Next Article in Journal
Contribution of Telomere Length to Systemic Sclerosis Onset: A Mendelian Randomization Study
Next Article in Special Issue
Anti-Obesity Effects of GABA in C57BL/6J Mice with High-Fat Diet-Induced Obesity and 3T3-L1 Adipocytes
Previous Article in Journal
A Retrospective Exploratory Analysis for Serum Extracellular Vesicles Reveals APRIL (TNFSF13), CXCL13, and VEGF-A as Prognostic Biomarkers for Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer
Previous Article in Special Issue
A Review of the Anti-Obesity Effects of Wild Edible Plants in the Mediterranean Diet and Their Active Compounds: From Traditional Uses to Action Mechanisms and Therapeutic Targets
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New Therapeutic Strategies for Obesity and Its Metabolic Sequelae: Brazilian Cerrado as a Unique Biome

by
Tamaeh Monteiro-Alfredo
1,2,3,4,5,
Maria Lígia Rodrigues Macedo
5,
Kely de Picoli Souza
4 and
Paulo Matafome
1,2,3,6,*
1
Coimbra Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
2
Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
3
Clinical Academic Center of Coimbra, 3000-075 Coimbra, Portugal
4
Research Group on Biotechnology and Bioprospection Applied to Metabolism and Cancer (GEBBAM), Federal University of Grande Dourados, Dourados 79804-970, MS, Brazil
5
Laboratório de Purificação de Proteínas e Suas Funções Biológicas (LPPFB), Federal University of Mato Grosso do Sul, Campo Grande 79070-900, MS, Brazil
6
Coimbra Health School (ESTeSC), Polytechnic University of Coimbra, Rua 5 de Outubro, 3046-854 Coimbra, Portugal
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(21), 15588; https://doi.org/10.3390/ijms242115588
Submission received: 31 August 2023 / Revised: 21 October 2023 / Accepted: 23 October 2023 / Published: 25 October 2023

Abstract

:
Brazil has several important biomes holding impressive fauna and flora biodiversity. Cerrado being one of the richest ones and a significant area in the search for new plant-based products, such as foods, cosmetics, and medicines. The therapeutic potential of Cerrado plants has been described by several studies associating ethnopharmacological knowledge with phytochemical compounds and therapeutic effects. Based on this wide range of options, the Brazilian population has been using these medicinal plants (MP) for centuries for the treatment of various health conditions. Among these, we highlight metabolic diseases, namely obesity and its metabolic alterations from metabolic syndrome to later stages such as type 2 diabetes (T2D). Several studies have shown that adipose tissue (AT) dysfunction leads to proinflammatory cytokine secretion and impaired free fatty acid (FFA) oxidation and oxidative status, creating the basis for insulin resistance and glucose dysmetabolism. In this scenario, the great Brazilian biodiversity and a wide variety of phytochemical compounds make it an important candidate for the identification of pharmacological strategies for the treatment of these conditions. This review aimed to analyze and summarize the current literature on plants from the Brazilian Cerrado that have therapeutic activity against obesity and its metabolic conditions, reducing inflammation and oxidative stress.

1. Introduction

In the last decades, there has been a significant increase in the life expectancy of the global population. According to the World Health Organization (WHO), between the years 2000 and 2015, life expectancy increased by an average of 5 years [1]. This remarkable improvement can be attributed to several factors, including a better quality of life, improved access to healthcare services, and the implementation of public policies aimed at disease prevention and treatment [1]. Although higher life expectancy is often associated with improved quality of life, it has also led to a higher incidence of chronic diseases worldwide, creating a significant burden on healthcare systems [2,3,4], requiring the development of more effective and affordable therapies to address this growing challenge.
Among the chronic illnesses that have been progressively more common lately, we highlight obesity, which is usually caused by unhealthy lifestyle habits—such as the consumption of processed foods rich in long-chain saturated fatty acids and added sugars [5], a sedentary lifestyle, and overweight [6]—and affects the overall quality of life of the population, both physically and economically, as well as psychologically [7]. Obesity is considered a major public health problem and is ranked as the fifth leading cause of mortality worldwide [8]. In addition to potentially leading to death, it can contribute to the development of other chronic disorders, including cancer [9], respiratory, hepatic, renal, and cardiovascular diseases (CVD) [10,11], and other metabolic disorders such as metabolic syndrome [8] and type 2 diabetes (T2D) [5,12].
The World Health Organization (WHO) has established overweight as a body mass index (BMI) between 25 and 29.9 kg/m2, and obesity is defined as a BMI above 30 kg/m2 [13]. Although BMI is a widely used parameter for determining obesity, there are limitations in its definition because this index does not differentiate between lean mass and fat, nor does it evaluate body fat distribution [14]. Additionally, besides defining parameters for the diagnosis of obesity, the WHO has also predicted that the adoption of unhealthy lifestyles will be responsible for 30% of deaths by the year 2030. Even though these data are alarming, they can be reversed through the identification of risk factors and the implementation of public policies promoting healthier habits [8]. Recent research [15,16,17] has observed that obesity-associated risks are more complex than simply weight excess. These studies highlight fat distribution as a crucial factor in the development of obesity-related comorbidities. Excess visceral fat also leads to the development of pro-oxidant conditions [11], increased proinflammatory cytokines, AT dysfunction, insulin resistance, pancreatic beta-cell dysfunction, and the hepatic accumulation of free fatty acids (FFA) [17,18]. Insulin resistance itself causes abnormal glucose and lipid metabolism in adipocytes, hepatocytes, and muscle cells, leading to a systemic metabolic dysregulation [19]. This systemic dysmetabolism results in redox imbalance which can trigger a variety of consequences linked with protein oxidation and glycation, lipid peroxidation, and the release of inflammatory signaling molecules.
In this context, it is important to highlight the importance of secondary metabolites of plants, which have therapeutic properties capable of contributing to the restoration of this imbalance in a comprehensive manner. These phytochemical compounds act through several mechanisms, such as capturing free radicals, activating or inhibiting enzymes, and specific transcriptional factors (such as Nrf2-Keap1, NF-κB, MAPK), regulating cellular activities, and also have modulatory effects on inflammatory processes (for example, in arachidonic acid metabolism, involving phospholipase A2 and COX, and in arginine metabolism, including NOS), enzymatic expression, and cytokine release [20,21]. The development of therapeutic options for obesity and its related diseases is crucial, especially if they have low cost, easy accessibility, low toxicity, and, most importantly, significant pharmacological potential. Approximately 65% to 80% of primary healthcare in developing countries depends on the consumption of medicinal plants (MP) [22], and more than 50% of the newly developed drugs are derived from them or their constituents [23]. However, MP accounts for only 15% of the existing 300,000 plant species worldwide [24]. Therefore, they offer a vast source of chemical compounds that are still underexplored and can be used in the development of new therapeutic alternatives [25].
Brazil is home to the world’s most extensive biodiversity, containing a rich variety of flora and fauna, including significant biomes such as the Cerrado [26]. This makes the country a crucial area for exploring new alternatives for producing plant-based products, including food, cosmetics, and medicines [27]. The high acceptance of plant-based treatments among the population highlights the importance of developing new treatment alternatives, especially in the field of obesity. The vast biodiversity of the Cerrado and the resulting diverse chemical composition of plants in it may reveal significant pharmacological effects. Several studies have explored this natural potential, and here we highlight the native plants of the Cerrado, which have demonstrated therapeutic effects, particularly in terms of anti-obesogenic properties, and/or their ability to assist in the treatment of its causes and consequences, such as AT inflammation, oxidative stress, CVD, and overall metabolic conditions.

2. Chemical Constitution of Native Plants of the Brazilian Cerrado

Brazilian biodiversity has significant economic value in the production of dietary and plant products, due to the diverse range of primary and secondary metabolites found in plants [27]. Secondary metabolites contribute to the chemical complexity of plants and are associated with their biological activities. The pharmacological potential of these plants, based on their phytochemical compounds, can aid in restoring the body’s redox balance [27] and chronic conditions [28,29]. Some metabolites that have biological effects related to the context of obesity and its complications are tannins [30,31], coumarins [32,33], terpenoids [34], anthocyanins [35,36], saponins [37], phenolic compounds, and flavonoids [38].
The efforts of science have demonstrated the increasing potential of MPs and their phytochemicals in reducing obesity. The use of multiple combinations of phytochemicals can enhance their beneficial effects at the molecular, cellular, and metabolic levels, providing advantages over treatments based on chemically synthesized drugs. Moreover, toxicity is usually found only for higher dosages, increasing the security of MPs. Plants and their derived compounds have the potential to control appetite, inhibit pancreatic lipase activity, induce lipolysis, stimulate thermogenesis, lipid metabolism and adipogenesis, and regulate fat cell cycle [39]. Various compounds extracted from plants are capable of regulating the different stages of adipocyte development, becoming important therapeutic strategies in the treatment of obesity [39]. Considering the wide range of compounds that have shown therapeutic activity in the field of obesity and its complications, we will provide a more detailed description of the two major groups that have demonstrated significant therapeutic potential: polyphenols and terpenes/sterols.

2.1. Polyphenols

Polyphenols constitute one of the largest classes of biologically active secondary metabolites. They are represented by over 8000 compounds identified in various plants, classified based on the number of phenolic rings, molecular structure, and functional groups attached to the phenolic rings. Their two main classes are flavonoids and non-flavonoids (phenolic acids, lignans, and stilbenes) [40]. These metabolites are responsible for a significant portion of the antioxidant activity found in plants. They exert their effects by chelating metal ions through redox reactions, inhibiting the transformation of hydroperoxides into reactive oxygen species (ROS), and acting as scavengers of free radicals by donating electrons from hydroxyl groups. The potential of phenolic compounds also lies in the ability to donate hydrogens or electrons, facilitating the stabilization of intermediate radicals [2].
The treatment of adipocytes with polyphenolic compounds has been shown to significantly stimulate the lipolysis and β-oxidation of fatty acids, reducing the accumulation of fatty acids and proinflammatory factors and exhibiting an inhibitory effect on their differentiation [39]. This is associated with data demonstrating the effect of polyphenols and their derivatives in improving the obesity condition and lipid profile of treated animals, associated with the increased utilization of fat for energy expenditure and glycemic balance [12,41,42]. These effects are associated with the activation of specific pathways that are crucial for the metabolic balance of the organism, namely the activation of peroxisome proliferator-activated receptors (PPARs), AMP-activated protein kinase (AMPK), and glucose transporters 2 and 4, which are further detailed in the following sections [12,39].
Regarding flavonoids, they not only possess antioxidant effects by reducing ROS accumulation through the capture of free radicals but also have a wide range of health benefits in humans due to their bioactive properties [43]. These include anti-inflammatory, anticancer, anti-aging, cardioprotective, neuroprotective, immunomodulatory, antidiabetic, antibacterial, antiparasitic, antiviral [44,45], and photoprotective activities [46]. Regarding the metabolic effect of flavonoids, they have shown lipolytic and synergistic effects when combined with other compounds, beyond their cytostatic and thermogenic potential [39]. In addition to the described effects of polyphenols per se, it is also important to highlight that these bioactive compounds undergo metabolism by various enzymes in the human body, leading to structural changes that result in improved bioavailability and enhanced effects [12,47]. Although the metabolic benefits of polyphenols have been extensively evaluated in animal models of obesity and metabolic disorders, their performance in clinical trials has not been so promising, and most of the studies until now demonstrate no effect or a very modest effect. Importantly, most of the studies use one single compound, and it is possible that a therapeutic effect may only be achieved with combinations, like in MP extracts, which have a mixture of compounds and may be the mixture that has therapeutic effects.

