome and Mediterranean diet in the context of obesity. Current knowledge, perspectives and potential therapeutic targets. Metabol. Open 2021, 9, 100081. [Google Scholar] [CrossRef]
  92. Bolori, P.; Setaysh, L.; Rasaei, N.; Jarrahi, F.; Yekaninejad, M.S.; Mirzaei, K. Adherence to a health plant diet may reduce inflammatory factors in obese and overweight women-a cross-sectional study. Diabetes Metab. Syndr. Clin. Res Rev. 2019, 13, 2795–2802. [Google Scholar] [CrossRef] [PubMed]
  93. Rajan, L.; Palaniswamy, D.; Mohankumar, S.K. Targeting obesity with plant-derived pancreatic lipase inhibitors: A comprehensive review. Pharmacol. Res. 2020, 155, 104681. [Google Scholar] [CrossRef]
  94. Shimoda, Y.; Okada, J.; Yamada, E.; Okada, K.; Okada, S.; Yamada, M. Association between percentage of obese people and water hardness in water purification plants. Obes. Med. 2020, 19, 100244. [Google Scholar] [CrossRef]
  95. Jupp, P.W. Selected environmental factors in a complex systems approach to managing obesity. Obes. Med. 2020, 19, 100275. [Google Scholar] [CrossRef]
  96. Sun, N.-N.; Wu, T.-W.; Chau, C.-F. Natural dietary and herbal products in anti-obesity treatment. Molecules 2016, 21, 1351. [Google Scholar] [CrossRef]
  97. Im, J.-Y.; Ki, H.-H.; Xin, M.; Kwon, S.-U.; Kim, Y.H.; Kim, D.-K.; Hong, S.-P.; Jin, J.-S.; Lee, Y.-M. Anti-obesity effect of Triticum aestivum sprout extract in high-fat-diet-induced obese mice. Biosci. Biotechnol. Biochem. 2015, 79, 1133–1140. [Google Scholar] [CrossRef] [Green Version]
  98. AbdelMassih, A.; Ye, J.; Kamel, A.; Mishriky, F.; Ismail, H.-A.; Ragab, H.A.; El Qadi, L.; Malak, L.; Abdu, M.; El-Husseiny, M.; et al. A multicenter consensus: A role of furin in the endothelial tropism in obese patients with COVID-19 infection. Obes. Med. 2020, 19, 100281. [Google Scholar] [CrossRef]
  99. Singh, M.; Thrimawithana, T.; Shukla, R.; Adhikari, B. Managing obesity through natural polyphenols: A review. Future Foods 2020, 1–2, 100002. [Google Scholar] [CrossRef]
  100. Marrelli, M.; Russo, N.; Chiocchio, I.; Statti, G.; Poli, F.; Conforti, F. Potential use in the treatment of inflammatory disorders and obesity of selected wild edible plants from Calabria region (Southern Italy). S. Afr. J. Bot. 2020, 128, 304–311. [Google Scholar] [CrossRef]
  101. Sargin, S.A. Plants used against obesity in Turkish folk medicine: A review. J. Ethnopharmacol. 2020, 270, 113841. [Google Scholar] [CrossRef]
  102. Cao, S.-Y.; Zhao, C.-N.; Xu, X.-Y.; Tang, G.-T.; Corke, H.; Gan, R.-Y.; Li, H.-B. Dietary plants, gut microbiota, and obesity: Effects and mechanisms. Trends Food Sci. Technol. 2019, 92, 194–204. [Google Scholar] [CrossRef]
  103. Lone, J.B.; Koh, W.Y.; Parray, H.A.; Paek, W.K.; Lim, J.; Rather, I.A.; Jan, A.T. Gut microbiome: Microglora association with obesity and obesity-related comorbidities. Microb. Pathog. 2018, 124, 266–271. [Google Scholar] [CrossRef]
  104. Naik, R.R.; Nemani, H.; Pothani, S.; Pothana, S.; Satyavani, M.; Qadri, S.S.Y.H.; Srinivas, M.; Parim, B. Obesity-alleviating capabilities of Acalypha indica, Pergulari ademia and Tinospora cardifolia leaves methanolic extracts in WNIN/GR-Ob rats. J. Nutr. Intermed. Metab. 2019, 16, 100090. [Google Scholar] [CrossRef]
  105. Njume, C.; Donkor, O.; McAinch, A.J. Predisposing factors of type 2 diabetes mellitus and the potential protective role of native plants with functional properties. J. Funct. Foods. 2019, 53, 115–124. [Google Scholar] [CrossRef]
  106. Aumeeruddy, M.Z.; Mahommodally, M.F. Traditional herbal medicines used in obesity management: A systematic review of ethnomedicinal surveys. J. Herb. Med. 2021, 28, 100435. [Google Scholar] [CrossRef]
  107. Moro, C.O.; Basile, G. Obesity and medicinal plants. Fitoterapia 2000, 71, S73–S82. [Google Scholar] [CrossRef]
  108. Afolayan, A.J.; Mbaebie, B.O. Ethnobotanical study of medicinal plants used as anti-obesity remedies in Nkonkobe Municipality of South Africa. Pharmacogn. J. 2010, 2, 368–373. [Google Scholar] [CrossRef] [Green Version]
  109. Cercato, L.M.; White, P.A.S.; Nampo, F.K.; Santos, M.R.V.; Camargo, E.A. A systematic review of medicinal plants used for weight loss in Brazil: Is there potential for obesity treatment? J. Ethnopharmacol. 2015, 176, 286–296. [Google Scholar] [CrossRef]
  110. Zhu, R.; Chen, B.; Bai, Y.; Miao, T.; Rui, L.; Zhang, H.; Xia, B.; Li, Y.; Gao, S.; Wang, X.-D.; et al. Lycopene in protection against obesity and diabetes: A mechanistic review. Pharmacol. Res. 2020, 159, 104966. [Google Scholar] [CrossRef]
  111. Benbouguerra, N.; Hornedo-Ortega, R.; Garcia, F.; El Khawand, T.; Saucier, C.; Richard, T. Stilbenes in grape berries and wine and their potential role as anti-obesity agents: A review. Trend Food Sci. Technol. 2021, 112, 362–381. [Google Scholar] [CrossRef]
  112. Batista, A.C.V.; Ribeiro, M.D.A.; Oliveira, K.A.D.; Freitas, P.A.D.; Santos, N.S.D.; Magalhaes, L.A.; Magalhaes, S.C.; Fonseca, S.G.D.C.; Aquino, J.D.S.; Souza, E.L.D.; et al. Effects of consumption of acerola, cashew and guava by-products on adiposity and redox homeostasis of adipose tissue in obese rats. Clin. Nutr. ESPEN 2021, 43, 283–289. [Google Scholar] [CrossRef] [PubMed]
  113. Vasileva, L.V.; Savova, M.S.; Tews, D.; Wabitsch, M.; Georgiev, M.I. Rosmarinic acid attenuates obesity and obesity-related inflammation in human adipocytes. Food Chem Toxicol. 2021, 149, 112002. [Google Scholar] [CrossRef] [PubMed]
  114. Adegbola, P.I.; Fadahunsi, O.S.; Ajilore, B.S.; Akintola, A.O.; Olorunnisola, O.S. Combined ginger and garlic extract improves serum lipid profile, oxidative stress markers and reduced IL-6 in diet induced obese rats. Obes. Med. 2021, 23, 100336. [Google Scholar] [CrossRef]
  115. Lu, K.-K.; Lin, S.-Z.; Dass, K.T.P.; Lin, W.-J.; Liu, S.-P.; Harn, H.-J. 3-N-butylphthalide protects against high-fat-diet-induced obesity in C57BL/6 mice and increases metabolism in lipid-accumulation cells. Biomed. Pharmacother. 2021, 139, 111687. [Google Scholar] [CrossRef]
  116. Shende, P.; Narvenker, R. Herbal nanotherapy: A new paradigm over conventional obesity treatment. J. Drug Deliv. Sci. Technol. 2021, 61, 102291. [Google Scholar] [CrossRef]
  117. Chukwuma, C.I.; Matsabisa, M.G.; Ibrahim, M.A.; Erukainure, O.L.; Chabalala, M.H.; Islam, M.S. Medicinal plants with concomitant anti-diabetic and anti-hypertensive effects as potential sources of dual acting therapies against diabetes and hypertension: A review. J. Ethnopharmacol. 2019, 235, 329–360. [Google Scholar] [CrossRef]