2.2. Terpenes and Sterols

Some plants composed of terpenoids and their derivatives, known as sterols, have shown potential effects in the treatment of metabolic syndrome and its complications. Terpenes are composed of isoprene units with five carbons and one carbon side chain. They include carotenoids such as β-carotene, a precursor of vitamin A [48], ursolic acid [49], tetraterpenes [50], and pentacyclic terpens [51,52], among others. The class of sterols includes sphytosterols as campesterol, sitosterol, and stigmasterol, which are an important part of dietary intake [48]. Regarding their biological effects, some plants containing terpenes have demonstrated antioxidant, antimicrobial, and anticancer activities.
Terpenes can also act as anxiolytic, immunomodulatory, analgesic, anti-inflammatory, neuroprotective, hepatoprotective, and wound-healing agents. They have also been studied for their potential against metabolic disorders, particularly obesity and T2D [53]. Specifically, concerning metabolic disorders, terpenes can activate PPARs, known for their anti-inflammatory action [34] (discussed in more detail in the section about inflammation and obesity).
In the article selection stage for this review, the scientific databases Science Direct and PubMed were used as primary sources. The keywords chosen for the search covered a variety of topics related to obesity and the Cerrado biome. Priority was given to articles that contained information about native plants, with a special focus on phytochemical composition. The inclusion of articles that met these criteria was carried out with the aim of adequately covering the intersection of topics of interest in this review. In summary, in Table 1 we highlight studies that link the chemical composition of plants to their various therapeutic effects, particularly regarding their potential for metabolic improvement, anti-obesogenic effects, and addressing of the causes and consequences of obesity, such as oxidative stress, inflammation, diabetes, and CVD. In addition, we summarized these data in Figure 1, correlating the therapeutic effect of the plants with the chemical constitution. These data support the importance of this biome as a source of bioactive compounds for the bioprospecting of future drugs and therapeutic strategies, given the complex chemical compositions and diverse metabolites found within it.
Table 1. Phytochemical constitution and biological properties related to obesity and its metabolic sequelae of medicinal plants from the Brazilian Cerrado.
Table 1. Phytochemical constitution and biological properties related to obesity and its metabolic sequelae of medicinal plants from the Brazilian Cerrado.
Medicinal PlantPhytochemical ConstituentsBiological PropertiesReferences
Acrocomia aculeataGallic, vanillic, caffeic, and ferulic acid, rutin, quercetin, campesterol, stigmasterol, β-sitosterol, lupeol, and lupeol acetateIn vitro and in vivo antioxidant activity, hypoglycemic, hypotriglyceridemic anticancer and cardioprotective effect[2,12,54]
Alibertia edulisCaffeic acid, quercetin 3-rhamnosyl-(1 → 6)-galactoside and iridois ioxideHypoglycemiant effect, protection against hemolysis and oxidative stress[55]
Alibertia verrucosaPhenolic compounds and tocopherolsAntioxidant activity[56]
Anacardium humilePhenolic compounds, anthocyanins and tocopherolsAntioxidant activity[56]
Annona crassifloraEpicatechin and quercetinAntioxidant, antiproliferative and wound healing [57]
Alkaloids, specially the isolated one, stevagallinAnti-obesity capacity, inhibition against pancreatic lipase with low cytotoxicity[58]
Annona muricataTotal phenolic compounds, flavonoids and proanthocyanidins (total quantification)Antioxidant activity, in vitro antidiabetic and inhibition of α-amylase, lipase, α-glucosidase, non-enzymatic glycation, and lipid peroxidation[59]
Phenols, flavonoids, saponins, tannins, steroids, and alkaloidsAntidiabetic and antiglycation[60]
Bactris setosaPhenolic compounds (anthocyanins and non-anthocyanin phenolic compounds) and carotenoidsOxidative and nitrosative protection[61,62]
Banisteriopsis argyrophyllaCatechin, flavonoids, glycosylated kaempferol, procyanidins, and megastigmane glucosidesα-amylase, α-glucosidase, lipase, and glycation inhibitors, antidiabetic and antioxidant [63]
Byrsonima verbascifoliaResveratrol and ferulic acidAntimutagenic, antigenotoxic and antioxidant activity[64]
Buchenavia tomentosaPhenols, carotenoids and tocopherolsAntioxidant activity[56,65]
Campomanesia cambessedeanaCatechin, ethyl gallate and propyl gallateAntimutagenic, antigenotoxic and antioxidant activity[64]
Caryocar brasilienseGallic acid, quinic acid, quercetin, and quercetin 3-O-arabinosAntioxidant activity[66]
Phenolic acids and tannins, including corilagin and geraniinAntidiabetic effect[67]
Cedrela odorataGallic acid, catechin and gallocatechinHyperglycemia reduction and antioxidant activity in vivo[68]
Dipteryx alataGallic acid and its derivatives, such as gallic acid esters and gallotanninsAntioxidant and antiproliferative activity[69]
p-Coumaric, ellagic, caffeic, ferulic, and gallic acid and hydroxybenzoic, catechin and epicatechinAntioxidant activity[70]
Phenolic compounds (total quantification)In vivo antioxidant activity[71]
Phenols and terpenesAntioxidant activity and Caenorhabditis elegans life expectancy increase[72]
Eschweilera nanatRutin and hyperosideAntioxidant activity[73]
Eugenia dysentericaProanthocyanidins, flavonoids, phenolic acids, quercetin, kaempferol derivatives, free and total ellagic acidAntioxidant activity, pancreatic lipase inhibition, lower body weight and fat mass, improved hyperglycemia, dyslipidemia and fecal triglycerides excretion[74]
Quercetin and gallic acidInhibitory effects on hydrolases, antioxidant and antiglycation properties[75]
Phenolic compounds (total quantification), myricetin, quercetin and kaempferolAntioxidant, antiproliferative and antimutagenic potential[76]
Eugenia klotzschianaMore than 35 compounds (see ref.)Antioxidant and antibacterial effect[77]
Guazuma ulmifoliaFlavan-3-ol-derived flavonoids, including monomers and dimers, condensed tannins, and glycosylated flavonoidsIn vitro and in vivo antioxidant activity[10]
Hancornia speciosaPhenolic compounds (total quantification)Antioxidant activity[78]
Hymenaea stignocarpaCaffeic acid, quercetin-3-rutinoside, kaempferol; quercetin-3-rhamnosideα-amylase and α-glucosidase inhibition, glycemic profile improved[79]
Hyptis Jacq.Phenolic acids, flavonoids, cinnamic acid derivatives, chlorogenic acid and rosmarinic acidAntioxidant activity[80]
Kielmeyera coriaceaProtocatechuic acid, procyanidins A, B, and C and epicatechin Antioxidant and antiglycation activity, and LPL inhibition.[81].
Mauritia flexuosaTotal phenolic compounds and β-carotene (total quantification)Antioxidant activity[82]
Carotenoids and polyphenols (catechin, quercetin and gallic acid)Antioxidant activity, antimutagenic, antimicrobial[83,84,84,85,86,87,88,89]
Mauritiella armataPalmitic, estearic, oleic, linoleic, linolenic acid, tocopherol, and α-tocopherolAntioxidant activity[90]
Myrcia bellaFlavonoids and phenolic acids derivativesAntimutagenic and antioxidant activity[91]
Passiflora setaceaPolyphenolsAntidiabetic and anti-inflammatory effect, with insulin, HOMA IR, PPAR-γ and IL-6 levels improvement[92]
Pouteria ramifloraFriedelin, epifriedelanol, taraxerol, triterpens and FFAAntioxidant and α-amylase inhibition[93]
Pouteria tortaPhenolic compounds, flavonoids, catechin and epicatechinAntioxidant and α-amylase inhibition[94]
Psidium cattleianumEpicatechin, gallic, coumaric, and ferulic acid, myricetin and quercetinAntioxidant and antimicrobial activity and antiproliferative effect on human cancer cells[95]
Rollinia mucosaPhenolic compounds and tocopherolsAntioxidant activity[56]
Schinus terebinthifolius RaddiO-glycosylated flavonols, gallotannins and gallic acid along with its derivativesAntioxidant, antidiabetic and antiproliferative activities[96,97]
Sterculia striataOleic acid, phytosterols β-sitosterol, stigmasteroland, campesterol γ-, δ-, α- and β-tocopherol, ellagic, ferulic, methoxyphenylacetic and protocatechuic acidsAntioxidant activity[98]
Solanum lycocarpu24 phenolic compounds (see ref.)Antioxidant activity[99]
Vochysiaceae speciesPolyphenols, such as flavonoids and condensed tanninsAntioxidant and inhibitory potential against human α-amylase and protein glycation[100]
Senna velutina21 compounds (see ref.)Antioxidant, in vitro and in vivo antitumor effects[101,102]
It is important to underscore that several studies have assessed the toxicity of various plants, with many confirming their safe use [2,54,103,104,104,105,106,107]. These studies provide additional evidence supporting their therapeutic potential, which is the focus of this work. While some plants are often safe for use, it is crucial to acknowledge the toxic potential of plants. Due to the richness of phytochemical compounds, its metabolization can generate toxic metabolites, mainly in the liver. The literature has not yet yielded definitive information on the mechanisms responsible for liver toxicity. However, factors such as high lipophilicity or elevated doses can potentially elicit such effects [108]. Furthermore, after demonstrating the importance and chemical richness of the plants found in the Brazilian Cerrado, we present their therapeutic potential on the pathophysiological mechanisms of obesity and its metabolic complications.

3. Obesity, Dyslipidemia and Inflammation

Obesity is most of the time characterized by AT dysfunction, which is closely associated with obesity-related diseases, such as CVD, hypertension, metabolic syndrome, and T2D [11,40,109,110,111]. AT dysfunction encompasses adipocyte hypertrophy, leukocyte infiltration and an exacerbated secretion of proinflammatory cytokines, including tumor necrosis factor alpha (TNF-α), interleukins IL-6, IL-1β, and IL-18, and monocyte chemoattractant protein-1 (MCP-1), resulting from nuclear factor kappa B (NF-κB) activation [112]. Furthermore, the accumulation of excessive free fatty acids (FFA) in the muscles and liver contributes to the activation of the c-Jun N-terminal kinase pathway (JNK). All these pathways hinder insulin-induced tyrosine phosphorylation and PI3K-AKT activation, resulting in insulin resistance [113]. AT inflammation culminates in impaired tissue plasticity, the installation of chronic low-grade inflammation, and persistent insulin resistance [34]. Adipocyte insulin resistance also increases the level of FFA in the bloodstream due to lower lipolysis inhibition [114]. Obesity also causes inflammation in the liver, skeletal muscle, and vessels (atherosclerosis), in part due to an increased efflux of FFA from AT to these tissues [114].
Visceral adipose tissue (VAT) has a higher rate of lipolysis and macrophage infiltration than subcutaneous adipose tissue (SAT) [115], especially proinflammatory M1 macrophages [114]. However, recent studies conducted in humans have suggested a subdivision between metabolically healthy and unhealthy obesity. Metabolically healthy obese individuals show better insulin sensitivity and have lower levels of ectopic fat, improved AT function, and lower cardiometabolic risk than metabolically unhealthy obese patients [114,116,117]. However, the molecular determinants of both conditions are yet to be established. Adipose tissue may also be affected by gluco- and lipotoxicity. The accumulation of AGEs and their precursors is also associated with local inflammation, impairing microcirculation and the expansion of AT, which can result in hypoxia and insulin resistance, especially when associated with high-fat diets and obesity [18,114]. Furthermore, elevated FFA levels increase superoxide (O2) generation, redox imbalance and lipid peroxidation [118]. Higher levels of FFA are associated with higher levels of 4-hydroxy-2-nonenal (4-HNE) in obese patients [119]. Abnormalities of mitochondrial function and biogenesis through a downregulation of PPARγ and PPAR co-activator 1-α (PGC-1α) are related to oxidative stress [120,121]. Mitochondrial dysfunction and reactive oxygen species (ROS) are associated with the dysregulation of adipokine secretion, fatty acid oxidation, and glucose homeostasis [11]. Table 2 summarizes the data found for plants that act on the regulation of inflammation, adipogenesis, browning, endocrine mechanisms, and enzyme activity.
Both crude extracts and isolated phytochemical compounds may become phytotherapeutic strategies for weight maintenance and metabolic homeostasis [115]. The main mechanisms by which phytochemical compounds from plants may improve obesity may include enzymatic inhibition, anti-inflammatory and antioxidant effects, appetite reduction, increased thermogenesis and lipid metabolism, and the regulation of adipogenesis [39,122,123]. The extract of Casearia sylvestris, composed mainly of the flavonoid glycosides and rutin, reduced dyslipidemia and atherogenesis caused by a high fat diet, besides improving the levels of cholesterol fractions and triglycerides, in Swiss mice [124]. The study conducted by Sousa et al. evaluated the effect of Davilla elliptica on diet-induced hepatic steatosis also in Swiss mice. Animals treated with the plant extract fraction and leaf powder showed reduced fat deposition in the liver, a lower body weight, and lower levels of total cholesterol and triglycerides [125].

3.1. Antilipidemic Effects of Cerrado Plants through Enzymatic Inhibition

Lipoprotein lipase (LPL) is responsible for metabolizing triglycerides from chylomicrons, allowing fatty acid uptake by adipocytes and triglyceride storage. Changes in LPL activity or its availability are directly related to higher concentrations of triglycerides in the blood, which can be stored in other tissues or metabolized [126]. The study conducted with Kielmeyera coriacea confirmed, in addition to antioxidant and antiglycation effects, the inhibitory effect on LPL, as well as other enzymes related to carbohydrate metabolism in vitro. The compounds found, including protocatechuic acid, epicatechin, and procyanidins A, B, and C, are likely responsible for the antioxidant, hyperglycemic, and hyperlipidemic effects of the plant [81].
Regarding digestive enzymes, some researchers have recently associated the inhibition of α-glucosidase and lipase with a reduction in postprandial glucose and appetite reduction, which can be positive for the treatment of obesity [127]. Many studies evaluated the inhibitory profile of phenolic compounds on digestive enzymes in vitro, including amylase, glucosidase, and phospholipase A2 [128]. Several studies also confirmed the inhibition of such enzymes by Cerrado plants, namely, B. argyrophylla [63], E. dysenterica [74], and A. muricata [59]. The study conducted by Pereira et al. on the species A. crassiflora, which is rich in chlorogenic acid, (epi)catechin, procyanidins, caffeoyl-hexosides, quercetin-glucosides, and kaempferol, suggested an antiobesogenic capacity of the isolated alkaloid compound stephalagine. Stephalagine demonstrated inhibitory activity in vitro against pancreatic lipase with low cytotoxicity [58].

3.2. Anti-Inflammatory Effect

Chronic inflammation in obesity and its comorbidities can also be treated by MPs, especially those native from the Cerrado. The phytochemical compounds, phenols, and flavonoids present in Xylopia aromatica were shown to reduce inflammation in overweight male BALB/c mice. The plant extract composed of flavonoids, namely rutin and caffeic and chlorogenic acid, was able to reduce glycemia during an oral glucose tolerance test, improve insulin sensitivity, attenuate inflammation and hepatic steatosis, and even showed potential in the release of anti-inflammatory cytokines, namely IL-4 and IL-13 in the liver and AT [123]. Pyrostegia venusta also exhibited anti-inflammatory effects in BALB/c mice induced with a high-carbohydrate-refined diet. Treatment with the extract of the plant showed an increase in IL-10 and a decrease in TNF-α, IL-6, IL-4, and IL-13 levels in AT. In the liver, the treatment decreased the TNF-α level. The plant extract was also able to reduce adipocyte area and adiposity, as well as to improve the metabolic profile of the animals [129].
In C57Bl/6 mice with a high-fat/high-sucrose diet, the reversal of the inflammatory condition was also achieved through E. dysenterica treatment, popularly known as cagaita. The plant extract is rich in polyphenols (total polyphenols, proanthocyanidins, ellagitannins, quercetin derivatives, kaempferol derivatives, and free ellagic acid) and was able to reduce NF-κB levels, protecting the animals from dyslipidemia and glucose alterations and reducing gluconeogenesis and hepatic inflammation [74,130]. Finally, the study conducted by Napolitano et al. demonstrated the anti-inflammatory effect of three other Cerrado plants, namely Serjania lethalis, Cupania vernalis, and Casearia sylvestris, through the measurement of nitric oxide released by macrophages [131].