  118. Song, H.; Shen, X.; Deng, R.; Zhang, Y.; Zheng, X. Dietary anthocyanin-rich extract of acai protects from diet-induced obesity, liver steatosis, and insulin resistance with modulation of gut microbiota in mice. Nutrition 2021, 86, 111176. [Google Scholar] [CrossRef]
  119. Xu, J.; Ge, J.; He, X.; Sheng, Y.; Zheng, S.; Zhang, C.; Xu, W.; Huang, K. Caffeic acid reduces body weight by regulating gut microbiota in diet-induced-obese mice. J. Funct. Foods 2020, 74, 104061. [Google Scholar] [CrossRef]
  120. Ilyas, Z.; Perna, S.; Al-thawadi, S.; Alalwan, T.A.; Riva, A.; Petrangolini, G.; Gasparri, C.; Infantino, V.; Peroni, G.; Rondanelli, M. The effect of Berberine on weight loss in order to prevent obesity: A systematic review. Biomed. Pharmacother. 2020, 127, 110137. [Google Scholar] [CrossRef]
  121. Wu, F.; Yang, X.; Hu, M.; Shao, Q.; Fang, K.; Li, J.; Zhao, Y.; Xu, L.; Zou, X.; Lu, F.; et al. Wu-Mei-Wan prevents high-fat diet-induced obesity by reducing white adipose tissue and enhancing brown adipose tissue function. Phytomedicine 2020, 76, 153258. [Google Scholar] [CrossRef] [PubMed]
  122. Wu, S.-J.; Huang, W.-C.; Yu, M.-C.; Chen, Y.-L.; Shen, S.-C.; Yeh, K.-W.; Liou, C.-J. Tomatidine ameliorates obesity-induced nonalcoholic fatty liver disease in mice. J. Nutr. Biochem. 2021, 91, 108602. [Google Scholar] [CrossRef]
  123. El-Shiekh, R.A.; Al-Mahdy, D.A.; Hifnawy, M.S.; Abdel-Sattar, E.A. In vitro screening of selected traditional medicinal plants for their anti-obesity and anti-oxidation activities. S. Afr. J. Bot. 2019, 123, 43–50. [Google Scholar] [CrossRef]
  124. Janson, B.; Prasomthong, J.; Malakul, W.; Boonsong, T.; Tunsophon, S. Hibiscus sabdariffa L. calyx extract prevents the adipogenesis of 3T3-L1 adipocytes, and obesity-related insulin resistance in high-fat diet-induced obese rats. Biomed. Pharmacother. 2021, 138, 111438. [Google Scholar] [CrossRef] [PubMed]
  125. Sergent, T.; Vanderstraeten, J.; Winand, J.; Beguin, P.; Schneider, Y.-J. Phenolic compounds and plant extracts as potential natural anti-obesity substances. Food Chem. 2012, 135, 68–73. [Google Scholar] [CrossRef]
  126. Pugliese, G.; Barrea, L.; Laudisio, D.; Aprano, S.; Castellucci, B.; Framondi, L.; Di Matteo, R.; Savastano, S.; Colao, A.; Muscogiuri, G. Mediterranean diet as tool to manage obesity in menopause: A narrative review. Nutrition 2020, 79–80, 110991. [Google Scholar] [CrossRef]
  127. Zhou, F.; Yin, M.; Liu, Y.; Han, X.; Guo, J.; Ren, C.; Wang, W.; Huang, W.; Zhang, J.; You, Y. Grape seed flour intake decreases adiposity gain in high-fat-diet induced obese mice by activating thermogenesis. J. Funct. Foods 2019, 62, 103509. [Google Scholar] [CrossRef]
  128. Zheng, S.; Huang, K.; Zhao, C.; Xu, W.; Sheng, Y.; Luo, Y.; He, X. Procyanidin attenuates weight gain and modifies the gut microbiota in high fat diet induced obese mice. J. Funct. Foods 2018, 49, 362–368. [Google Scholar] [CrossRef]
  129. Perez-Ramirez, I.F.; Gonzalez-Davalos, M.L.; Mora, O.; Gallegos-Corona, M.A.; Reynoso-Camacho, R. Effect of Ocimum sanctum and Crataegus pubescens aqueous extracts on obesity, inflammation, and glucose metabolism. J. Funct. Foods 2017, 35, 24–31. [Google Scholar] [CrossRef]
  130. Li, W.; Wan, H.; Yan, S.; Yan, Z.; Chen, Y.; Guo, P.; Ramesh, T.; Cui, Y.; Ning, L. Gold nanoparticles synthesized with Poria cocos modulates the anti-obesity parameters in high-fat diet and streptozotocin induced obese diabetes rat model. Arab J. Chem. 2020, 13, 5966–5977. [Google Scholar] [CrossRef]
  131. Hussein, M.M.A.; Samy, M.; Arisha, A.H.; Saadeldin, I.M.; Alshammari, G.M. Anti-obesity effects of individual or combination treatment with Spirulina platensis and green coffee bean aqueous extracts in high-fat diet-induced obese rats. All Life 2020, 13, 328–338. [Google Scholar] [CrossRef]
  132. Cercato, L.M.; Oliveira, J.O.; Souza, M.T.S.; Andrade, N.; Martel, F.; Camargo, E.A. Effect of flavonoids in preclinical models of experimental obesity. Pharma Nutr. 2021, 16, 100260. [Google Scholar] [CrossRef]
  133. Freitas, D.F.D.; Guimaraes, V.H.D.; Borem, L.M.A.; Mafra, V.; Costa, D.V.D.; Costa, T.O.; Vieira, C.R.; Paula, A.M.B.D.; Guimaraes, A.L.S.; Santos, S.H.S. Acosmium dasycarpum (Vog.) Yakovlev root bark reduces obesity induced by hypercaloric diet in mice: Acosmium dasycarpum improves obesity. Phytochem. Lett. 2021, 44, 23–30. [Google Scholar] [CrossRef]
  134. Yu, S.; Li, H.; Cui, T.; Cui, M.; Piao, C.; Wang, S.; Ju, M.; Liu, X.; Zhou, G.; Xu, H.; et al. Onion (Allium cepa L.) peel extract effects on 3T3-L1 adipocytes and high-fat diet-induced obese mice. Food Biosci. 2021, 41, 101019. [Google Scholar] [CrossRef]
  135. Walid, R.; Hafida, M.; Haci, E.; Abdelhamid, I.; Reda, B.; Rachid, A.; Mohamed, B. Beneficial effects of Aloe vera gel on lipid profile, lipase activities and oxidant/antioxidant status in obese rats. J. Funct. Foods 2018, 48, 525–532. [Google Scholar] [CrossRef]
  136. Sirotkin, A.V. Can konjac (Amorphophallus konjac K. Koch) and it constituent glucomannan be useful for treatment of obesity? Obes. Med. 2021, 24, 100343. [Google Scholar] [CrossRef]
  137. Li, J.; Pang, B.; Shao, D.; Jiang, C.; Hu, X.; Shi, J. Artemisia sphaerocephala Krasch polysaccharide mediates lipid metabolism and metabolic endotoxaemia in associated with the modulation of gut microbiota in diet-induced obese mice. Int. J. Biol. Macromol. 2020, 147, 1008–1017. [Google Scholar] [CrossRef] [PubMed]
  138. Li, J.; Jin, H.; Yan, X.; Shao, D.; Hu, X.; Shi, J. The anti-obesity effects exerted by different fractions of Artemisia sphaerocephala Krasch polysaccharide in diet-induced obese mice. Int. J. Biol. Macromol. 2021, 182, 825–837. [Google Scholar] [CrossRef]
  139. Goyal, A.; Kaur, R.; Sharma, D.; Sharma, M. Protective effect of Betula utilis bark extract on high fat diet induced obesity in Wistar rats. Obes. Med. 2019, 15, 100123. [Google Scholar] [CrossRef]
  140. Wu, L.; Yan, Q.; Chen, F.; Cao, C.; Wang, S. Bupleuri radix extract ameliorates impaired lipid metabolism in high-fat diet-induced obese mice via gut microbial-mediated regulation of FGF21 signaling pathway. Biomed. Pharmacother. 2021, 135, 111187. [Google Scholar] [CrossRef]