3.3. Adipocyte Differentiation, Adipogenesis, and Browning

PPARs are members of a large family of nuclear receptors activated by fatty acids and their derivatives, regulating the metabolism of lipids and carbohydrates. They are predominantly expressed in the digestive tract, liver, and cardiac muscle and are involved in lipid catabolism. PPAR activators are widely used in the treatment of dyslipidemia, such as the pharmacological class of fibrates or Glitazones [34]. PPARs have also been described as a key transcription factor involved in adipogenesis [132], and they contribute to the reduction of inflammation and the transcriptional regulation of proinflammatory factors [133,134]. Another plant native to the Brazilian Cerrado, Hippeastrum stapfianum, showed a selective activation of PPAR-α and PPAR-γ and exhibited antioxidant effects. Although the focus of the study was not specifically metabolic diseases, such results suggest a possible effect on obesity [135]. Lastly, the study by Duarte et al. highlighted the properties of wild passion fruit (Passiflora setacea), which is rich in polyphenols. In individual male volunteers treated with P. setacea pulp, a significant reduction in insulin levels, HOMA IR, and IL-6 was observed. The plasma from the same individuals was used as a treatment in BV-2 microglial cells, and as a result, higher levels of PPAR-γ were observed in the cells, indicating an anti-inflammatory and antidiabetic effect [92].
There are no reports in the literature regarding the modulation of proteins such as C/EBP, FAS, and SREBP induced by Cerrado plants. However, some studies have shown a relationship between the presence of polyphenols and the increase in these proteins [136]. In addition, studies have also found an anti-obesity effect of catechins, which have been shown to reduce fat droplets in 3T3-L1 cells and the expression of transcription factors related to adipogenesis, such as SREBP-1c, C/EBP α, and PPAR γ [137]. An alternative to reduce obesity is the process of converting white adipocytes into brown adipocytes, known as browning. In brown adipocytes, PGC-1α promotes lipid oxidation and releases heat within the mitochondrial electron transport chain through the uncoupling protein 1 (UCP-1), a process called adaptive thermogenesis [138,139]. Although there are currently no studies relating Cerrado phytochemical compounds to browning, some reports observed this effect in plants rich in compounds such as those found in Cerrado plants, especially the great group of phenolic compounds and terpenes. They have been associated with the browning effect, adaptive thermogenesis, and the activation of target molecules such as PGC-1α, PPARγ, SIRT1, and AMPK [140]. This further justifies the investigation of these plants to determine their potential for future obesity treatment.

3.4. Modulation of Neuroendocrine Mechanisms

Appetite reduction is considered the primary approach for regulating and restoring body weight, being regulated by more than 40 hormones, enzymes, neuropeptides, and their respective receptors. Phytochemical compounds that have appetite-inhibiting activity include epigallocatechin, steroid glycosides, and saponins. It is believed that a significant portion of appetite inhibition occurs through signals from the brain–gastrointestinal system axis [39]. The hormones involved in this mechanism can also have their expression increased by medicinal plants. Although studies on this subject are still scarce, considering the main effect of body weight control, we can suggest that this mechanism of action may occur with the Baru nut, which has shown significant effects in improving satiety and in reducing body mass in adults [141,142]. Furthermore, the fruits of Solanum lycocarpum, known by the population as ‘fruta-de-lobo’, are rich in polysaccharides that promote delayed gastric emptying and induce changes in the endocrine system, affecting gastrointestinal hormones and consequently improving glycemic and lipid profiles [143]. Additionally, Rocha and colleagues conducted a study with various native Brazilian nuts, including cashew nuts (Anacardium occidentale—native to the Cerrado region), and they found that, although not altering leptin levels, their consumption for a period of 8 weeks showed potential in reducing ghrelin levels and consequently in decreasing appetite and cardiometabolic risk [144]. Another study conducted by Torres et al. evaluated the metabolic effect of pequi (C. brasiliense) administered to rats for 21 days. The oil from pequi kernels demonstrated the ability to reduce serum leptin levels, IL-6, leukotrienes 4 and 5, and TNF receptor. The treatment also showed a reduction in liver lesions and biochemical markers of liver function, AST and ALT, and increased serum concentrations of HDL. In addition, it exhibited antioxidant effects by increasing the expression of enzymes such as glutathione peroxidase and reductase [145].
Table 2. The role of medicinal plants in regulating inflammation, adipogenesis, browning, endocrine mechanisms and enzyme activity.
Table 2. The role of medicinal plants in regulating inflammation, adipogenesis, browning, endocrine mechanisms and enzyme activity.
Therapeutic PropertiesMedicinal PlantExperimental
Condition
Refs.Medicinal PlantExperimentalConditionModelTreatment/DoseRefs.
In VitroIn Vivo
Antilipidemic effect---Casearia sylvestrisHigh fat dietSwiss and C57BL/6 LDLr-null mice 250 and 500 mg/kg extract [124]
---Eugenia dysenterica High-fat high-sucrose diet C57BL/6J mice 7 and 14 mg gallic acid equivalent of extract/kg [74]
Enzymatic inhibitionKielmeyera coriacea α-amylase, α-glucosidase, and pancreatic lipase inhibition, antioxidant and antiglycation assays [81]-----
Banisteriopsis argyrophylla α-amylase, α-glucosidase, and pancreatic lipase inhibition, antioxidant and antiglycation assays [63]-----
Annona muricata α-amylase, α-glucosidase, and pancreatic lipase inhibition, antioxidant, antiglycation assays and cytotoxic assays [59]-----
Annona crassiflora Lipase inhibition and cytotoxic assay [58]-----
Anti-inflammatory effectSerjania lethalis, Cupania vernalis, Casearia sylvestris Determination of nitric oxide production and cytotoxicity assay [131]Xylopia aromaticaHigh carbohydrateBALB/c mice 50, 100 and 200 mg/kg extract [123]
---Pyrostegia venustaHigh-carbohydrate-refined dietBALB/c 300 mg/kg extract [129]
--- Eugenia dysenterica High-fat high-sucrose diet C57BL/6J mice 7 and 14 mg gallic acid equivalent of extract/kg [74,130]
Adipocyte differentiation, adipogenesis, and browning Hippeastrum stapfianum Activation of PPAR-α, PPAR-γ and antioxidant assays[135]Davilla elliptica High-lard/high-sugar diet Swiss mice 0.26 mg/kg extract [125]
---Passiflora setaceaClinical trialOverweight male volunteers and BV-2 microglial cells50 g, 150 g of pulp in two phases (humans), phenolic mebolites (cells)[92]
Modulation of neuroendocrine mechanisms---Anacardium occidentaleClinical trialWomen at cardiometabolic risk 15 g of Brazil nuts + 30 g of cashew nuts [144]
---Caryocar. brasilienseLiver injury induction with carbon tetrachlorideWistar rats 3 or 6 mL/kg of almond oil [145]

4. Type 2 Diabetes, Oxidative Stress and Glycation

Metabolic syndrome (MetS) is a common metabolic consequence of overweight and obesity and is cluster of metabolic abnormalities characterized by the simultaneous presence of the following risk factors: (I) abdominal obesity, (II) T2D, (III) hyperinsulinemia and/or glucose intolerance, (IV) dyslipidemia, and (V) hypertension [19], which can be either genetically predisposed or acquired throughout life due to certain habits. Typically, T2D manifests within the framework of metabolic syndrome, which is characterized by concurrent conditions such as obesity, hyperlipidemia, hypertension, and coagulation disorders. These interrelated factors significantly contribute to the development of CVD [146], the primary global cause of mortality [147]. CVD encompasses a wide range of conditions, including cerebrovascular disease, peripheral artery disease, cardiomyopathy, coronary heart disease, arrhythmias, and sudden death [148]. It is crucial to note that diabetic individuals face a significantly elevated risk, ranging from 150% to 400%, of experiencing stroke episodes [146].
Assuming that there is a vicious cycle among the three mentioned conditions, MP with antioxidant potential will likely show an improvement in common metabolic conditions. Given the antioxidant effect of plants from the Cerrado biome, as described by several studies already mentioned using extracts rich in compounds with phenolic structure, many of them also have antidiabetic effects [149]. Table 3 summarizes the data found for plants showing their action on the regulation of oxidative stress, T2D and glycation.

4.1. Antidiabetic Effects of Cerrado Plants

Several drugs used in the treatment of non-insulin-dependent diabetes slow down the digestion of starch by suppressing the activity of starch-hydrolyzing enzymes such as pancreatic α-amylase and intestinal α-glucosidase [60]. Although medications such as acarbose, voglibose, miglitol, and other starch blockers have been successfully used to manage postprandial hyperglycemia in T2D, their associated side effects have sparked interest in alternative therapies. Recent research suggests that plant-derived bioactive phytonutrients, such as alkaloids, cardiac glucosides, flavonoids, tannins, phenols, and steorids, with antioxidant properties can function as inhibitors of pancreatic α-amylase and intestinal α-glucosidase activities [150]. This inhibitory effect, in the context of phytotherapy, is attributed to the redox potential of their hydroxyl groups, which serve as effective enzyme inhibitors [151]. Besides enzymatic inhibition, several studies have also demonstrated the antidiabetic effects of Cerrado plants. For instance, the leaves of Annona muricata, used in ethnopharmacology for the treatment of T2D and its complications, were evaluated by Justino et al. regarding their in vitro antidiabetic effect. They determined the inhibition of lipidic and glycosidic hydrolases such as α-amylase, α-glucosidase, and pancreatic lipase, associated with antioxidant and antiglycation effects performed through DPPH, ORAC, FRAP assays, the reduction of Fe2+, the reduction of lipid peroxidation, and albumin and arginine glycation. By preparing various fractions of the leaf extract, the authors confirmed the presence of bioactive compounds described as antioxidants, such as quercetin and caffeic acid. They demonstrated the antioxidant effect of A. muricata in the conducted tests and the reduction of albumin glycation in the assays with methylglyoxal. The authors associate the phenolic group and the presence of hydroxyl groups with the effects observed [59]. Another in vitro study conducted with A. muricata by Olasehinde et al. also revealed and associated the plant’s antidiabetic and antiglycation potentials with its phytochemical constitution (phenols, flavonoids, saponins, tannins, steroids, and alkaloids) [60]. Finally, Pinto et al. showed the antidiabetic effect of this same plant but now analyzed the oil from the seeds in a model of streptozotocin (STZ)-induced type 1 diabetes (T1D) in BALB/c mice and in erythrocytes of diabetic patients. The oil from A. muricata seeds was able to preserve pancreatic islets and hepatic tissue and improve the patients’ anti-inflammatory profile with higher levels of IL-4 and 10 and lower levels of IFN-γ in the patients’ blood [152].
Additionally, in the study conducted by Florence et al. carried out in STZ-induced rats treated for 28 days, a single administration reduced blood glucose levels from 58.22% to 75%, depending on the dose. The extract was also able to reduce serum creatinine, MDA levels, AST, ALT, and LDL cholesterol and restore triglyceride and total cholesterol levels, as well as the antioxidant enzymes superoxide dismutase (SOD) and catalase (CAT) [153]. Another plant with demonstrated antidiabetic potential is Acrocomia aculeata. Besides acting as antioxidant in several experimental models, the aqueous extract of its leaves was able to reduce blood glucose and triglyceride levels in non-obese type 2 diabetic rats (Goto-Kakizaki) after 30 days’ treatment, as well as increasing proteins involved in glucose and lipid metabolism, such as GLUT-4, PPARγ, AMPK, and IR. Regarding the redox balance, the authors observed increased cell viability and higher catalase levels in 3T3-L1 preadipocytes exposed to hydrogen peroxide [2]. The extract also improved vascular function in diabetic animals and exhibited an important antioxidant effect on dermal microvascular cells, HMVec-D. The authors also associate the therapeutic effects with the phenolic composition of the extract (gallic, vanillic, ferulic, and caffeic acids), although other metabolites may be produced in vivo after the extract administration to the animals [12]. Another study was conducted with the same plant but using the kernel oil. It also showed an improvement in metabolic parameters, such as the reduction of hyperglycemia, increased insulin secretion, decreased insulin resistance, and enhanced function and number of pancreatic β-cells in both diabetic and non-diabetic rats. Similar to the study conducted with the leaf extract, kernel oil was able to improve the lipid profile and promoted better liver function [154]. Moreover, Da Silva’s work performed on the pulp of the same plant demonstrated a positive effect on the glycemic metabolism of rats and an antioxidant potential, without showing evidence of cytotoxicity [155]. These data contribute to an understanding of the extensive therapeutic potential of just one Cerrado plant and its various parts.
Other studies have also demonstrated the relationship between chemical composition, antioxidant effects, and metabolic improvement. A study conducted with Banisteriopsis argyrophylla showed the presence of phytochemical compounds such as glycosylated flavonoids, catechins, procyanidins, kaempferol, and glycosides, as well as its antioxidant activity by DPPH and ORAC, and the inhibition of non-enzymatic glycation, α-amylase, α-glucosidase, and lipase [63]. Jatobá-do-cerrado (Hymenaea stignocarpa Mart.) presented similar data regarding the inhibition of starch hydrolases with phytochemicals such as caffeic acid, kaempferol, quercetin-3-rutinoside, and quercetin-3-rhamnoside. Silva et al. proposed the substitution of a part of wheat flour by jatobá flour in bread and observed a less dose-dependent glycemic response. They confirmed the synergistic effect of dietary fibers and the phenolic compounds of Jatobá on glucose metabolism [79]. Eugenia dysenterica was also evaluated for its potential inhibitory effects on hydrolases, as well as its antioxidant and antiglycation properties. The authors found quercetin and gallic acid in its composition and confirmed the antidiabetic, antiglycation, and antioxidant effects of the plant [75]. Another interesting study conducted with Pequi (Caryocar brasiliense) demonstrated the presence of phenolic acids and tannins as constituents, including corilagin and geraniin, and linked them with the inhibition of insulin resistance-related factors, such as TNF-alpha and IL-1B, and inhibitory effects over α-glucosidase. In a starch tolerance test in mice, Pequi was able to reduce blood glucose levels [67]. Terminalia phaeocarpa also demonstrated enzymatic inhibition, while reducing in vivo blood glucose and TNF-alpha and IL-1B levels [156].
Other studies demonstrated the vast potential of the Cerrado plants Eugenia florida, Alibertia edulis, and Terminalia phaeocarpa in diabetic animal models, also with phytochemical composition mainly composed of phenols. They showed improvements in biochemical profiles, including higher levels of HDL cholesterol, improved liver function through AST and ALT markers, reduced uric acid, and reduced renal damage. They also presented a better redox condition, with lower levels of malondialdehyde and higher levels of glutathione (GSH) [55,157,158].
Regarding cashew (Anacardium othonianum), it is rich in vitamin C and was able to improve glucose levels during the glucose tolerance test, serum lipid levels, and systolic blood pressure in control women [159]. In a study conducted by Talpo et al. in C57Bl/6J mice, Siolmatra brasiliensis was able to reduce insulin resistance and lower serum cholesterol levels. Animals induced with HFD diet and treated with the extract showed a reduction in body weight and caloric intake, besides a 17% reduction in fasting blood glucose. Treatment also decreased biochemical markers related to glycation and lipid peroxidation, such as AGEs and TBARs, and increased antioxidant defenses such as CAT and SOD [160].
Finally, based on ethnopharmacological knowledge, cagaita (E. dysenterica) was studied for its antidiabetic potential in dysglycaemic female volunteers with metabolic syndrome. Araujo et al. showed that the treatment with cagaita juice was able to reduce 53% of the incremental area under the curve (iAUC) of glucose, 38% of insulin, 78% of glucose-dependent insulinotropic polypeptide (GIPb), and 58% of C-peptide, after juice consumption and bread intake, compared to control water. Glucagon-like peptide-1 (GLP-1) and glucagon values were not affected by the treatment with cagaita [161].
Table 3. Medicinal plants with potential therapeutic effects for treating T2D, oxidative stress, and glycation.
Table 3. Medicinal plants with potential therapeutic effects for treating T2D, oxidative stress, and glycation.
Therapeutic PropertiesMedicinal PlantExperimental
Condition
Refs.Medicinal PlantExperimental
Condition
ModelTreatment/
Dose
Refs.
In VitroIn Vivo/Clinical Studies
Enzymatic inhibition and antidiabetic effectsAnnona muricataα-amylase, α-glucosidase, and pancreatic lipase, associated with antioxidant and antiglycation assays[59]Annona muricataMice—streptozotocin (STZ)-induced T1DMale BALB/c mice and in erythrocytes of diabetic patients 1.0 mg/kg seed oil [152]
Annona muricataantidiabetic, and antiglycation potentials[60]Annona muricataSTZ-induced T2DMale wistar rats 100 mg/kg or 200 mg/kg extract [153]
Acrocomia aculeataAntioxidant and cytotoxic assays[2]Acrocomia aculeataNormal and non-obese T2D ratsMale wistar and Goto-Kakizaki rats 200 mg/kg extract [12]
---Acrocomia aculeataSTZ- and low HFD induction Male wistar rats 40 or 160 g of kernel oil [154]
Banisteriopsis argyrophyllaInhibition of α-amylase, α-glucosidase, and lipase and antioxidant assays[63]Acrocomia aculeataNormal, STZ and frutose-induced dietMale wistar rats 3, 30 or 300 mg/kg pulp oil [155]
Eugenia dysentericaInhibitory effects on hydrolases, antioxidant and antiglycation properties[75]Hymenaea stignocarpaNormal and healthyWomen Replacement of normal flour with pulp flour at 10, 20 and 30% [79]
---Caryocar
brasiliense
Normal ratsMale swiss mice and THP-1, CCL-13 and CR-1458 cell lines 100 mg/kg extract and fractions [67]
---Terminalia phaeocarpaNormal ratsMale swiss mice and THP-1 cell line 100 mg/kg extract and fractions [156]
---Eugenia floridaNormal and Alloxan-inducedMale Wistar rats 200 mg/kg extract [43]
---Alibertia edulisNormal and HDF-inducedMale swiss mice 200 and 400 mg/kg extract [53]
---Anacardium othonianumNormal and healthyWomen 400 mL juice [159]
---Siolmatra brasiliensisNormal and HDF-induceMale C57Bl/6J mice 125 or 250 mg/kg extract [160]
---Eugenia dysentericaMetabolic syndromeWoman 300 mL juice [161]
Redox imbalance and antioxidant potential Mauritia flexuosa Free radical scavenging[85]Acrocomia aculeataOxidative stress inductionC. elegans500–1000  μg/mL[2]
Mauritia flexuosa Free radical and hydroxyl scavenging, and reducing iron [82]Acrocomia aculeataNormal and non-obese T2D ratsWistar and Goto-Kakizaki rats 200 mg/kg [12]
Annona crassifloraABTS free radicals capture[162]--- - -
Solanum lycocarpumDPPH, FRAP and ORAC techniques[99,163]--- - -
Dipteryx alataAntioxidant profile with antiproliferative activity[69]--- - -

4.2. The Redox Imbalance and Antioxidant Potential of Phytochemical Compounds from the Cerrado Plants

Oxidative stress is an important factor transversal to various conditions such as cancer, premature aging, CVD, and metabolic disorders such as T2D and obesity [164]. ROS formation is a consequence of secondary glucose metabolism and the upregulation of alternative pathways that lead to glucotoxicity [165,166,167,168,169,170]. Oxidative damage plays a crucial role in neuronal damage and microvascular and cardiovascular complications of diabetes, such as nephropathy, retinopathy, and neuropathy, mainly due to glucotoxicity [12,171]. In hyperglycaemia, the glycolytic pathway, Krebs cycle, and mitochondrial electron transport chain (METC) are upregulated. This leads to higher levels of ATP and hyperpolarization of METC, promoting electron “escape” and partial oxygen reduction, resulting in higher levels of ROS, especially O2•− [172]. Additionally, glycolysis alternative pathways are activated, including protein kinase C (PKC), aldose reductase (polyol pathway), increased hexosamine flow, and advanced glycation end product (AGEs) formation [173]. All these alternative pathways are associated with ROS increase and are linked to the development of oxidative stress and T2D complications. Clinical indicators of oxidative stress include 4-hydroxy-2-nonenal (HNE) [169,170], 8-hydroxy-2-deoxyguanosine (8-OHdG) [165], and heme oxygenase-1 (HMOX1) [166,167]. The antioxidant enzymatic system is formed by enzymes such as SOD, glutathione peroxidase (GPx), and CAT [174]. The non-enzymatic system includes substances such as uric acid, lipoic acid, melatonin, bilirubin, and GSH. Exogenous antioxidants can be divided into two groups: natural and synthetic. Natural antioxidants are represented by phenolic compounds, carotenoids, flavonoids, and vitamins E, A, and C, among others [175]. They neutralize ROS from metabolism and exogenous sources, thereby preventing macromolecule damage. Synthetic antioxidants are more effective, stable, and cheap than natural ones but are associated with more side effects [176].
Several recent studies have reported an association between oxidative stress and AT dysfunction, namely elevated levels of FFA, dysregulation in adipokine production, and a reduced expression of antioxidant enzymes [177,178]. Thus, improving redox imbalance may attenuate the dysregulation of white AT and improve its function. The development of T2D is also closely related to the redox condition, being associated with reduced insulin biosynthesis and secretion [179]. The reduction of insulin biosynthesis in pancreatic β cells is further exacerbated by oxidative stress, which hampers the activity of the insulin gene’s promoter and transcription factors pancreatic duodenal homeobox-1 (PDX-1) and MAF bZIP transcription factor A (MafA) [180,181]. Besides decreasing the expression of insulin mRNA, the development, differentiation, maintenance, and regeneration of β cells are also compromised [179]. Consequently, the combination of hyperglycemia and oxidative stress leads to a decrease in insulin biosynthesis and secretion, thereby perpetuating a vicious cycle of sustained hyperglycemia. AGEs result from glucose auto-oxidation or the interaction of precursor metabolites (namely glyoxal and methylglyoxal, which are metabolic byproducts of glycolysis) with proteins and amino acids. Arginine is particularly important when assessing protein glycation caused by reactive carbonyl compounds. The inhibition of theses reactions, as well as the subsequent oxidative products, is of the utmost importance for the prevention and reduction of T2D complications, including the impairment of vessel function [182] and insulin resistance [183].
Natural compounds can act as antioxidants in various ways. Besides neutralizing ROS, their action includes the repair of ROS-induced injury, increasing the synthesis of antioxidant enzymes, and reducing oxidative damage [184,185]. One example of a Cerrado plant that is a source of antioxidant agents is A. aculeata, which has shown significant effects in capturing free radicals and reducing the oxidation of macromolecules (DNA, cell membrane lipids, and proteins). Extracts prepared from its leaves have been shown to be rich in phenolic compounds, tannins, and saponins (mainly gallic acid, vanillic acid, caffeic acid, ferulic acid, rutin, and quercetin) and to increase the survival of nematodes Caenorhabiditis elegans and normal cells Cos-7 exposed to oxidative damage. Furthermore, they have been found to increase the expression of protective transcription factors such as NRF2 and SIRT1 and antioxidant enzymes such as CAT, which were associated with the protective in vivo effects in diabetic rats [2,12].
The buriti palm (Mauritia flexuosa L.), a native palm tree of Latin America, especially from the Cerrado biome, is an important source of bioactive compounds such as carotenoids and polyphenols. In the analysis conducted by Leite et al., catechin, quercetin, and gallic acid were the major compounds in the fruit peel and pulp, demonstrating antioxidant effects in the scavenging of the ABTS free radical [85]. Candido et al. performed a comparative analysis between samples collected from two different Brazilian biomes, the Cerrado and the Amazon rainforest. Samples from the Cerrado exhibited a higher content of phenolic compounds and showed superior in vitro antioxidant activity. The buriti palm from the Cerrado displayed greater potential in capturing free radicals, reducing iron, and scavenging hydroxyl radicals, as confirmed by DPPH, ABTS, FRAP, and ORAC analyses, compared to the samples from the Amazon [82]. Other studies in the literature also confirmed the antioxidant effect of the buriti palm, associating this effect with its phenolic composition [83,84,86,87,89] and corroborating the previously mentioned antimicrobial activity data [84]. Furthermore, studies confirm its safety through antimutagenicity and micronucleus assays [88].
The study conducted by Rios de Souza et al. on five different species highlights the antioxidant potential of Marolo (Annona crassiflora Mart.), which showed a significant effect in capturing ABTS free radicals, a notable content of ascorbic acid, and a higher phenolic compound content, compared to the other four evaluated plants [162]. The study conducted with Solanum lycocarpum St. Hil., another native plant of the Brazilian Cerrado, linked the antioxidant effect determined in different extracted fractions of its fruits by DPPH and FRAP techniques. The chemical composition of this assay revealed the presence of phytosterols and terpenes [163]. The same plant was studied by Pereira et al., demonstrating once again its antioxidant potential, this time using the ORAC technique, in addition to the presence of phenolic compounds in its composition [99]. Dipteryx alata Vog. is another medicinal plant with therapeutic potential. It was analyzed by Oliveira-Alves et al., who associated its antioxidant profile with antiproliferative activity in human colon cancer cells HT29 and Caco-2. The extract rich in phenolic compounds such as gallic acid and its derivatives demonstrated its potential through the ORAC assay and hydroxyl radical scavenging capacity (HOSC) assay [69]. These are crucial studies that contribute to the characterization of the chemical composition and antioxidant profile of different species in the Cerrado biome. These plants are promising in reducing redox unbalance and thus in preventing the oxidative stress associated with metabolic disorders.

4.3. Limitations of the Study

In this review, our primary aim was to summarize the studies exclusively conducted on Cerrado plants. While there is a significant number of publications, many studies lack comprehensive descriptions of the plants’ phytochemical composition, and it is not yet possible to establish a composition–effect relationship. However, we chose not to exclude studies that did not present the full characterization of bioactive compounds, emphasizing the Cerrado’s extensive biodiversity even though it remains poorly documented. Methodological descriptions remain relatively underdeveloped, especially with regard to the study of mechanistic pathways related to bioactive compounds and their biological effects. In Figure 2, we summarize the possible mechanisms of action that may be related to the therapeutic potential of Cerrado plants for the treatment of obesity and its consequences.
Cerrado plants exhibit remarkable therapeutic potential, as proven in the course of our work. However, despite their promise, there are also few studies involving humans, and no reports were found of plants being evaluated in clinical studies for the development of new drugs. It is important to highlight that there is an incentive for the use of medicinal plants applied in phytotherapy, such as the National Policy on Medicinal Plants and Phytotherapy (PNPMF) of Brazil, which aims to guarantee the population’s safe access to medicinal plants and herbal medicines, promoting the responsible use of these natural resources. Despite these phytochemical compounds being strong candidates for the development of new allopathic drugs, extensive research is still needed.
Additionally, the low bioavailability of secondary metabolites, especially phenolic compounds and flavonoids, represents a significant challenge for medicinal plant research. The results obtained in in vitro studies are often not reflected in in vivo tests [186] due to characteristics such as low water solubility and instability in the gastrointestinal tract, in addition to rapid metabolism—most often hepatic—and low membrane penetration [187]. On the other hand, hepatic metabolism and the microbiota can contribute to increased therapeutic efficacy, producing more active metabolites, as we have already reported in another article published by our group [12]. In this context, two equally important aspects require further investigation: the identification of both inactive and more active metabolites, as well as the assessment of the therapeutic efficacy and reproducibility of data obtained in in vitro assays in in vivo models. Comprehensive studies are necessary to fully realize the potential of these plants.

5. Conclusions

The Brazilian mega-biodiversity is still a largely unexplored universe and a source of diverse bioactive compounds with promising pharmacological properties. These phytochemical compounds are potential candidates for the development and bioprospection of new therapeutic strategies that can be used to treat obesity and its metabolic sequelae. With all the reports described here, we have demonstrated the incredible potential that is available to be explored, with anti-inflammatory, antioxidant, anti-obesogenic, and antidiabetic properties, among others, which can contribute to the maintenance and restoration of metabolic health. Based on the evidence presented here, we aim to encourage the conduction and development of new research that allows the exploration of these still less-known plants, as well as emphasizing the need to raise nature consciousness towards biodiversity preservation while promoting scientific and technological development.

Funding

This work was supported by Portugal Foundation for Science and Technology (strategic projects UIDB/04539/2020, UIDP/04539/2020, and LA/P/0058/2020: CIBB). This work was also supported by Fundação de Apoio ao Desenvolvimento do Ensino, Ciência e Tecnologia do Estado de Mato Grosso do Sul (FUNDECT), Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES), and Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq).

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.