  141. Golandaz, G.; Pal, A.K.; Uplanchiwar, V.; Gautam, R.K. Butea monosperma flower extract partially reduces high-fat diet induced obesity in experimental rats. Obes. Med. 2020, 17, 100179. [Google Scholar] [CrossRef]
  142. Astell, K.J.; Mathai, M.L.; McAinch, A.J.; Stathis, C.G.; Su, X.Q. A pilot study investigating the effect of Caralluma fimbriata extract on the risk factors of metabolic syndrome in overweight and obese subjects: A randomized controlled clinical trial. Complement. Ther. Med. 2013, 21, 180–189. [Google Scholar] [CrossRef]
  143. Lee, D.-H.; Park, S.-H.; Lee, E.; Seo, H.-D.; Ahn, J.; Jang, Y.-J.; Ha, T.-Y.; Im, S.S.; Jung, C.H. Withaferin A exerts an anti-obesity effect by increasing energy expenditure through thermogenic gene expression in high-fat diet-fed obese mice. Phytomedicine 2021, 82, 153457. [Google Scholar] [CrossRef] [PubMed]
  144. Mohseni, F.; Ahmadiani, E.S.; Hekmatdoost, A. The effect of cumin on anthropometric measurements: A systematic review of randomized controlled clinical trials. Obes. Med. 2021, 23, 100243. [Google Scholar] [CrossRef]
  145. Zare, R.; Heshmati, F.; Fallahzadeh, H.; Nadjarzadeh, A. Effect of cumin powder on body composition and lipid profile in overweight and obese women. Complement. Ther. Clin. Pract. 2014, 20, 297–301. [Google Scholar] [CrossRef]
  146. Jafarnejad, S.; Tsang, C.; Taghizadeh, M.; Asemi, Z.; Keshavarz, S.A. A meta-analysis of cumin (Cuminum cyminim L.) consumption on metabolic and anthropometric indices in overweight and type 2 diabetics. J. Funct. Foods 2018, 44, 313–321. [Google Scholar] [CrossRef] [Green Version]
  147. Kazemipoor, M.; Hajifaraji, M.; Radzi, C.W.J.B.W.M.; Shamshirband, S.; Petkovic, D.; Kiah, M.L.M. Appraisal of adaptive neuro-fuzzy computing technique for estimating anti-obesity properties of a medicinal plant. Comput. Methods Programs Biomed. 2015, 118, 69–76. [Google Scholar] [CrossRef]
  148. Jack, B.U.; Malherbe, C.J.; Mamushi, M.; Muller, C.J.F.; Joubert, E.; Louw, J.; Pheiffer, C. Adipose tissue as a possible therapeutic target for polyphenols: A case for Cyclopia extracts as anti-obesity nutraceuticals. Biomed. Pharmacother. 2019, 120, 109439. [Google Scholar] [CrossRef]
  149. Mahboubi, M. Cynara scolymus (artichoke) and its efficacy in management of obesity. Bull. Fac. Pharm. Cairo Univ. 2018, 56, 115–120. [Google Scholar] [CrossRef]
  150. Seyedan, A.; Mohamed, Z.; Alshagga, M.A.; Koosha, S.; Alshawsh, M.A. Cynometra cauliflora Linn. attenuates metabolic abnormalities in high-fat diet-induced obese mice. J. Ethnopharmacol. 2019, 236, 173–182. [Google Scholar] [CrossRef]
  151. Chen, J.; Leong, P.K.; Leung, H.Y.; Chan, W.M.; Wong, H.S.; Ko, K.M. 48Biochemical mechanisms of the anti-obesity effect of a triterpenoid-enriched extract of Cynomorium songaricum in mice with high-fat-diet-induced obesity. Phytomedicine 2020, 73, 153038. [Google Scholar] [CrossRef]
  152. Al-Rimawi, F.; Jaradat, N.; Qneibi, M.; Hawash, M.; Emwas, N. Free radicals and enzymes inhibitory potentials of the traditional medicinal plant Echium angustifolium. Eur. J. Integr. Med. 2020, 38, 101196. [Google Scholar] [CrossRef]
  153. Acin, S.; Munoz, D.L.; Guillen, A.; Soscue, D.; Castano, A.; Echeverri, F.; Balcazar, N. Triterpene-enriched fractions from Eucalyptus tereticornis ameliorate metabolic alterations in a mouse model of diet-induced obesity. J. Ethnopharmacol. 2011, 265, 113298. [Google Scholar] [CrossRef]
  154. Maia-Landim, A.; Lancho, C.; Poblador, M.S.; Lancho, J.L.; Ramirez, J.M. Garcinia cambogia and Glucomannan reduce weight, change body composition and ameliorate lipid and glucose blood profiles in overweight/obese patients. J. Herb. Med. 2021, 26, 100424. [Google Scholar] [CrossRef]
  155. Arika, W.M.; Kibiti, C.M.; Njagi, J.M.; Ngugi, M.P. Anti-obesity effects of dichloromethane leaf extract of Gnidia glauca in high fat diet-induced obese rats. Heliyon 2019, 5, e02800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Sang, T.; Guo, C.; Guo, D.; Wu, J.; Wang, Y.; Wang, Y.; Chen, J.; Chen, C.; Wu, K.; Na, K.; et al. Suppression of obesity and inflammation by polysaccharide from sporoderm-broken spore of Ganoderma lucidum via gut microbiota regulation. Carbohydr. Polym. 2021, 256, 117594. [Google Scholar] [CrossRef]
  157. Alarcon-Aguilar, F.J.; Zamilpa, A.; Perez-Garcia, M.D.; Almanza-Perez, J.C.; Romero-Nunez, 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] [PubMed]
  158. Martinet, A.; Hostettmann, K.; Schutz, Y. Thermogenic effects of commercially available plant preparations aimed at treating human obesity. Phytomedicine 1999, 6, 231–238. [Google Scholar] [CrossRef]
  159. Anigboro, A.A.; Avwioroko, O.J.; Akeghware, O.; Tonukari, N.J. Anti-obesity, antioxidant and in silico evaluation of Justicia carnea bioactive compounds as potential inhibitors of an enzyme linked with obesity: Insights from kinetics, semi-empirical quantum mechanics and molecular docking analysis. Biophys. Chem. 2021, 274, 106607. [Google Scholar] [CrossRef]
  160. Bais, S.; Patel, N.J. In vitro anti diabetic and anti obesity effect of J. communis extract on 3T3L1 mouse adipocytes: A possible role of MAPK/ERK activation. Obes. Med. 2020, 18, 100219. [Google Scholar] [CrossRef]
  161. Yang, R.-M.; Liu, F.; He, Z.-D.; Ji, M.; Chu, X.-X.; Kang, Z.-Y.; Cai, D.-Y.; Gao, N.-N. Anti-obesity effect of total phenylpropanoid glycosides from Ligustrumrobustum Blume in fatty diet-fed mice via up-regulating leptin. J. Ethnopharmacol. 2015, 169, 459–465. [Google Scholar] [CrossRef] [PubMed]
  162. Zhang, X.; Hu, P.; Zhang, X.; Li, X. Chemical structure elucidation of an inulin-type fructan isolated from Lobelia chinensis lour with anti-obesity activity on diet-induced mice. Carbohyd. Polym. 2020, 240, 116357. [Google Scholar] [CrossRef] [PubMed]
  163. Bharathi, V.; Rengarajan, R.L.; Radhakrishnan, R.; Hashem, A.; Abd-Allah, E.F.; Alqarawi, A.A.; Anand, A.V. Effects of a medicinal plant Macrotylomauniflorum (Lam.) Verdc. Formulation (MUF) on obesity-associated oxidative stress-induced liver injury. Saudi J. Biol. Sci. 2018, 25, 1115–1121. [Google Scholar] [CrossRef]
  164. Kumaraswamy, A.; Gurunagarjan, S.; Pemiah, B. Scientific evaluation of anti-obesity potential of aqueous seed kernel extract of Mangifera indica Linn. in high fat diet induced obese rats. Obes. Med. 2020, 19, 100264. [Google Scholar] [CrossRef]