References

  1. WHO. Life Expectancy Increases by 5 Years, but Inequalities Persist. Available online: https://www.who.int/news/item/19-05-2016-life-expectancy-increased-by-5-years-since-2000-but-health-inequalities-persist (accessed on 8 May 2023).
  2. Monteiro-Alfredo, T.; Matafome, P.; Iacia, B.P.; Antunes, K.Á.; dos Santos, J.M.; da Silva Melo da Cunha, J.; Oliveira, S.; Oliveira, A.S.; Campos, J.F.; Magalhães, M.; et al. Acrocomia aculeata (Jacq.) Lodd. Ex Mart. Leaves Increase SIRT1 Levels and Improve Stress Resistance. Oxidative Med. Cell. Longev. 2020, 2020, e5238650. [Google Scholar] [CrossRef] [PubMed]
  3. Vatner, S.F.; Zhang, J.; Oydanich, M.; Berkman, T.; Naftalovich, R.; Vatner, D.E. Healthful Aging Mediated by Inhibition of Oxidative Stress. Ageing Res. Rev. 2020, 64, 101194. [Google Scholar] [CrossRef] [PubMed]
  4. WHO. World Health Organization—Background—The Global Burden of Chronic. Available online: https://www.who.int/nutrition/topics/2_background/en/ (accessed on 26 July 2019).
  5. Monteiro-Alfredo, T.; Caramelo, B.; Arbeláez, D.; Amaro, A.; Barra, C.; Silva, D.; Oliveira, S.; Seiça, R.; Matafome, P. Distinct Impact of Natural Sugars from Fruit Juices and Added Sugars on Caloric Intake, Body Weight, Glycaemia, Oxidative Stress and Glycation in Diabetic Rats. Nutrients 2021, 13, 2956. [Google Scholar] [CrossRef] [PubMed]
  6. Borghi-Silva, A.; Back, G.D.; Garcia de Araújo, A.S.; Oliveira, M.R.; da Luz Goulart, C.; Silva, R.N.; Bassi, D.; Mendes, R.G.; Arena, R. COVID-19 Seen from a Syndemic Perspective: Impact of Unhealthy Habits and Future Perspectives to Combat These Negative Interactions in Latin America. Prog. Cardiovasc. Dis. 2022, 71, 72–78. [Google Scholar] [CrossRef]
  7. Chu, D.-T.; Minh Nguyet, N.T.; Nga, V.T.; Thai Lien, N.V.; Vo, D.D.; Lien, N.; Nhu Ngoc, V.T.; Son, L.H.; Le, D.-H.; Nga, V.B.; et al. An Update on Obesity: Mental Consequences and Psychological Interventions. Diabetes Metab. Syndr. Clin. Res. Rev. 2019, 13, 155–160. [Google Scholar] [CrossRef]
  8. Safaei, M.; Sundararajan, E.A.; Driss, M.; Boulila, W.; Shapi’i, A. A Systematic Literature Review on Obesity: Understanding the Causes & Consequences of Obesity and Reviewing Various Machine Learning Approaches Used to Predict Obesity. Comput. Biol. Med. 2021, 136, 104754. [Google Scholar] [CrossRef]
  9. Srinivas, U.S.; Tan, B.W.Q.; Vellayappan, B.A.; Jeyasekharan, A.D. ROS and the DNA Damage Response in Cancer. Redox Biol. 2018, 25, 101084. [Google Scholar] [CrossRef] [PubMed]
  10. dos Santos, J.M.; Alfredo, T.M.; Antunes, K.Á.; da Cunha, J.d.S.M.; Costa, E.M.A.; Lima, E.S.; Silva, D.B.; Carollo, C.A.; Schmitz, W.O.; Boleti, A.P.d.A.; et al. Guazuma ulmifolia Lam. Decreases Oxidative Stress in Blood Cells and Prevents Doxorubicin-Induced Cardiotoxicity. Oxidative Med. Cell. Longev. 2018, 2018, e2935051. [Google Scholar] [CrossRef] [PubMed]
  11. Manna, P.; Jain, S.K. Obesity, Oxidative Stress, Adipose Tissue Dysfunction, and the Associated Health Risks: Causes and Therapeutic Strategies. Metab. Syndr. Relat. Disord. 2015, 13, 423–444. [Google Scholar] [CrossRef] [PubMed]
  12. Monteiro-Alfredo, T.; Oliveira, S.; Amaro, A.; Rosendo-Silva, D.; Antunes, K.; Pires, A.S.; Teixo, R.; Abrantes, A.M.; Botelho, M.F.; Castelo-Branco, M.; et al. Hypoglycaemic and Antioxidant Properties of Acrocomia aculeata (Jacq.) Lodd Ex Mart. Extract Are Associated with Better Vascular Function of Type 2 Diabetic Rats. Nutrients 2021, 13, 2856. [Google Scholar] [CrossRef] [PubMed]
  13. WHO. Obesity. Available online: https://www.who.int/health-topics/obesity (accessed on 13 April 2023).
  14. Nevill, A.M.; Duncan, M.J.; Myers, T. BMI Is Dead; Long Live Waist-Circumference Indices: But Which Index Should We Choose to Predict Cardio-Metabolic Risk? Nutr. Metab. Cardiovasc. Dis. 2022, 32, 1642–1650. [Google Scholar] [CrossRef]
  15. Elffers, T.W.; de Mutsert, R.; Lamb, H.J.; de Roos, A.; van Dijk, K.W.; Rosendaal, F.R.; Jukema, J.W.; Trompet, S. Body Fat Distribution, in Particular Visceral Fat, Is Associated with Cardiometabolic Risk Factors in Obese Women. PLoS ONE 2017, 12, e0185403. [Google Scholar] [CrossRef] [PubMed]
  16. Frank, A.P.; de Souza Santos, R.; Palmer, B.F.; Clegg, D.J. Determinants of Body Fat Distribution in Humans May Provide Insight about Obesity-Related Health Risks. J. Lipid Res. 2019, 60, 1710–1719. [Google Scholar] [CrossRef] [PubMed]
  17. Jensen, M.D. Role of Body Fat Distribution and the Metabolic Complications of Obesity. J. Clin. Endocrinol. Metab. 2008, 93, S57–S63. [Google Scholar] [CrossRef] [PubMed]
  18. Goossens, G.H. The Metabolic Phenotype in Obesity: Fat Mass, Body Fat Distribution, and Adipose Tissue Function. Obes. Facts 2017, 10, 207–215. [Google Scholar] [CrossRef]
  19. Finicelli, M.; Squillaro, T.; Di Cristo, F.; Di Salle, A.; Melone, M.A.B.; Galderisi, U.; Peluso, G. Metabolic Syndrome, Mediterranean Diet, and Polyphenols: Evidence and Perspectives. J. Cell. Physiol. 2019, 234, 5807–5826. [Google Scholar] [CrossRef]
  20. Hussain, T.; Tan, B.; Yin, Y.; Blachier, F.; Tossou, M.C.B.; Rahu, N. Oxidative Stress and Inflammation: What Polyphenols Can Do for Us? Oxidative Med. Cell. Longev. 2016, 2016, 7432797. [Google Scholar] [CrossRef] [PubMed]
  21. Pérez-Torres, I.; Castrejón-Téllez, V.; Soto, M.E.; Rubio-Ruiz, M.E.; Manzano-Pech, L.; Guarner-Lans, V. Oxidative Stress, Plant Natural Antioxidants, and Obesity. Int. J. Mol. Sci. 2021, 22, 1786. [Google Scholar] [CrossRef] [PubMed]
  22. WHO. World Health Organization—2011 the World Traditional Medicines Situation. In Traditional Medicines: Global Situation, Issues and Challenges; WHO: Geneva, Switzerland, 2011; Volume 3, pp. 1–14. [Google Scholar]
  23. Teng, Z.; Shen, Y. Research progress of genetic engineering on medicinal plants. Zhongguo Zhong Yao Za Zhi 2015, 40, 594–601. [Google Scholar]
  24. De Luca, V.; Salim, V.; Atsumi, S.M.; Yu, F. Mining the Biodiversity of Plants: A Revolution in the Making. Science 2012, 336, 1658–1661. [Google Scholar] [CrossRef] [PubMed]
  25. de Carvalho, J.T.G.; Da Silva Baldivia, D.; de Castro, D.T.H.; dos Santos, H.F.; dos Santos, C.M.; Oliveira, A.S.; Alfredo, T.M.; Vilharva, K.N.; de Picoli Souza, K.; dos Santos, E.L. The Immunoregulatory Function of Polyphenols: Implications in Cancer Immunity. J. Nutr. Biochem. 2020, 85, 108428. [Google Scholar] [CrossRef]
  26. Fonseca, C.R.; Venticinque, E.M. Biodiversity Conservation Gaps in Brazil: A Role for Systematic Conservation Planning. Nat. Conserv. 2018, 16, 61–67. [Google Scholar] [CrossRef]
  27. de Carvalho, A.P.A.; Conte-Junior, C.A. Health Benefits of Phytochemicals from Brazilian Native Foods and Plants: Antioxidant, Antimicrobial, Anti-Cancer, and Risk Factors of Metabolic/Endocrine Disorders Control. Trends Food Sci. Technol. 2021, 111, 534–548. [Google Scholar] [CrossRef]
  28. Dos Reis, S.O.; da Luz, T.C.; da Silva Couto, C.V.M.; Dalbó, J.; Nunes, L.d.C.; Martins, M.C.; Silva, P.I.; da Silva, A.M.A.; Trivilin, L.O. Juçara (Euterpe edulis Mart.) Supplementation Reduces Aberrant Crypt Foci and Increases SOD1 Expression in the Colorectal Mucosa of Carcinogenesis-Induced Rats. Nutr. Cancer 2020, 72, 610–619. [Google Scholar] [CrossRef] [PubMed]
  29. Seraglio, S.K.T.; Valese, A.C.; Daguer, H.; Bergamo, G.; Azevedo, M.S.; Nehring, P.; Gonzaga, L.V.; Fett, R.; Costa, A.C.O. Effect of in Vitro Gastrointestinal Digestion on the Bioaccessibility of Phenolic Compounds, Minerals, and Antioxidant Capacity of Mimosa Scabrella Bentham Honeydew Honeys. Food Res. Int. 2017, 99, 670–678. [Google Scholar] [CrossRef] [PubMed]
  30. Fang, J.; Zeng, L.; He, Y.; Liu, X.; Zhang, T.; Wang, Q. Effects of Dietary Tannic Acid on Obesity and Gut Microbiota in C57BL/6J Mice Fed with High-Fat Diet. Foods 2022, 11, 3325. [Google Scholar] [CrossRef] [PubMed]
  31. Manzoor, F.; Nisa, M.U.; Hussain, H.A.; Khan, M.K.; Ahmad, R.S.; Ahmad, N.; Imran, M.; Umbreen, H. Effect of Hydrolysable Tannin on Nutrient Intake Obesity and Other Associated Metabolic Risk Factors in Polycystic Rats. Transl. Med. Commun. 2021, 6, 10. [Google Scholar] [CrossRef]
  32. Khalil, H.E.; Abdelwahab, M.F.; Ibrahim, H.-I.M.; AlYahya, K.A.; Altaweel, A.A.; Alasoom, A.J.; Burshed, H.A.; Alshawush, M.M.; Waz, S. Cichoriin, a Biocoumarin, Mitigates Oxidative Stress and Associated Adverse Dysfunctions on High-Fat Diet-Induced Obesity in Rats. Life 2022, 12, 1731. [Google Scholar] [CrossRef]
  33. Singh, L.R.; Kumar, A.; Upadhyay, A.; Gupta, S.; Palanati, G.R.; Sikka, K.; Siddiqi, M.I.; Yadav, P.N.; Sashidhara, K.V. Discovery of Coumarin-Dihydroquinazolinone Analogs as Niacin Receptor 1 Agonist with in-Vivo Anti-Obesity Efficacy. Eur. J. Med. Chem. 2018, 152, 208–222. [Google Scholar] [CrossRef]
  34. Goto, T.; Takahashi, N.; Hirai, S.; Kawada, T. Various Terpenoids Derived from Herbal and Dietary Plants Function as PPAR Modulators and Regulate Carbohydrate and Lipid Metabolism. PPAR Res. 2010, 2010, 483958. [Google Scholar] [CrossRef] [PubMed]
  35. Song, H.; Shen, X.; Deng, R.; Zhang, Y.; Zheng, X. Dietary Anthocyanin-Rich Extract of Açai Protects from Diet-Induced Obesity, Liver Steatosis, and Insulin Resistance with Modulation of Gut Microbiota in Mice. Nutrition 2021, 86, 111176. [Google Scholar] [CrossRef]
  36. Xu, H.; Liu, M.; Liu, H.; Zhao, B.; Zheng, M.; Liu, J. Anthocyanins from Purple Corn Ameliorated Obesity in High Fat Diet-Induced Obese Mice through Activating Hepatic AMPK. J. Funct. Foods 2021, 84, 104582. [Google Scholar] [CrossRef]
  37. Abdel-Mottaleb, Y.; Ali, H.S.; El-Kherbetawy, M.K.; Elkazzaz, A.Y.; ElSayed, M.H.; Elshormilisy, A.; Eltrawy, A.H.; Abed, S.Y.; Alshahrani, A.M.; Hashish, A.A.; et al. Saponin-Rich Extract of Tribulus Terrestris Alleviates Systemic Inflammation and Insulin Resistance in Dietary Obese Female Rats: Impact on Adipokine/Hormonal Disturbances. Biomed. Pharmacother. 2022, 147, 112639. [Google Scholar] [CrossRef]
  38. Tafolla-Arellano, J.C.; González-León, A.; Tiznado-Hernández, M.E.; Zacarías García, L.; Báez-Sañudo, R. Composición, Fisiología y Biosíntesis de La Cutícula En Plantas. Rev. Fitotec. Mex. 2013, 36, 3–12. [Google Scholar] [CrossRef]
  39. Saad, B.; Ghareeb, B.; Kmail, A. Metabolic and Epigenetics Action Mechanisms of Antiobesity Medicinal Plants and Phytochemicals. Evid. Based Complement. Altern. Med. 2021, 2021, 9995903. [Google Scholar] [CrossRef]
  40. Francini-Pesenti, F.; Spinella, P.; Calò, L.A. Potential Role of Phytochemicals in Metabolic Syndrome Prevention and Therapy. Diabetes Metab. Syndr. Obes. 2019, 12, 1987–2002. [Google Scholar] [CrossRef]
  41. Speer, H.; D’Cunha, N.M.; Alexopoulos, N.I.; McKune, A.J.; Naumovski, N. Anthocyanins and Human Health-A Focus on Oxidative Stress, Inflammation and Disease. Antioxidants 2020, 9, 366. [Google Scholar] [CrossRef] [PubMed]
  42. Verpoorte, R. Exploration of Nature’s Chemodiversity: The Role of Secondary Metabolites as Leads in Drug Development. Drug Discov. Today 1998, 5, 232–238. [Google Scholar] [CrossRef]
  43. Jucá, M.M.; Cysne Filho, F.M.S.; de Almeida, J.C.; Mesquita, D.d.S.; Barriga, J.R.d.M.; Dias, K.C.F.; Barbosa, T.M.; Vasconcelos, L.C.; Leal, L.K.A.M.; Ribeiro, J.E.; et al. Flavonoids: Biological Activities and Therapeutic Potential. Nat. Prod. Res. 2020, 34, 692–705. [Google Scholar] [CrossRef]
  44. Araújo, J. Química de Alimentos—Teoria e Prática—7a Edição. Available online: https://www.agrolivros.com.br/nutricao-e-tecnologia-de-alimentos/livro-quimica-de-alimentos-teoria-e-pratica--p (accessed on 9 September 2019).
  45. Dias, M.C.; Pinto, D.C.G.A.; Silva, A.M.S. Plant Flavonoids: Chemical Characteristics and Biological Activity. Molecules 2021, 26, 5377. [Google Scholar] [CrossRef] [PubMed]
  46. Nunes, Â.A.; Buccini, D.F.; Jaques, J.A.S.; Portugal, L.C.; Guimarães, R.C.A.; Favaro, S.P.; Caldas, R.A.; Carvalho, C.M.E. Effect of Acrocomia aculeata Kernel Oil on Adiposity in Type 2 Diabetic Rats. Plant Foods Hum. Nutr. 2018, 73, 61–67. [Google Scholar] [CrossRef]
  47. Mir, S.A.; Shah, M.A.; Ganai, S.A.; Ahmad, T.; Gani, M. Understanding the Role of Active Components from Plant Sources in Obesity Management. J. Saudi Soc. Agric. Sci. 2019, 18, 168–176. [Google Scholar] [CrossRef]
  48. Patel, M.D.; Thompson, P.D. Phytosterols and Vascular Disease. Atherosclerosis 2006, 186, 12–19. [Google Scholar] [CrossRef]
  49. Tang, S.; Fang, C.; Liu, Y.; Tang, L.; Xu, Y. Anti-Obesity and Anti-Diabetic Effect of Ursolic Acid against Streptozotocin/High Fat Induced Obese in Diabetic Rats. J. Oleo Sci. 2022, 71, 289–300. [Google Scholar] [CrossRef] [PubMed]
  50. 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] [PubMed]
  51. Sharma, H.; Kumar, P.; Deshmukh, R.R.; Bishayee, A.; Kumar, S. Pentacyclic Triterpenes: New Tools to Fight Metabolic Syndrome. Phytomedicine 2018, 50, 166–177. [Google Scholar] [CrossRef] [PubMed]
  52. Liu, J.; Lee, J.; Salazar Hernandez, M.A.; Mazitschek, R.; Ozcan, U. Treatment of Obesity with Celastrol. Cell 2015, 161, 999–1011. [Google Scholar] [CrossRef] [PubMed]
  53. Garcia-Oliveira, P.; Fraga-Corral, M.; Pereira, A.G.; Lourenço-Lopes, C.; Jimenez-Lopez, C.; Prieto, M.A.; Simal-Gandara, J. Scientific Basis for the Industrialization of Traditionally Used Plants of the Rosaceae Family. Food Chem. 2020, 330, 127197. [Google Scholar] [CrossRef] [PubMed]
  54. Monteiro-Alfredo, T.; Maurino dos Santos, J.; Ávila Antunes, K.; Melo, J.; Botelho, W.; Boleti, A.P.; Pires, A.S.; Marques, I.; Abrantes, A.M.; Botelho, M.F.; et al. Acrocomia aculeata Associated with Doxorubicin: Cardioprotection and Anticancer Activity. Front. Pharmacol. 2023, 14, 1223933. [Google Scholar] [CrossRef] [PubMed]
  55. Figueiredo de Santana Aquino, D.; Monteiro, T.A.; Lima Cardoso, C.A.; Heredia Vieira, S.C.; Vieira, M.d.C.; de Picoli Souza, K.; Amaya-Farfan, J.; Borges Castro Carvalho, G.C.; Moura, C.S.; Morato, P.N. Investigation of the Antioxidant and Hypoglycemiant Properties of Alibertia edulis (L.C. Rich.) A.C. Rich. Leaves. J. Ethnopharmacol. 2020, 253, 112648. [Google Scholar] [CrossRef] [PubMed]
  56. Borges, P.R.S.; Edelenbos, M.; Larsen, E.; Hernandes, T.; Nunes, E.E.; de Barros Vilas Boas, E.V.; Pires, C.R.F. The Bioactive Constituents and Antioxidant Activities of Ten Selected Brazilian Cerrado Fruits. Food Chem. X 2022, 14, 100268. [Google Scholar] [CrossRef] [PubMed]
  57. Prado, L.G.; Arruda, H.S.; Peixoto Araujo, N.M.; de Oliveira Braga, L.E.; Banzato, T.P.; Pereira, G.A.; Figueiredo, M.C.; Ruiz, A.L.T.G.; Eberlin, M.N.; de Carvalho, J.E.; et al. Antioxidant, Antiproliferative and Healing Properties of Araticum (Annona crassiflora Mart.) Peel and Seed. Food Res. Int. 2020, 133, 109168. [Google Scholar] [CrossRef] [PubMed]
  58. Pereira, M.N.; Justino, A.B.; Martins, M.M.; Peixoto, L.G.; Vilela, D.D.; Santos, P.S.; Teixeira, T.L.; da Silva, C.V.; Goulart, L.R.; Pivatto, M.; et al. Stephalagine, an Alkaloid with Pancreatic Lipase Inhibitory Activity Isolated from the Fruit Peel of Annona crassiflora Mart. Ind. Crops Prod. 2017, 97, 324–329. [Google Scholar] [CrossRef]
  59. Justino, A.B.; Miranda, N.C.; Franco, R.R.; Martins, M.M.; da Silva, N.M.; Espindola, F.S. Annona muricata Linn. Leaf as a Source of Antioxidant Compounds with in Vitro Antidiabetic and Inhibitory Potential against α-Amylase, α-Glucosidase, Lipase, Non-Enzymatic Glycation and Lipid Peroxidation. Biomed. Pharmacother. 2018, 100, 83–92. [Google Scholar] [CrossRef]
  60. Olasehinde, O.R.; Afolabi, O.B.; Owolabi, O.V.; Akawa, A.B.; Omiyale, O.B. GC–MS Analysis of Phytochemical Constituents of Methanolic Fraction of Annona muricata Leaf and Its Inhibition against Two Key Enzymes Linked to Type II Diabetes. Sci. Afr. 2022, 16, e01178. [Google Scholar] [CrossRef]
  61. Boeing, J.S.; Ribeiro, D.; Chisté, R.C.; Visentainer, J.V.; Costa, V.M.; Freitas, M.; Fernandes, E. Chemical Characterization and Protective Effect of the Bactris setosa Mart. Fruit against Oxidative/Nitrosative Stress. Food Chem. 2017, 220, 427–437. [Google Scholar] [CrossRef]
  62. Dantas, M.B.V.C.; Júnior, O.R.P.; Campos, L.T.d.P.; Campos, É.G. Compounds of Tucum-Do-Cerrado (Bactris setosa) Fruit with Antioxidant Activity. Nat. Prod. Res. 2023, 37, 793–797. [Google Scholar] [CrossRef] [PubMed]
  63. Quaresma, D.M.O.; Justino, A.B.; Sousa, R.M.F.; Munoz, R.A.A.; de Aquino, F.J.T.; Martins, M.M.; Goulart, L.R.; Pivatto, M.; Espindola, F.S.; de Oliveira, A. Antioxidant Compounds from Banisteriopsis argyrophylla Leaves as α-Amylase, α-Glucosidase, Lipase, and Glycation Inhibitors. Bioorg. Chem. 2020, 105, 104335. [Google Scholar] [CrossRef] [PubMed]
  64. Malta, L.G.; Ghiraldini, F.G.; Reis, R.; Oliveira, M.d.V.; Silva, L.B.; Pastore, G.M. In Vivo Analysis of Antigenotoxic and Antimutagenic Properties of Two Brazilian Cerrado Fruits and the Identification of Phenolic Phytochemicals. Food Res. Int. 2012, 49, 604–611. [Google Scholar] [CrossRef]
  65. Ferreira, B.A.; da Silva, A.R.A.; Filbido, G.S.; Narita, I.M.P.; Pinheiro, A.P.d.O.; da Cruz e Silva, D.; Nascimento, E.; Villa, R.D.; de Oliveira, A.P. In Vitro Bioaccessibility of the Bioactive Compounds and Minerals in the Pulp and Peel of Buchenavia Tomentosa Eichler Fruits and Their Antioxidant Capacities. Meas. Food 2022, 8, 100064. [Google Scholar] [CrossRef]
  66. Roesler, R.; Catharino, R.R.; Malta, L.G.; Eberlin, M.N.; Pastore, G. Antioxidant Activity of Caryocar brasiliense (Pequi) and Characterization of Components by Electrospray Ionization Mass Spectrometry. Food Chem. 2008, 110, 711–717. [Google Scholar] [CrossRef]
  67. Caldeira, A.S.P.; Mbiakop, U.C.; Pádua, R.M.; van de Venter, M.; Matsabisa, M.G.; Campana, P.R.V.; Cortes, S.F.; Braga, F.C. Bioguided Chemical Characterization of Pequi (Caryocar brasiliense) Fruit Peels towards an Anti-Diabetic Activity. Food Chem. 2021, 345, 128734. [Google Scholar] [CrossRef] [PubMed]
  68. Giordani, M.A.; Collicchio, T.C.M.; Ascêncio, S.D.; de Oliveira Martins, D.T.; Balogun, S.O.; Bieski, I.G.C.; da Silva, L.A.; Colodel, E.M.; de Souza, R.L.; de Souza, D.L.P.; et al. Hydroethanolic Extract of the Inner Stem Bark of Cedrela Odorata Has Low Toxicity and Reduces Hyperglycemia Induced by an Overload of Sucrose and Glucose. J. Ethnopharmacol. 2015, 162, 352–361. [Google Scholar] [CrossRef] [PubMed]
  69. Oliveira-Alves, S.C.; Pereira, R.S.; Pereira, A.B.; Ferreira, A.; Mecha, E.; Silva, A.B.; Serra, A.T.; Bronze, M.R. Identification of Functional Compounds in Baru (Dipteryx alata Vog.) Nuts: Nutritional Value, Volatile and Phenolic Composition, Antioxidant Activity and Antiproliferative Effect. Food Res. Int. 2020, 131, 109026. [Google Scholar] [CrossRef]
  70. Lemos, M.R.B.; Siqueira, E.M.d.A.; Arruda, S.F.; Zambiazi, R.C. The Effect of Roasting on the Phenolic Compounds and Antioxidant Potential of Baru Nuts [Dipteryx alata Vog.]. Food Res. Int. 2012, 48, 592–597. [Google Scholar] [CrossRef]
  71. Siqueira, E.M.d.A.; Marin, A.M.F.; da Cunha, M.d.S.B.; Fustinoni, A.M.; de Sant’Ana, L.P.; Arruda, S.F. Consumption of Baru Seeds [Dipteryx alata Vog.], a Brazilian Savanna Nut, Prevents Iron-Induced Oxidative Stress in Rats. Food Res. Int. 2012, 45, 427–433. [Google Scholar] [CrossRef]
  72. Leite, N.R.; de Araújo, L.C.A.; Dos Santos da Rocha, P.; Agarrayua, D.A.; Ávila, D.S.; Carollo, C.A.; Silva, D.B.; Estevinho, L.M.; de Picoli Souza, K.; Dos Santos, E.L. Baru Pulp (Dipteryx alata Vogel): Fruit from the Brazilian Savanna Protects against Oxidative Stress and Increases the Life Expectancy of Caenorhabditis Elegans via SOD-3 and DAF-16. Biomolecules 2020, 10, 1106. [Google Scholar] [CrossRef]
  73. Outuki, P.M.; de Francisco, L.M.B.; Hoscheid, J.; Bonifácio, K.L.; Barbosa, D.S.; Cardoso, M.L.C. Development of Arabic and Xanthan Gum Microparticles Loaded with an Extract of Eschweilera nana Miers Leaves with Antioxidant Capacity. Colloids Surf. A Physicochem. Eng. Asp. 2016, 499, 103–112. [Google Scholar] [CrossRef]
  74. Donado-Pestana, C.M.; Belchior, T.; Genovese, M.I. Phenolic Compounds from Cagaita (Eugenia dysenterica DC.) Fruit Prevent Body Weight and Fat Mass Gain Induced by a High-Fat, High-Sucrose Diet. Food Res. Int. 2015, 77, 177–185. [Google Scholar] [CrossRef]
  75. Justino, A.B.; Guerra Silva, H.C.; Franco, R.R.; de Oliveira Cavalcante Pimentel, I.; Silva, N.F.; Saraiva, A.L.; Espindola, F.S. Flavonoids and Proanthocyanidins-Rich Fractions from Eugenia dysenterica Fruits and Leaves Inhibit the Formation of Advanced Glycation End-Products and the Activities of α-Amylase and α-Glucosidase. J. Ethnopharmacol. 2022, 285, 114902. [Google Scholar] [CrossRef]
  76. Neri-Numa, I.A.; Carvalho-Silva, L.B.; Morales, J.P.; Malta, L.G.; Muramoto, M.T.; Ferreira, J.E.M.; de Carvalho, J.E.; Ruiz, A.L.T.G.; Maróstica Junior, M.R.; Pastore, G.M. Evaluation of the Antioxidant, Antiproliferative and Antimutagenic Potential of Araçá-Boi Fruit (Eugenia stipitata Mc Vaugh—Myrtaceae) of the Brazilian Amazon Forest. Food Res. Int. 2013, 50, 70–76. [Google Scholar] [CrossRef]
  77. Carneiro, N.S.; Alves, C.C.F.; Alves, J.M.; Egea, M.B.; Martins, C.H.G.; Silva, T.S.; Bretanha, L.C.; Balleste, M.P.; Micke, G.A.; Silveira, E.V.; et al. Chemical Composition, Antioxidant and Antibacterial Activities of Essential Oils from Leaves and Flowers of Eugenia Klotzschiana Berg (Myrtaceae). An. Acad. Bras. Ciênc. 2017, 89, 1907–1915. [Google Scholar] [CrossRef] [PubMed]
  78. de Lima, J.P.; Fante, C.A.; Freitas Pires, C.R.; Nunes, E.E.; Alves, R.R.; de Siqueira Elias, H.H.; Nunes, C.A.; de Barros Vilas Boas, E.V. The Antioxidative Potential and Volatile Constituents of Mangaba Fruit over the Storage Period. Sci. Hortic. 2015, 194, 1–6. [Google Scholar] [CrossRef]