  165. Lee, D.; Shin, Y.; Jang, J.; Park, Y.; Ahn, J.; Jeong, S.; Shin, S.S.; Yoon, M. The herbal extract ALS-L1023 from Melissa officinalis alleviates visceral obesity and insulin resistance in obese female C57BL/6J mice. J. Ethnopharmacol. 2020, 253, 112646. [Google Scholar] [CrossRef]
  166. Sunil, V.; Shree, N.; Venkataranganna, M.V.; Bhonde, R.R.; Majumdar, M. The anti diabetic and anti obesity effect of Memecylon umbellatum extract in high fat diet induced obese mice. Biomed. Pharmacother. 2017, 89, 880–886. [Google Scholar] [CrossRef]
  167. Metwally, F.M.; Rashad, H.M.; Ahmed, H.H.; Mahmoud, A.A.; Raouf, E.R.A.; 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]
  168. Ezzat, S.M.; El Bishbishy, M.H.; Aborehab, N.M.; Salama, M.M.; Hasheesh, A.; Motaal, A.A.; Rashad, H.; Metwally, F.M. Upregulation of MC4R and PPAR-α expression mediates the anti-obesity activity of Moringa oleifera Lam. in high-fat diet-induced obesity in rats. J. Ethnopharmacol. 2020, 251, 112541. [Google Scholar] [CrossRef] [PubMed]
  169. Greish, S.M.; Kader, G.S.A.; Abdelaziz, E.Z.; Eltamany, D.A.; Sallam, H.S.; Abogresha, N.M. Lycopene is superior to moringa in improving fertility markers in diet-induced obesity male rats. Saudi J. Biol. Sci. 2021, 28, 2956–2963. [Google Scholar] [CrossRef] [PubMed]
  170. Alkhudhayri, D.A.; Osman, M.A.; Alshammari, G.M.; Maiman, S.A.A.; Yahya, M.A. Moringa peregrina leaf extracts produce anti-obesity, hypoglycemic, anti-hyperlipidemic and hepatoprotective effects on high-fat diet fed rats. Saudi J. Biol Sci. 2021, 28, 3333–3342. [Google Scholar] [CrossRef] [PubMed]
  171. Mahboubi, M. Morus alba (mulberry), a natural potent compound in management of obesity. Pharmacol. Res. 2019, 146, 104341. [Google Scholar] [CrossRef]
  172. Namazi, N.; Larijani, B.; Ayati, M.H.; Abdollahi, M. The effects of Nigellasativa L. on obesity: A systematic review and meta-analysis. J. Ethnopharmacol. 2018, 219, 173–181. [Google Scholar] [CrossRef]
  173. Mangal, P.; Khare, P.; Jagtap, S.; Bishnoi, M.; Kondepudi, K.K.; Bhutani, K.K. Screening of six Ayurvedic medicinal plants for anti-obesity potential: An investigation on bioactive constituents from Oroxylum indicum (L.) Kurz bark. J. Ethnopharmacol. 2017, 197, 138–146. [Google Scholar] [CrossRef]
  174. Vuolo, M.M.; Lima, G.C.; Batista, A.G.; Carazin, C.B.B.; Cintra, D.E.; Prado, M.A.; Marostica, M.R., Jr. Passion fruit peel intake decreases inflammatory response and reverts lipid peroxidation and adiposity in diet-induced obese rats. Nutr. Res. 2020, 76, 106–117. [Google Scholar] [CrossRef] [PubMed]
  175. Oliveira, A.M.D.; Freitas, A.F.S.D.; Costa, M.D.D.S.; Torres, M.K.D.S.; Castro, Y.A.D.A.; Almeida, A.M.R.; Paiva, P.M.G.; Carvalho, B.M.; Napoleao, T.H. Pilosocereus gounellei (Cactaceae) stem extract decreases insulin resistance, inflammation, oxidative stress, and cardio-metabolic risk in diet-induced obese mice. J. Ethnopharmacol. 2021, 265, 113327. [Google Scholar] [CrossRef] [PubMed]
  176. Naidu, P.B.; Nemani, H.; Meriga, B.; Mehar, S.K.; Potana, S.; Ramgopalrao, S. Mitigating efficacy of piperine in the physiological derangements of high fat diet induced obesity in Sprague Dawley rats. Chem. Biol. Interact. 2014, 221, 42–51. [Google Scholar] [CrossRef]
  177. Harbilas, D.; Brault, A.; Vallerand, D.; Martineau, L.C.; Saleem, A.; Arnason, J.T.; Musallam, L.; Haddad, P.S. Populus balsamifera L. (Salicaceae) mitigates the development of obesity and improves insulin sensitivity in a diet-induced obese mouse model. J. Ethnopharmacol. 2012, 141, 1012–1020. [Google Scholar] [CrossRef]
  178. Diaz-de-Cerio, E.; Rodriguez-Nogales, A.; Algieri, F.; Romero, M.; Verardo, V.; Segura-Carretero, A.; Duarte, J.; Galvez, J. The hypoglycemic effects of guava leaf (Psidium guajava L.) extract are associated with improving endothelial dysfunction in mice with diet-induced obesity. Food Res. Int. 2017, 96, 64–71. [Google Scholar] [CrossRef] [PubMed]
  179. Gao, L.; Hu, Y.; Hu, D.; Li, Y.; Yang, S.; Dong, X.; Alharbi, S.A.; Liu, H. Anti-obesity activity of gold nanoparticles synthesized from Salacia chinensis modulates the biochemical alterations in high-fat diet-induced obese rat model via AMPK signaling pathway. Arab J. Chem. 2020, 13, 6589–6597. [Google Scholar] [CrossRef]
  180. Souza, T.D.; Silva, S.V.D.; Fonte-Faria, T.; Nascimento-Silva, V.; Barja-Fidalgo, C.; Citelli, C. Chia oil induces browning of white adipose tissue in high-fat diet-induced obese mice. Mol. Cell Endocrinol. 2020, 507, 110772. [Google Scholar] [CrossRef]
  181. Hamidpour, M.; Hamidpour, R.; Hamidpour, S.; Shahlari, M. Chemistry, pharmacology and medicinal property of sage (Salvia) to prevent and cure illnesses such as obesity, diabetes, depression, dementia, lupus, autism, heart disease and cancer. J. Tradit. Complement. Med. 2014, 4, 82–88. [Google Scholar] [CrossRef] [Green Version]
  182. Li, X.; Yang, L.; Xu, M.; Qiao, G.; Li, C.; Lin, L.; Zheng, G. Smilax china L. polyphenols alleviates obesity and inflammation by modulating gut microbiota in high fat/high sucrose diet-fed C57BL/6J mice. J. Funct. Foods 2021, 77, 104332. [Google Scholar] [CrossRef]
  183. Ansari, S.-A.; Bari, A.; Ullah, R.; Mathanmohun, M.; Veeraraghavan, V.P.; Sun, Z. Gold nanoparticles synthesized with Smilax glabra rhizome modulates the anti-obesity parameters in high-fat diet and streptozotocin induced obese diabetes rat model. J. Photochem. Photobiol. Biol. 2019, 201, 111643. [Google Scholar] [CrossRef] [PubMed]
  184. El-Shiekh, R.A.; Al-Mahdy, D.A.; Mouneir, S.M.; Hifnawy, M.S.; Abdel-Sattar, E.A. Anti-obesity effect of argel (Solenostemma argel) on obese rats fed a high fat diet. J. Ethnopharmacol. 2019, 238, 111893. [Google Scholar] [CrossRef] [PubMed]
  185. Iwamoto, K.; Fukuda, Y.; Tokikura, C.; Noda, M.; Yamamoto, A.; Yamamoto, M.; Yamashita, M.; Zaima, N.; Iida, A.; Moriyama, T. The anti-obesity effect of Taheebo (Tabebuia avellanedae Lorentz ex Griseb) extract in ovariectomized mice and the identification of a potential anti-obesity compound. Biochem. Biophys. Res. Commun. 2016, 478, 1136–1140. [Google Scholar] [CrossRef] [PubMed]
  186. Singh, H.; Sharma, A.K.; Gupta, M.; Singh, A.P.; Kaur, G. Tinosporacordifolia attenuates high fat diet-induced obesity and associated hepatic and renal dysfunctions in rats. PharmaNutrition 2020, 13, 100189. [Google Scholar] [CrossRef]
  187. Singh, H.; Bajaj, P.; Kalotra, S.; Bhandari, A.; Kaur, T.; Singh, A.P.; Kaur, G. Tinospora cordifolia ameliorates brain functions impairments associated with high fat diet induced obesity. Neurochem. Int. 2021, 143, 104937. [Google Scholar] [CrossRef]
  188. Fan, S.; Raychaudhuri, S.; Page, R.; Shahinozzaman, M.; Obanda, D.N. Metagenomic insights into the effects of Urtica dioica vegetable on the gut microbiota of C57BL/6J obese mice, particularly the composition of Clostridia. J. Nutr. Biochem. 2021, 91, 108594. [Google Scholar] [CrossRef] [PubMed]
  189. Kianbakht, S.; Hashem-Dabaghian, F. Antihypertensive efficacy and safety of Vaccinium arctostaphylos berry extract in overweight/obese hypertensive patients: A randomized, double-blind and placebo-controlled clinical trial. Complement. Ther. Med. 2019, 44, 296–300. [Google Scholar] [CrossRef]
  190. Lee, H.-B.; Oh, M.-J.; Do, M.H.; Kim, Y.-S.; Park, H.-Y. Molokhia leaf extract prevents gut inflammation and obesity. J. Ethnopharmacol. 2020, 257, 112866. [Google Scholar] [CrossRef]
  191. Zheng, C.-D.; Duan, Y.-Q.; Gao, J.-M.; Ruan, Z.-G. Screening for anti-lipase properties of 37 traditional Chinese medicinal herbs. J. Chin. Med. Associat. 2010, 73, 319–324. [Google Scholar] [CrossRef] [Green Version]
  192. Chahibakhsh, N.; Hosseini, E.; Islam, M.S.; Rahbar, A.R. Bitter almond gum reduces body mass index, serum triglyceride, hyperinsulinemia and insulin resistance in overweight subjects with hyperlipidemia. J. Funct. Foods 2019, 55, 343–351. [Google Scholar] [CrossRef]
  193. Jeepipalli, S.P.K.; Du, B.; Sabitaliyevich, U.Y.; Xu, B. New insights into potential nutritional effects of dietary saponins in protecting against the development of obesity. Food Chem. 2020, 318, 126474. [Google Scholar] [CrossRef]
  194. Patil, R.; Patil, R.; Ahirwar, B.; Ahirwar, D. Current status of Indian medicinal plants with antidiabetic potential: A review. Asian Pac. J. Trop. Biomed. 2011, 1, S291–S298. [Google Scholar] [CrossRef]
  195. Kadkhoda, G.; Zarkesh, M.; Saidpour, A.; Oghaza, M.H.; Hedayati, M.; Khalaj, A. Association of dietary intake of fruit and green vegetables with PTEN and P53 mRNA gene expression in visceral and subcutaneous adipose tissues of obese and non-obese adults. Gene 2020, 733, 144353. [Google Scholar] [CrossRef]
  196. Majewski, M.; Lis, B.; Juskiewicz, J.; Ognik, K.; Jedrejek, D.; Stochmal, A.; Olas, B. The composition and vascular/antioxidant properties of Taraxacum officinale flower water syrup in a normal-fat diet using an obese rat model. J. Ethnopharmacol. 2021, 265, 113393. [Google Scholar] [CrossRef] [PubMed]
  197. Fang, P.; He, B.; Yu, M.; Shi, M.; Zhu, Y.; Zhang, Z.; Bo, P. Treatment with celastrol protects against obesity through suppression of galanin-induced fat intake and activation of PGC-1αGLUT4 axis-mediated glucose consumption. Biochim. Biophys. Acta Mol. Basis Dis. 2019, 1865, 1341–1350. [Google Scholar] [CrossRef] [PubMed]
  198. Abolghasemi, J.; Jahromi, M.A.F.; Sharifi, M.H.; Mazloom, Z.; Hosseini, L.; Zamani, N.; Nimrouzi, M. Effects of Zataria oxymel on obesity, insulin resistance and lipid profile: A randomized controlled, triple-blind trial. J. Integr. Med. 2020, 18, 401–408. [Google Scholar] [CrossRef]
  199. 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] [PubMed]
  200. Tung, Y.-C.; Shih, Y.-A.; Nagabhushanam, K.; Ho, C.-T.; Cheng, A.-C.; Pan, M.-H. Coleus forskohlii and Garcinia indica extracts attenuated lipid accumulation by regulating energy metabolism and modulating gut microbiota in obese mice. Food Res. Int. 2021, 142, 110143. [Google Scholar] [CrossRef] [PubMed]
  201. Uti, D.E.; Atangwho, I.J.; Eyong, E.U.; Umoru, G.U.; Egbung, G.E.; Nna, V.U.; Udeozor, P.A. African walnuts attenuate ectopic fat accumulation and associated peroxidation and oxidative stress in monosodium glutamate-obese Wistar rats. Biomed. Pharmacother. 2020, 124, 109879. [Google Scholar] [CrossRef]
  202. Li, L.; Ma, H.; Liu, T.; Ding, Z.; Liu, W.; Gu, Q.; Mu, Y.; Xu, J.; Seeram, N.P.; Huang, X.; et al. Glucitol-core containing gallotannins-enriched red maple (Acerrubrum) leaves extract alleviated obesity via modulating short-chain fatty acid producing in high-fat diet-fed mice. J. Funct. Foods 2020, 70, 103970. [Google Scholar] [CrossRef]
  203. Sharma, P.P.; Baskaran, V. Polysaccharide (Laminaran and fucoidan), fucoxanthin and lipids as functional components from brown algae (Padina tetrastromatica) modulates adipogenesis and thermogenesis in diet-induced obesity in C57BL6 mice. Algal Res. 2021, 54, 102187. [Google Scholar] [CrossRef]
  204. Fonda, Y.B.; Tom, E.N.L.; Atsamo, A.D.; Bonabe, C.; Dimo, C. Effects of stem bark aqueous extract of Fagara tessmannii Engl (Rutaceae) on cardiovascular risks related to monosodium glutamate-induced obesity in rat: In vivo and in vitro assessments. J. Ethnopharmacol. 2020, 260, 112972. [Google Scholar] [CrossRef]
  205. Yuan, J.; Shi, W.; Chen, J.; Lu, J.; Wang, L.; Qiu, M.; Liu, J. Effects of 23-epi-26-deoxyactein on adipogenesis in 3T3-L1 preadipocytes and diet-induced obesity in C57BL/6 mice. Phytomedicine 2020, 76, 153264. [Google Scholar] [CrossRef]
  206. Majeed, M.; Majeed, S.; Nagabhushanam, K.; Lawrence, L.; Mundkur, L. Garcinia indica extract standardized for 20% Garcinol reduces adipogenesis and high fat diet-induced obesity in mice by alleviating endoplasmic reticulum stress. J. Funct. Foods 2020, 67, 103863. [Google Scholar] [CrossRef]
  207. Pai, S.A.; Martis, E.A.; Munshi, R.P.; Gursahani, M.S.; Mestry, S.N.; Juvekar, A.R. Chrysin mitigated obesity by regulating energy intake and expenditure in rats. J. Tradit. Complement. Med. 2020, 10, 577–585. [Google Scholar] [CrossRef] [PubMed]
  208. Mirtaheri, E.; Namazi, N.; Alizadeh, M.; Sargheini, N.; Karimi, S. Effects of dried licorice extract with low-calorie diet on lipid profile and atherogenic indices in overweight and obese subjects: A randomized controlled clinical trial. Eur. J. Inegr. Med. 2015, 7, 287–293. [Google Scholar] [CrossRef]
  209. Bahadori, M.B.; Maggi, F.; Zengin, G.; Asghari, B.; Eskandani, M. Essential oils of hedgenettles (Stachys inflata, S. lavandulifolia, and S. byzantine) have antioxidant, anti-Alzheimer, antidiabetic, and anti-obesity potential: A comparative study. Ind. Crops Prod. 2020, 145, 112089. [Google Scholar] [CrossRef]
  210. Hao, M.; Guan, Z.; Gao, Y.; Xing, J.; Zhou, X.; Wang, C.; Xu, J.; Li, W. Huang-Qi San ameliorates hyperlipidemia with obesity rats via activating brown adipocytes and converting white adipocytes into brown-like adipocytes. Phytomedicine 2020, 78, 153292. [Google Scholar] [CrossRef] [PubMed]