  79. da Silva, C.P.; Soares-Freitas, R.A.M.; Sampaio, G.R.; Santos, M.C.B.; do Nascimento, T.P.; Cameron, L.C.; Ferreira, M.S.L.; Arêas, J.A.G. Identification and Action of Phenolic Compounds of Jatobá-Do-Cerrado (Hymenaea stignocarpa Mart.) on α-Amylase and α-Glucosidase Activities and Flour Effect on Glycemic Response and Nutritional Quality of Breads. Food Res. Int. 2019, 116, 1076–1083. [Google Scholar] [CrossRef]
  80. dos Santos, K.P.; Sedano-Partida, M.D.; Sala-Carvalho, W.R.; Loureiro, B.O.S.J.; da Silva-Luz, C.L.; Furlan, C.M. Biological Activity of Hyptis Jacq. (Lamiaceae) Is Determined by the Environment. Ind. Crops Prod. 2018, 112, 705–715. [Google Scholar] [CrossRef]
  81. Justino, A.B.; Santana, E.C.; Franco, R.R.; Queiroz, J.S.; Silva, H.C.G.; de Lima, J.P.; Saraiva, A.L.; Martins, M.M.; Lemos de Morais, S.A.; de Oliveira, A.; et al. Antioxidant Compounds of Kielmeyera coriacea Mart. with α-Amylase, Lipase and Advanced Glycation End-Product Inhibitory Activities. J. Pharm. Biomed. Anal. 2021, 206, 114387. [Google Scholar] [CrossRef]
  82. Cândido, T.L.N.; Silva, M.R.; Agostini-Costa, T.S. Bioactive Compounds and Antioxidant Capacity of Buriti (Mauritia flexuosa L.f.) from the Cerrado and Amazon Biomes. Food Chem. 2015, 177, 313–319. [Google Scholar] [CrossRef] [PubMed]
  83. de Oliveira, D.M.; Siqueira, E.P.; Nunes, Y.R.F.; Cota, B.B. Flavonoids from Leaves of Mauritia flexuosa. Rev. Bras. Farmacogn. 2013, 23, 614–620. [Google Scholar] [CrossRef]
  84. Koolen, H.H.F.; da Silva, F.M.A.; Gozzo, F.C.; de Souza, A.Q.L.; de Souza, A.D.L. Antioxidant, Antimicrobial Activities and Characterization of Phenolic Compounds from Buriti (Mauritia flexuosa L. f.) by UPLC–ESI-MS/MS. Food Res. Int. 2013, 51, 467–473. [Google Scholar] [CrossRef]
  85. Leite, P.I.P.; Barreto, S.M.A.G.; Freitas, P.R.; de Araújo, A.C.J.; Paulo, C.L.R.; de Almeida, R.S.; de Assis, C.F.; Padilha, C.E.A.; Ferrari, M.; de Sousa Junior, F.C. Extraction of Bioactive Compounds from Buriti (Mauritia flexuosa L.) Fruit by Eco-Friendly Solvents: Chemical and Functional Characterization. Sustain. Chem. Pharm. 2021, 22, 100489. [Google Scholar] [CrossRef]
  86. Nobre, C.B.; Sousa, E.O.; Camilo, C.J.; Machado, J.F.; Silva, J.M.F.L.; Filho, J.R.; Coutinho, H.D.M.; Costa, J.G.M. Antioxidative Effect and Phytochemical Profile of Natural Products from the Fruits of “Babaçu” (Orbignia speciose) and “Buriti” (Mauritia flexuosa). Food Chem. Toxicol. 2018, 121, 423–429. [Google Scholar] [CrossRef]
  87. Resende, L.M.; Franca, A.S.; Oliveira, L.S. Buriti (Mauritia flexuosa L. f.) Fruit by-Products Flours: Evaluation as Source of Dietary Fibers and Natural Antioxidants. Food Chem. 2019, 270, 53–60. [Google Scholar] [CrossRef] [PubMed]
  88. Ribeiro, J.; Antunes, L.; Darin, J.; Mercadante, A.; Bianchi, M.D.L. Buriti (Mauritia flexuosa) Oil: Evaluation of the Mutagenic and Antimutagenic Potential by the Micronucleus Test in Vivo. Toxicol. Lett. 2010, 196, S163–S164. [Google Scholar] [CrossRef]
  89. Rudke, A.R.; Andrade, K.S.; Mazzutti, S.; Zielinski, A.A.F.; Alves, V.R.; Vitali, L.; Ferreira, S.R.S. A Comparative Study of Phenolic Compounds Profile and in Vitro Antioxidant Activity from Buriti (Mauritia flexuosa) by-Products Extracts. LWT 2021, 150, 111941. [Google Scholar] [CrossRef]
  90. de Souza, F.G.; Náthia-Neves, G.; de Araújo, F.F.; Dias Audibert, F.L.; Delafiori, J.; Neri-Numa, I.A.; Catharino, R.R.; de Alencar, S.M.; de Almeida Meireles, M.A.; Pastore, G.M. Evaluation of Antioxidant Capacity, Fatty Acid Profile, and Bioactive Compounds from Buritirana (Mauritiella armata Mart.) Oil: A Little-Explored Native Brazilian Fruit. Food Res. Int. 2021, 142, 110260. [Google Scholar] [CrossRef]
  91. Serpeloni, J.M.; Leal Specian, A.F.; Ribeiro, D.L.; Tuttis, K.; Vilegas, W.; Martínez-López, W.; Dokkedal, A.L.; Saldanha, L.L.; de Syllos Cólus, I.M.; Varanda, E.A. Antimutagenicity and Induction of Antioxidant Defense by Flavonoid Rich Extract of Myrcia bella Cambess. in Normal and Tumor Gastric Cells. J. Ethnopharmacol. 2015, 176, 345–355. [Google Scholar] [CrossRef]
  92. Duarte, I.; de Souza, M.C.M.; Curinga, R.M.; Mendonça, H.M.; de Oliveira, L.d.L.; Milenkovic, D.; Hassimotto, N.M.A.; Costa, A.M.; Malaquias, J.V.; Borges, T.K.d.S. Effect of Passiflora Setacea Juice and Its Phenolic Metabolites on Insulin Resistance Markers in Overweight Individuals and on Microglial Cell Activity. Food Funct. 2022, 13, 6498–6509. [Google Scholar] [CrossRef] [PubMed]
  93. Rodrigues, P.M.; Dutra Gomes, J.V.; Jamal, C.M.; Cunha Neto, Á.; Santos, M.L.; Fagg, C.W.; Fonseca-Bazzo, Y.M.; Magalhães, P.d.O.; de Sales, P.M.; Silveira, D. Triterpenes from Pouteria ramiflora (Mart.) Radlk. Leaves (Sapotaceae). Food Chem. Toxicol. 2017, 109, 1063–1068. [Google Scholar] [CrossRef]
  94. de Sales, P.M.; de Souza, P.M.; Dartora, M.; Resck, I.S.; Simeoni, L.A.; Fonseca-Bazzo, Y.M.; de Oliveira Magalhães, P.; Silveira, D. Pouteria Torta Epicarp as a Useful Source of α-Amylase Inhibitor in the Control of Type 2 Diabetes. Food Chem. Toxicol. 2017, 109, 962–969. [Google Scholar] [CrossRef] [PubMed]
  95. Medina, A.L.; Haas, L.I.R.; Chaves, F.C.; Salvador, M.; Zambiazi, R.C.; da Silva, W.P.; Nora, L.; Rombaldi, C.V. Araçá (Psidium cattleianum Sabine) Fruit Extracts with Antioxidant and Antimicrobial Activities and Antiproliferative Effect on Human Cancer Cells. Food Chem. 2011, 128, 916–922. [Google Scholar] [CrossRef]
  96. dos Santos da Rocha, P.; de Araújo Boleti, A.P.; do Carmo Vieira, M.; Carollo, C.A.; da Silva, D.B.; Estevinho, L.M.; dos Santos, E.L.; de Picoli Souza, K. Microbiological Quality, Chemical Profile as Well as Antioxidant and Antidiabetic Activities of Schinus Terebinthifolius Raddi. Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2019, 220, 36–46. [Google Scholar] [CrossRef]
  97. Olinto, S.C.F.; Calil-Silveira, J.; Dias, R.B.A.; Valle, M.M.R.; Serrano-Nascimento, C.; Rocha, P.S.; Monteiro-Alfredo, T.; dos Santos, E.L.; de Picoli Souza, K.; Nunes, M.T. Antiproliferative and Cytotoxic Effects of Schinus Terebinthifolia Leaf Extract on Thyroid Follicular Cells. Rev. Bras. Farmacogn. 2020, 30, 693–700. [Google Scholar] [CrossRef]
  98. de Britto Policarpi, P.; Turcatto, L.; Demoliner, F.; Ferrari, R.A.; Bascuñan, V.L.A.F.; Ramos, J.C.; Jachmanián, I.; Vitali, L.; Micke, G.A.; Block, J.M. Nutritional Potential, Chemical Profile and Antioxidant Activity of Chichá (Sterculia striata) Nuts and Its by-Products. Food Res. Int. 2018, 106, 736–744. [Google Scholar] [CrossRef]
  99. Pereira, A.P.A.; Angolini, C.F.F.; Paulino, B.N.; Lauretti, L.B.C.; Orlando, E.A.; Silva, J.G.S.; Neri-Numa, I.A.; Souza, J.D.R.P.; Pallone, J.A.L.; Eberlin, M.N.; et al. A Comprehensive Characterization of Solanum lycocarpum St. Hill and Solanum oocarpum Sendtn: Chemical Composition and Antioxidant Properties. Food Res. Int. 2019, 124, 61–69. [Google Scholar] [CrossRef]
  100. Franco, R.R.; Justino, A.B.; Martins, M.M.; Silva, C.G.; Campana, P.R.V.; Lopes, J.C.D.; De Almeida, V.L.; Espindola, F.S. Phytoscreening of Vochysiaceae Species: Molecular Identification by HPLC-ESI-MS/MS and Evaluating of Their Antioxidant Activity and Inhibitory Potential against Human α-Amylase and Protein Glycation. Bioorg. Chem. 2019, 91, 103122. [Google Scholar] [CrossRef]
  101. Campos, J.F.; de Castro, D.T.H.; Damião, M.J.; Vieira Torquato, H.F.; Paredes-Gamero, E.J.; Carollo, C.A.; Estevinho, L.M.; de Picoli Souza, K.; Dos Santos, E.L. The Chemical Profile of Senna velutina Leaves and Their Antioxidant and Cytotoxic Effects. Oxidative Med. Cell. Longev. 2016, 2016, 8405957. [Google Scholar] [CrossRef]
  102. Castro, D.T.H.; Campos, J.F.; Damião, M.J.; Torquato, H.F.V.; Paredes-Gamero, E.J.; Carollo, C.A.; Rodrigues, E.G.; de Picoli Souza, K.; dos Santos, E.L. Ethanolic Extract of Senna velutina Roots: Chemical Composition, In Vitro and In Vivo Antitumor Effects, and B16F10-Nex2 Melanoma Cell Death Mechanisms. Oxidative Med. Cell. Longev. 2019, 2019, e5719483. [Google Scholar] [CrossRef]
  103. Luna, E.M.; Freitas, T.S.; Campina, F.F.; Costa, M.S.; Rocha, J.E.; Cruz, R.P.; Sena Júnior, D.L.; Silveira, Z.S.; Macedo, N.S.; Pinheiro, J.C.A.; et al. Evaluation of Phytochemical Composition, Toxicity in Drosophila Melanogaster and Effects on Antibiotics Modulation of Plathymenia Reticulata Benth Extract. Toxicol. Rep. 2021, 8, 732–739. [Google Scholar] [CrossRef]
  104. Marinho, B.M.; Guimarães, V.H.D.; Sousa, J.N.; Moraes, D.S.; Gomes, E.S.B.; Vieira, C.R.; dos Reis, S.T.; Costa, T.d.O.; Farias, L.C.; Guimarães, A.L.S.; et al. Brazilian Cerrado Plant (Arnica) Lychnophora ericoides Mart. (Asteraceae) Toxicity Characterization in Mice. Phytomed. Plus 2022, 2, 100154. [Google Scholar] [CrossRef]
  105. Mendonça, L.a.B.M.; Matias, R.; Zanella, D.F.P.; Porto, K.R.A.; Guilhermino, J.F.; Moreira, D.L.; Roel, A.R.; Pott, A.; Carvalho, C.M.E. Toxicity and Phytochemistry of Eight Species Used in the Traditional Medicine of Sul-Mato-Grossense, Brazil. Braz. J. Biol. 2020, 80, 574–581. [Google Scholar] [CrossRef] [PubMed]
  106. Palozi, R.A.C.; Guarnier, L.P.; Romão, P.V.M.; Nocchi, S.R.; dos Santos, C.C.; Lourenço, E.L.B.; Silva, D.B.; Gasparotto, F.M.; Gasparotto Junior, A. Pharmacological Safety of Plinia cauliflora (Mart.) Kausel in Rabbits. Toxicol. Rep. 2019, 6, 616–624. [Google Scholar] [CrossRef] [PubMed]
  107. Traesel, G.K.; de Souza, J.C.; de Barros, A.L.; Souza, M.A.; Schmitz, W.O.; Muzzi, R.M.; Oesterreich, S.A.; Arena, A.C. Acute and Subacute (28 Days) Oral Toxicity Assessment of the Oil Extracted from Acrocomia aculeata Pulp in Rats. Food Chem. Toxicol. 2014, 74, 320–325. [Google Scholar] [CrossRef] [PubMed]
  108. Frenzel, C.; Teschke, R. Herbal Hepatotoxicity: Clinical Characteristics and Listing Compilation. Int. J. Mol. Sci. 2016, 17, 588. [Google Scholar] [CrossRef]
  109. Murphy, R.; Carroll, R.W.; Krebs, J.D. Pathogenesis of the Metabolic Syndrome: Insights from Monogenic Disorders. Mediat. Inflamm. 2013, 2013, e920214. [Google Scholar] [CrossRef]
  110. Rochlani, Y.; Pothineni, N.V.; Kovelamudi, S.; Mehta, J.L. Metabolic Syndrome: Pathophysiology, Management, and Modulation by Natural Compounds. Ther. Adv. Cardiovasc. Dis. 2017, 11, 215–225. [Google Scholar] [CrossRef]
  111. Rodrigues, T.; Matafome, P.; Santos-Silva, D.; Sena, C.; Seiça, R. Reduction of Methylglyoxal-Induced Glycation by Pyridoxamine Improves Adipose Tissue Microvascular Lesions. Available online: https://www.hindawi.com/journals/jdr/2013/690650/ (accessed on 8 February 2021).
  112. Teixeira, G.P.; Faria, R.X. Influence of Purinergic Signaling on Glucose Transporters: A Possible Mechanism against Insulin Resistance? Eur. J. Pharmacol. 2021, 892, 173743. [Google Scholar] [CrossRef]
  113. Saadeldeen, F.S.A.; Niu, Y.; Wang, H.; Zhou, L.; Meng, L.; Chen, S.; Sun-Waterhouse, D.; Waterhouse, G.I.N.; Liu, Z.; Kang, W. Natural Products: Regulating Glucose Metabolism and Improving Insulin Resistance. Food Sci. Hum. Wellness 2020, 9, 214–228. [Google Scholar] [CrossRef]
  114. Rodrigues, T.; Matafome, P.; Sereno, J.; Almeida, J.; Castelhano, J.; Gamas, L.; Neves, C.; Gonçalves, S.; Carvalho, C.; Arslanagic, A.; et al. Methylglyoxal-Induced Glycation Changes Adipose Tissue Vascular Architecture, Flow and Expansion, Leading to Insulin Resistance. Sci. Rep. 2017, 7, 1698. [Google Scholar] [CrossRef] [PubMed]
  115. Saad, B. Prevention and Treatment of Obesity-Related Inflammatory Diseases by Edible and Medicinal Plants and Their Active Compounds. Immuno 2022, 2, 609–629. [Google Scholar] [CrossRef]
  116. Boonchaya-anant, P.; Apovian, C.M. Metabolically Healthy Obesity—Does It Exist? Curr. Atheroscler. Rep. 2014, 16, 441. [Google Scholar] [CrossRef]
  117. Rutkowski, J.M.; Davis, K.E.; Scherer, P.E. Mechanisms of Obesity and Related Pathologies: The Macro- and Microcirculation of Adipose Tissue. FEBS J. 2009, 276, 5738–5746. [Google Scholar] [CrossRef] [PubMed]
  118. Bakker, S.J.; IJzerman, R.G.; Teerlink, T.; Westerhoff, H.V.; Gans, R.O.; Heine, R.J. Cytosolic Triglycerides and Oxidative Stress in Central Obesity: The Missing Link between Excessive Atherosclerosis, Endothelial Dysfunction, and Beta-Cell Failure? Atherosclerosis 2000, 148, 17–21. [Google Scholar] [CrossRef] [PubMed]
  119. Russell, A.P.; Gastaldi, G.; Bobbioni-Harsch, E.; Arboit, P.; Gobelet, C.; Dériaz, O.; Golay, A.; Witztum, J.L.; Giacobino, J.-P. Lipid Peroxidation in Skeletal Muscle of Obese as Compared to Endurance-Trained Humans: A Case of Good vs. Bad Lipids? FEBS Lett. 2003, 551, 104–106. [Google Scholar] [CrossRef] [PubMed]
  120. Martínez, J.A. Mitochondrial Oxidative Stress and Inflammation: An Slalom to Obesity and Insulin Resistance. J. Physiol. Biochem. 2006, 62, 303–306. [Google Scholar] [CrossRef] [PubMed]
  121. Mastrototaro, L.; Roden, M. Insulin Resistance and Insulin Sensitizing Agents. Metabolism 2021, 125, 154892. [Google Scholar] [CrossRef] [PubMed]
  122. Gooda Sahib, N.; Saari, N.; Ismail, A.; Khatib, A.; Mahomoodally, F.; Abdul Hamid, A. Plants’ Metabolites as Potential Antiobesity Agents. Sci. World J. 2012, 2012, e436039. [Google Scholar] [CrossRef]
  123. Oliveira, V.B.; Ferreira, A.V.M.; Oliveira, M.C.; Teixeira, M.M.; Brandão, M.G.L. Effects of Xylopia aromatica (Lam.) Mart. Fruit on Metabolic and Inflammatory Dysfunction Induced by High Refined Carbohydrate-Containing-Diet in Mice. Food Res. Int. 2014, 62, 541–550. [Google Scholar] [CrossRef]
  124. Frediani Brant, N.M.; Mourão Gasparotto, F.; de Oliveira Araújo, V.; Christian Maraschin, J.; Lima Ribeiro, R.d.C.; Botelho Lourenço, E.L.; Cardozo Junior, E.L.; Gasparotto Junior, A. Cardiovascular Protective Effects of Casearia Sylvestris Swartz in Swiss and C57BL/6 LDLr-Null Mice Undergoing High Fat Diet. J. Ethnopharmacol. 2014, 154, 419–427. [Google Scholar] [CrossRef]
  125. Sousa, J.N.; Mafra, V.; Marinho, B.M.; Guimarães, V.H.D.; Oliveira, L.P.; dos Reis, S.T.; Costa, T.; Vieira, C.R.; de Paula, A.M.B.; Guimarães, A.L.S.; et al. Oral Treatment with Davilla elliptica A. St,-Hil. Leaves Improves Liver Steatosis and Lipid Metabolism on a Diet-Induced Obese Mice Model. Phytomed. Plus 2021, 1, 100130. [Google Scholar] [CrossRef]
  126. Brisson, D.; Larouche, M.; Chebli, J.; Khoury, E.; Gaudet, D. Correlation between Chylomicronemia Diagnosis Scores and Post-Heparin Lipoprotein Lipase Activity. Clin. Biochem. 2023, 114, 67–72. [Google Scholar] [CrossRef]
  127. Holmbäck, U.; Forslund, A.; Grudén, S.; Alderborn, G.; Söderhäll, A.; Hellström, P.M.; Lennernäs, H. Effects of a Novel Combination of Orlistat and Acarbose on Tolerability, Appetite, and Glucose Metabolism in Persons with Obesity. Obes. Sci. Pract. 2020, 6, 313–323. [Google Scholar] [CrossRef]
  128. Pu, Y.; Chen, L.; He, X.; Cao, J.; Jiang, W. Soluble Polysaccharides Decrease Inhibitory Activity of Banana Condensed Tannins against Porcine Pancreatic Lipase. Food Chem. 2023, 418, 136013. [Google Scholar] [CrossRef]
  129. Veloso, C.d.C.; de Oliveira, M.C.; Oliveira, C.d.C.; Rodrigues, V.G.; Giusti-Paiva, A.; Teixeira, M.M.; Duarte, I.D.; Ferreira, A.V.M.; Perez, A.d.C. Hydroethanolic Extract of Pyrostegia venusta (Ker Gawl.) Miers Flowers Improves Inflammatory and Metabolic Dysfunction Induced by High-Refined Carbohydrate Diet. J. Ethnopharmacol. 2014, 151, 722–728. [Google Scholar] [CrossRef]
  130. Donado-Pestana, C.M.; dos Santos-Donado, P.R.; Daza, L.D.; Belchior, T.; Festuccia, W.T.; Genovese, M.I. Cagaita Fruit (Eugenia dysenterica DC.) and Obesity: Role of Polyphenols on Already Established Obesity. Food Res. Int. 2018, 103, 40–47. [Google Scholar] [CrossRef] [PubMed]
  131. Napolitano, D.R.; Mineo, J.R.; de Souza, M.A.; de Paula, J.E.; Espindola, L.S.; Espindola, F.S. Down-Modulation of Nitric Oxide Production in Murine Macrophages Treated with Crude Plant Extracts from the Brazilian Cerrado. J. Ethnopharmacol. 2005, 99, 37–41. [Google Scholar] [CrossRef]
  132. Semple, R.K.; Chatterjee, V.K.K.; O’Rahilly, S. PPAR Gamma and Human Metabolic Disease. J. Clin. Investig. 2006, 116, 581–589. [Google Scholar] [CrossRef]
  133. Vanden Berghe, W.; Vermeulen, L.; Delerive, P.; De Bosscher, K.; Staels, B.; Haegeman, G. A Paradigm for Gene Regulation: Inflammation, NF-κB and PPAR. Adv. Exp. Med. Biol. 2003, 544, 181–196. [Google Scholar] [CrossRef] [PubMed]
  134. Wang, Y.-X.; Lee, C.-H.; Tiep, S.; Yu, R.T.; Ham, J.; Kang, H.; Evans, R.M. Peroxisome-Proliferator-Activated Receptor Delta Activates Fat Metabolism to Prevent Obesity. Cell 2003, 113, 159–170. [Google Scholar] [CrossRef]
  135. Gomes-Copeland, K.K.P.; Meireles, C.G.; Gomes, J.V.D.; Torres, A.G.; Sinoti, S.B.P.; Fonseca-Bazzo, Y.M.; Magalhães, P.d.O.; Fagg, C.W.; Simeoni, L.A.; Silveira, D. Hippeastrum stapfianum (Kraenzl.) R.S.Oliveira & Dutilh (Amaryllidaceae) Ethanol Extract Activity on Acetylcholinesterase and PPAR-α/γ Receptors. Plants 2022, 11, 3179. [Google Scholar] [CrossRef] [PubMed]
  136. Hithamani, G.; Ganesan, P. Polyphenols from Indian Cereal Grains Inhibit 3T3-L1 Adipogenesis through Modulating Early and Late Phase Adipogenic Markers. Food Biosci. 2022, 50, 102075. [Google Scholar] [CrossRef]
  137. Li, K.K.; Peng, J.M.; Zhu, W.; Cheng, B.H.; Li, C.M. Gallocatechin Gallate (GCG) Inhibits 3T3-L1 Differentiation and Lipopolysaccharide Induced Inflammation through MAPK and NF-κB Signaling. J. Funct. Foods 2017, 30, 159–167. [Google Scholar] [CrossRef]
  138. Bartelt, A.; Heeren, J. Adipose Tissue Browning and Metabolic Health. Nat. Rev. Endocrinol. 2014, 10, 24–36. [Google Scholar] [CrossRef] [PubMed]
  139. Roth, C.L.; Molica, F.; Kwak, B.R. Browning of White Adipose Tissue as a Therapeutic Tool in the Fight against Atherosclerosis. Metabolites 2021, 11, 319. [Google Scholar] [CrossRef] [PubMed]
  140. Ma, P.-Y.; Li, X.-Y.; Wang, Y.-L.; Lang, D.-Q.; Liu, L.; Yi, Y.-K.; Liu, Q.; Shen, C.-Y. Natural Bioactive Constituents from Herbs and Nutraceuticals Promote Browning of White Adipose Tissue. Pharmacol. Res. 2022, 178, 106175. [Google Scholar] [CrossRef] [PubMed]
  141. Alves-Santos, A.M.; Fernandes, D.C.; Naves, M.M.V. Baru (Dipteryx alata Vog.) Fruit as an Option of Nut and Pulp with Advantageous Nutritional and Functional Properties: A Comprehensive Review. NFS J. 2021, 24, 26–36. [Google Scholar] [CrossRef]
  142. Mattes, R.D.; Kris-Etherton, P.M.; Foster, G.D. Impact of Peanuts and Tree Nuts on Body Weight and Healthy Weight Loss in Adults12. J. Nutr. 2008, 138, 1741S–1745S. [Google Scholar] [CrossRef]
  143. Dall’Agnol, R.; Lino von Poser, G. The Use of Complex Polysaccharides in the Management of Metabolic Diseases: The Case of Solanum lycocarpum Fruits. J. Ethnopharmacol. 2000, 71, 337–341. [Google Scholar] [CrossRef]
  144. Mayumi Usuda Prado Rocha, D.; Paula Silva Caldas, A.; Simões e Silva, A.C.; Bressan, J.; Hermana Miranda Hermsdorff, H. Nut-Enriched Energy Restricted Diet Has Potential to Decrease Hunger in Women at Cardiometabolic Risk: A Randomized Controlled Trial (Brazilian Nuts Study). Nutr. Res. 2023, 109, 35–46. [Google Scholar] [CrossRef] [PubMed]
  145. Torres, L.R.d.O.; de Santana, F.C.; Torres-Leal, F.L.; de Melo, I.L.P.; Yoshime, L.T.; Matos-Neto, E.M.; Seelaender, M.C.L.; Araújo, C.M.M.; Cogliati, B.; Mancini-Filho, J. Pequi (Caryocar brasiliense Camb.) Almond Oil Attenuates Carbon Tetrachloride-Induced Acute Hepatic Injury in Rats: Antioxidant and Anti-Inflammatory Effects. Food Chem. Toxicol. 2016, 97, 205–216. [Google Scholar] [CrossRef]
  146. Fowler, M.J. Microvascular and Macrovascular Complications of Diabetes. Clin. Diabetes 2008, 26, 77–82. [Google Scholar] [CrossRef]
  147. WHO. Cardiovascular Diseases. Available online: https://www.who.int/westernpacific/health-topics/cardiovascular-diseases (accessed on 11 October 2021).
  148. Viigimaa, M.; Sachinidis, A.; Toumpourleka, M.; Koutsampasopoulos, K.; Alliksoo, S.; Titma, T. Macrovascular Complications of Type 2 Diabetes Mellitus. Curr. Vasc. Pharmacol. 2020, 18, 110–116. [Google Scholar] [CrossRef]
  149. Mohamed, S.M.; Shalaby, M.A.; El-Shiekh, R.A.; El-Banna, H.A.; Emam, S.R.; Bakr, A.F. Metabolic Syndrome: Risk Factors, Diagnosis, Pathogenesis, and Management with Natural Approaches. Food Chem. Adv. 2023, 3, 100335. [Google Scholar] [CrossRef]
  150. Olasehinde, O.R.; Afolabi, O.B. Identification of Bioactive Constituents of Chloroform Fraction from Annona muricata Leaf, Its Antioxidant Activity and Inhibitory Potential against Carbohydrate-Hydrolyzing α-Amylase and α-Glucosidase Activities Linked to Type II Diabetes Mellitus: In Vitro Study. J. Herbmed. Pharmacol. 2022, 12, 100–108. [Google Scholar] [CrossRef]
  151. Ali Asgar, M. Anti-Diabetic Potential of Phenolic Compounds: A Review. Int. J. Food Prop. 2013, 16, 91–103. [Google Scholar] [CrossRef]
  152. Pinto, L.C.; Cerqueira-Lima, A.T.; Suzarth, S.d.S.; de Souza, R.; Tosta, B.R.; da Silva, H.B.; Pires, A.d.O.; Queiroz, G.d.A.; Teixeira, T.O.; Dourado, K.M.C.; et al. Anonna muricata L. (Soursop) Seed Oil Improves Type 1 Diabetes Parameters in Vivo and in Vitro. PharmaNutrition 2018, 6, 1–8. [Google Scholar] [CrossRef]
  153. Florence, N.T.; Benoit, M.Z.; Jonas, K.; Alexandra, T.; Désiré, D.D.P.; Pierre, K.; Théophile, D. Antidiabetic and Antioxidant Effects of Annona muricata (Annonaceae), Aqueous Extract on Streptozotocin-Induced Diabetic Rats. J. Ethnopharmacol. 2014, 151, 784–790. [Google Scholar] [CrossRef]
  154. Nunes, Â.A.; Buccini, D