  211. Sahebkar-Khorasani, M.; Jarahi, L.; Cramer, H.; Safarian, M.; Naghedi-Baghdar, H.; Salari, R.; Behravanrad, P.; Azizi, H. Herbal medicines for suppressing appetite: A systematic review of randomized clinical trials. Complement. Ther. Med. 2019, 44, 242–252. [Google Scholar] [CrossRef]
  212. Tavvafian, N.; Darabi, H.; Ahani, A.; Naghizadeh, H.; Hajiaghaee, R.; Rahmati-Ahmadabad, S.; Azarbayjani, M.A. Effects of glycyrrhizic acid supplementation during nonlinear resistance training on inflammatory markers and muscular damage indices in overweight young men. Obes. Med. 2020, 17, 100178. [Google Scholar] [CrossRef]
  213. Alonso-Castro, A.J.; Ruiz-Padilla, A.J.; Ramirez-Morales, M.A.; Alcocer-Garcia, S.G.; Ruiz-Noa, Y.; Ibarra-Reynoso, L.D.R.; Solorio-Alvarado, C.R.; Zapata-Morales, J.R.; Mendoza-Macias, C.L.; Deveze-Alvarez, M.A.; et al. Self-treatment with herbal products for weight-loss among overweight and obese subjects from central Mexico. J. Ethnopharmacol. 2019, 234, 21–26. [Google Scholar] [CrossRef]
  214. Carrageta, D.F.; Dias, T.R.; Alves, M.G.; Oliveira, P.F.; Monteiro, M.P.; Silva, B.M. Anti-obesity potential of natural methylxanthines. J. Funct. Foods. 2018, 43, 84–94. [Google Scholar] [CrossRef]
  215. Xiong, P.; Niu, L.; Talaei, S.; Kord-Varkaneh, H.; Clark, C.C.T.; Gaman, M.-A.; Rahmani, J.; Dorosti, M.; Mousavi, S.M.; Zarezadeh, M.; et al. The effect of berberine supplementation on obesity indices: A dose-response meta-analysis and systematic review of randomized controlled trials. Complement. Ther. Clin. Pract. 2020, 39, 101113. [Google Scholar] [CrossRef] [PubMed]
  216. Hou, D.; Zhao, Q.; Yousaf, L.; Khan, J.; Xue, Y.; Shen, Q. Consumption of mung bean (Vigna radiata L.) attenuates obesity, ameliorates lipid metabolic disorders and modified the gut microbiota composition in mice fed a high-fat diet. J. Funct. Foods 2020, 64, 103687. [Google Scholar] [CrossRef]
  217. Zhang, Y.; Xu, L.; Ding, M.; Su, G.; Zhao, Y. Anti-obesity effect of garlic oil on obese rats via Shenque point administration. J. Ethnopharmacol. 2019, 231, 486–493. [Google Scholar] [CrossRef]
  218. Lin, S.; Wang, Z.; Lin, Y.; Ge, S.; Hamzah, S.S.; Hu, J. Bound phenolics from fresh lotus seeds exert anti-obesity effects in 3T3-L1 adipocytes and high-fat diet-fed mice by activation of AMPK. J. Funct. Foods 2019, 58, 74–84. [Google Scholar] [CrossRef]
  219. Kaur, A.; Behl, T.; Makkar, R.; Goyal, A. Effect of ethanolic extract of Cuscuta reflexa on high fat diet-induced obesity in Wistar rats. Obes. Med. 2019, 14, 100082. [Google Scholar] [CrossRef]
  220. Adeneye, A.A.; Adeyemi, O.O.; Agbaje, E.O. Anti-obesity and antihyperlipidaemic effect of Hunteria umbellata seed extract in experimental hyperlipidaemia. J. Ethnopharmacol. 2010, 130, 307–314. [Google Scholar] [CrossRef]
  221. Nadeem, S. Synergistic effect of Commiphora mukul (gum resin) and Lagenariasiceraria (fruit) extracts in high fat diet induced obese rats. Asian Pac. J. Trop. Dis. 2012, 2, S883–S886. [Google Scholar] [CrossRef]
  222. Zheng, Y.; Lee, J.; Shin, K.-O.; Park, K.; Kang, I.-J. Synergistic action of Erigeron annuus L. Pers and Borago officinalis L. enhances anti-obesity activity in a mouse model of diet-induced obesity. Nutr. Res. 2019, 69, 58–66. [Google Scholar] [CrossRef] [PubMed]
  223. Shi, M.; Loftus, H.; McAinch, A.J.; Su, X.Q. Blueberry as a source of bioactive compounds for the treatment of obesity, type 2 diabetes and chronic inflammation. J. Funct. Foods 2017, 30, 16–29. [Google Scholar] [CrossRef] [Green Version]
  224. Xie, M.-X.; Long, M.; Liu, Y.; Qin, C.; Wang, Y.-D. Characterization of the interaction between human serum albumin and morin. Biochim. Biophys. Acta Gen. Subj. 2006, 1760, 1184–1191. [Google Scholar] [CrossRef]
  225. Caselli, A.; Cirri, P.; Santi, A.; Paoli, P. Morin: A promising natural Drug. Curr. Med. Chem. 2016, 23, 774–791. [Google Scholar] [CrossRef] [PubMed]
  226. Madkhali, H.A. Morin attenuates high-fat diet induced-obesity related vascular endothelial dysfunction in Wistar albino rats. Saudi Pharm. J. 2020, 28, 300–307. [Google Scholar] [CrossRef] [PubMed]
  227. Yamakawa, J.-I.; Moriya, J.; Takeuchi, K.; Nakatou, M.; Motoo, Y.; Kobayashi, J. Significant of Kampo, Japanese traditional medicine, in the treatment of obesity: Basic and clinical evidence. Evid.-Based Complement. Altern. Med. 2013, 2013, 943075. [Google Scholar] [CrossRef]
  228. Rahman, A.H.M.; Rahman, M.D.M. Medicinal plants having anti-obesity potentiality available in Bangladeh: A review. Biol. Med. Case Rep. 2018, 2, 4–11. [Google Scholar] [CrossRef]
  229. Roh, C.; Jung, U. Screening of crude plant extracts with anti-obesity activity. Int. J. Mol. Sci. 2012, 13, 1710–1719. [Google Scholar] [CrossRef] [Green Version]
  230. Bahmani, M.; Eftekhari, Z.; Saki, K.; Fazeli-Moghadam, E.; Jelodari, M.; Rafieian-Kopaei, M. Obesity phytotherapy: Review of native herbs used in traditional medicine for obesity. J. Evid.-Based Complement. Altern. Med. 2016, 21, 228–234. [Google Scholar] [CrossRef]
  231. Liu, Y.; Sun, M.; Yao, H.; Liu, Y.; Gao, R. Herbal medicine for the treatment of obesity: An overview of scientific evidence from 2007 to 2017. Evid.-Based Complement. Altern. Med. 2017, 2017, 8943059. [Google Scholar] [CrossRef]
  232. Li, C.; Zhang, H.; Li, X. The mechanism of traditional Chinese medicine for the treatment of obesity. Diabetes Metab. Syndr. Obes. Targets Ther. 2020, 13, 3371–3381. [Google Scholar] [CrossRef] [PubMed]
  233. Chang, Y.C.; Yang, M.Y.; Chen, S.C.; Wang, C.J. Mulberry leaf polyphenol extract improves obesity by inducing adipocyte apoptosis and inhibiting preadipocyte differentiation and hepatic lipogenesis. J. Funct. Foods 2016, 21, 249–262. [Google Scholar] [CrossRef]
  234. Sung, J.; Lee, J. Capsicoside G, a furostanol saponin from pepper (Capsicum annuum L.) seeds, suppresses adipogenesis through activation of AMP-activated protein kinase in 3T3-L1 cells. J. Funct. Foods 2016, 20, 148–158. [Google Scholar] [CrossRef]
  235. Ali, F.; Ismail, A.; Esa, N.M.; Pei, C.P.; Kersten, S. Hepatic genome-wide expression of lipid metabolism in diet-induced obesity rats treated with cocoa polyphenols. J. Funct. Foods 2015, 17, 969–978. [Google Scholar] [CrossRef]
  236. Seo, C.R.; Yi, B.; Oh, S.; Kwon, S.M.; Kim, S.; Song, N.J.; Cho, J.Y.; Park, K.M.; Ahn, J.Y.; Hong, J.W.; et al. Aqueous extracts of hulled barley containing coumaric acid and ferulic acid inhibit adipogenesis in vitro and obesity in vivo. J. Funct. Foods 2015, 12, 208–218. [Google Scholar] [CrossRef]
  237. Kim, S.Y.; Wi, H.R.; Choi, S.; Ha, T.J.; Lee, B.W.; Lee, M. Inhibitory effect of anthocyanin-rich black soybean testa (Glycine max (L.) Merr.) on the inflammation-induced adipogenesis in a DIO mouse model. J. Funct. Foods 2015, 14, 623–633. [Google Scholar] [CrossRef]
  238. Okamatsu-Ogura, Y.; Tsubota, A.; Ohyama, K.; Nogusa, Y.; Saito, M.; Kimura, K. Capsinoids suppress diet-induced obesity through uncoupling protein 1-dependent mechanism in mice. J. Funct. Foods 2015, 19, 1–9. [Google Scholar] [CrossRef]
  239. Chen, Y.C.; Kao, T.H.; Tseng, C.Y.; Chang, W.T.; Hsu, C.L. Methanolic extract of black garlic ameliorates diet-induced obesity via regulating adipogenesis, adipokine biosynthesis, and lipolysis. J. Funct. Foods 2014, 9, 98–108. [Google Scholar] [CrossRef]
  240. Mashmoul, M.; Azlan, A.; Yusof, B.N.M.; Khazaai, H.; Mohtarrudin, M.; Boroushaki, M.T. Effects of saffron extract and crocin on anthropometrical, nutritional and lipid profile parameters of rats fed a high fat diet. J. Funct. Foods 2014, 8, 180–187. [Google Scholar] [CrossRef]
  241. Goto, Y.; Teraminami, A.; Lee, J.Y.; Ohyama, K.; Funakoshi, K.; Kim, Y.I.; Hirai, S.; Uemura, T.; Yu, R.; Takahashi, N.; et al. Tiliroside, a glycosidic flavonoids ameliorates obesity-induced metabolic disorders via activation of adiponectin signaling followed by enhancement of fatty acid oxidation in liver and skeletal muscle in obese-diabetic mice. J. Nutr. Biochem. 2012, 23, 768–776. [Google Scholar] [CrossRef] [PubMed]
  242. Murase, T.; Yokoi, Y.; Misawa, K.; Ominami, H.; Suzuki, Y.; Shibuya, Y.; Hase, T. Coffee polyphenols modulae whole-body substrate oxidation and suppress postprandial hyperglycaemia, hyperinsulinaemia and hyperlipidaemia. Br. J. Nutr. 2012, 107, 1757–1765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Suzuki, R.; Tanaka, M.; Takanashi, M.; Hussain, A.; Yuan, B.; Toyoda, H.; Kuroda, M. Anthocyanidins-enriched bilberry extracts inhibit 3T3-L1 adipocyte differentiation via the insulin pathway. Nutr. Metabol. 2011, 8, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. Anti-obesity potential of some medicinal plants and fruits.
Table 1. Anti-obesity potential of some medicinal plants and fruits.
PlantPlant FamilyKey PointReferences
Acosmium dasycarpum (Vog.) Yakovlev
(Unha D/anta)
FabaceaeIt is a Brazilian medicinal plant known as “Unha D/anta”, especially found in the Cerrado (Brazilian Savanna) area.[133]
Ascomium dasycarpum promoted metabolic syndrome; its root bark clearly ameliorates triglycerides; it has beneficial impacts on adiposity and weight reduction. [133]
Allium cepa L.
(Onion)
AmaryllidaceaeOnion peel extract meaningfully reduced the lipids of 3T3-L1 cells and restricted lipid accumulation by decreasing the expression of lipogenesis-related genes, with impressive anti-obesity impacts. [134]
Aloe barbadensis Miller
(Aloe vera)
AsphodelaceaeAloe vera gel application regulated adipose tissue accumulation in obese rats. [135]
Aloe vera gel regulated the dyslipidemia as well as oxidative stress in obese rats. [135]
Amorphophallus konjac K. Koch
(Konjac)
AraceaeIt has positive impacts on prevention and treatment of obesity because of its components konjak and glucomannan. [136]
Artemisia sphaerocephala Krasch
(Artemisia)
AsteraceaeIts polysaccharide fractions prevent obesity. [137,138]
Betula utilis
(Himalayan birch)
BetulaceaeBetula utilis bark is rich in pentacyclic triterpenoids thus it can be considered for management of overweight and associated comorbidities. [139]
Bupleuri Radix
(Chaihu)
ApiaceaeBupleuri Radix extract (BupE) increased HFD-induced lipid disorders through FGF21 signaling pathway. [140]
BupE reversed obesity-related changes in structure and function of gut microbial communities. [140]
Butea monosperma
(Sacred Tree)
FabaceaeIts flower extract can normalize the weight gain parameters. [141]
Caralluma fimbriataApocynaceaeApplication with C. fimbriata extract while controlling overall dietary intake and physical activity may be useful in curbing central obesity, and the key component of metabolic syndrome. [142]
Corchorus olitorius L.
(Nalta jute)
MalvaceaeMolokhia leaf extract ameliorated guy dysbiosis and high-fat diet-induced obesity. [143]
Cuminum cyminum L.
(Cumin)
ApiaceaeIt contains essential fatty acids, flavonoids, volatile oils, etc., and may have influence on weight and other anthropometric indices in obese and overweight people. [144]
Carum carvi L.
(Caraway)
ApiaceaeCumin promoted anthropometric and metabolic indices in overweight and type 2 diabetic subjects. [145,146]
As a traditional medicine, it may attenuate body mass index, body fat percentage, and body weight loss. [147]
Cyclopia spp.
(Honeybush)
FabaceaeIt has been used as the herbal tea (honeybush), which shows anti-obesity effects, especially by targeting adipose tissue. [148]
Cynara scolymus L.
(Artichoke)
AsteraceaeArtichoke has tremendous potency as anti-obese agent. [149]
Cynometra cauliflora Linn.
(Nam-nam)
FabaceaeIt can be used for obesity management. [150]
Cynomorium songaricum Rupr.
(Suo Yang)
CynomoriaceaeHCY2, a triterpenoid-enriched extract of Cynomorri Herba treat obesity via the regulation of AMPK/PGC1 pathways. [151]
Echium angustifolium Mill. BoraginaceaeIt can be considered as the potent candidate for oxidative stress, diabetes, and obesity. [152]
OBE100 and OBE104 are natural Eu extracts which are rich in pentacyclic triterpenes, and can be used to combat obesity and diabetes, and treatment with OBE100 had better effects than OBE104.[153]
Garcinia cambogia
(Garcinia)
ClusiaceaeGarcinia cambogia and Glucomannan decrease weight, alter body composition, regulate lipid and glucose blood profiles in overweight/obese patients. [154]
Gnidia glauca (Fres.) Gilg
(Fish Posion Bush)
ThymelaeaceaeIt may reduce the body weight, organ weights, anthropometric indices, organo-somatic indices, the total fat content, adiposity index, atherogenic index, and various lipid profiles. [155]
Ganoderma lucidum sensu stricto
(Mushroom of immortality)
GanodermataceaeIt has shown anti-obesity effects. [155,156]
Hibiscus sabdariffa L.
(Roselle)
MalvaceaeCholesterol and triglycerides levels indicated non-significant reductions in animals treated with Hibiscus sabdariffa, with its anti-obesity effect. [157]
Ilex paraguariensis
(Yerba mate)
AquifoliaceaeAfter treatment with Ilex paraguariensis, a drop in respiratory quotient (RQ) was observed, revealing a rise in the proportion of fat oxidized. [158]
Justicia carnea Lindl.
(The Brazilian plume flower)
AcanthaceaeThe methanolic leaf extract of Justicia carnea is a rich natural source of antioxidant and anti-obesity agents which could be optimized for development of new anti-obesity drugs. [159]
Juniperus communis L.
(Common Juniper)
CupressaceaeIt is an indigenous plant with significant in vitro anti-obesity impacts in adipocytes differentiation. [160]
Ligustrum robustum BlumeOleaceaeIn Chinese folk medicine, its leaves are used in the treatment of obesity and hyperlipidemia, and its anti-obesity impact was associated with up-regulation of leptin. [161]
Lobelia chinensis lour
(Asian lobelia)
CampanulaceaePolysaccharide from Lobelia chinensis lour is an insulin-type fructan with Mw of 2.6 kDa that indicated the potential of anti-obesity effect in HFD-induced mice. [162]
Macrotyloma uniflorum (Lam.) Verdc.
(Horse gram)
FabaceaeMacrotyloma uniflurom formulation may increase the activity of enzymatic superoxide dismutase, glutathione peroxidase, catalase, and non-enzymatic antioxidants and could be applied in treatment and prevention of fat-induced oxidative stress and inflammation. [163]
Mangifera indica Linn.
(Mango)
AnacardiaceaeIts anti-obesity activity may be mediated partially via pancreatic lipase inhibitory activity and partially through reduction in food intake and improvement of antioxidant status. [164]
Melissa officinalis L.
(Lemon balm)
LabiataeThe herbal extract ALS-L1023 may block visceral obesity, and also decreases the increased fasting blood glucose, impaired glucose tolerance, and pancreatic dysfunction seen in female obese mice. [165]
Memecylon umbellatum Burm. f.
(Ironwood)
MelastomataceaeThe oral administration of methanolic extract in mice may decrease hyperglycemia, triglycerides, body weight, and ameliorates insulin resistance. [166]
Moringa oleifera Lam.
(Drumstick tree)
MoringaceaeIts extract up-regulated adiponectin gene expression in obese rats relative, and down-regulated mRNA expression of leptin and resistin. [167]
Traditionally, M. oleifera leaves considered as anti-obesity herbal medicine. [168]
Its oil extract is considered to have cholesterol-lowering properties and a potential to treat obesity, while lycopene is a potent antioxidant. [169]
Moringa peregrine (Forssk.) Fiori.MoringaceaeIts bark and leaf extracts revealed potential anti-obesity and hepatoprotective activity through reduced lipid absorption, anti-hyperlipidemic impacts, and hepatic antioxidant effects. [170]
Morus alba L.
(Common mulberry)
MoraceaeThe inhibitory impacts of mulberry on digestive enzymes and adipocyte differentiation, and its stimulatory influences on energy expenditures, and lipid metabolism may have a role in obesity regulation. [171]
Nigella sativa L.
(Fennel flower)
RanunculaceaeIt shows a moderate impact on reduction in body mass index and body weight. [172]
Oroxylum indicum (L.) Kurz
(Kyaung shar)
BignoniaceaeOroxylin A, chrysin and baicalein were suppressed lipid accumulation in 3T3-L1 preadipocytes and PL enzyme. [173]
Oroxylin A and chrysin also suppressed PPARγ and C/EBPα, main adipogenic transcription factors, in 3T3L-1 preadipocytes during adipogenesis process at 50 μM dose. [173]
Passiflora edulis Sims
(Passion fruit)
PassifloraceaePassiflora edulis peel flour could be considered as an adjuvant to control of early parameters in obesity dysfunction. [174]
Pilosocereus gounellei (F.A.C. Weber) Byles and G.D. RowleyCactusIts stem extract enhanced serum lipid profile of the animals, lessened atherogenic indices, liver steatosis, epididymal fat, and pro-inflammatory cytokines. [175]
Piper nigrum L.
(Black pepper)
PiperaceaePiperine was separated from methanolic extract of Piper nigrum seeds, which may have suppressed role in body weight, increase insulin and leptin sensitivity, ultimately leading to balance obesity. [176]
Populus balsamifera L.
(Balsam poplar)
SalicaceaeIt has been known as a culturally adapted therapeutic approach for the treatment and care of diabetes and obesity.[177]
Psidium guajava L.
(Common guava)
MyrtaceaeGuava leaves promoted the vascular dysfunction in obese mice, and its application may have a positive effect on metabolic functions in obese mice. [178]
Salacia chinensis L.
(Chinese salacia)
CelastraceaeIt is generally regarded that Saptrangi and Salacia chinensis-loaded gold nanoparticles ameliorate body weight, adipose index, resistin, inflammatory markers and AMPKα1, liver marker enzymes, leptin, and lipid profile. [179]
Salvia hispanica L.
(Chia)
LamiaceaeChia oil increased glucose metabolism and it has revealed potential to protect against the development of obesity-related diseases.[180]
Salvia officinalis L.
(Common sage)
LamiaceaeCommon sage provides novel natural treatments for the relief or cure of obesity. [181]
Smilax china L.
(Baqia)
SmilaceaeSmilax china L. polyphenols may be applied as a potential candidate to prevent obesity. [182]
Smilax glabra Roxb.
(Glabrous greenbrier)
SmilaceaeSmilax glabra rhizome may change the anti-obesity constraints in high-fat diet and obese diabetes in animal models. [183]
Solenostemma argel HayneApocynaceaeArgel is a promising Egyptian natural substitute, as it is rich in pregnane glycosides. [184]
TabebuiaavellanedaeLorentz ex GrisebBignoniaceaeThe n-BuOH extract of Taheebo prohibited ovariectomy-induced obesity and reduce fat mass in ovariectomized mice. [185]
Tinospora cordifolia (Thunb.) Miers.MenispermaceaeIt was discovered to be efficacious in regulating body weight in a high-fat diet (HFD)-fed rats by keeping energy metabolism and cellular homeostasis. [186,187]
Urtica dioica L.
(Common nettle)
UrticaceaeIt may decrease diet-induced weight gain and insulin resistance. [188]
Vaccinium arctostaphylos L.
(Caucasian whortleberry)
EricaceaeVaccinium arctostaphylos berry may have antihypertensive influence, and significantly lowers systolic and diastolic blood pressures in overweight/obese hypertensive patients. [189]
Withania somnifera (L.) Dunal
(Winter cherry or poison gooseberry)
SolanaceaeWithaferin A (WFA) which is the principal component of Withania somnifera extract, restricted HFD-induced obesity, and regulated mitogen-activated protein kinase (MAPK) signaling and AMP-activated protein kinase (AMPK) in adipose tissue. [190]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sun, W.; Shahrajabian, M.H.; Cheng, Q. Natural Dietary and Medicinal Plants with Anti-Obesity Therapeutics Activities for Treatment and Prevention of Obesity during Lock Down and in Post-COVID-19 Era. Appl. Sci. 2021, 11, 7889. https://doi.org/10.3390/app11177889

AMA Style

Sun W, Shahrajabian MH, Cheng Q. Natural Dietary and Medicinal Plants with Anti-Obesity Therapeutics Activities for Treatment and Prevention of Obesity during Lock Down and in Post-COVID-19 Era. Applied Sciences. 2021; 11(17):7889. https://doi.org/10.3390/app11177889

Chicago/Turabian Style

Sun, Wenli, Mohamad Hesam Shahrajabian, and Qi Cheng. 2021. "Natural Dietary and Medicinal Plants with Anti-Obesity Therapeutics Activities for Treatment and Prevention of Obesity during Lock Down and in Post-COVID-19 Era" Applied Sciences 11, no. 17: 7889. https://doi.org/10.3390/app11177889

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop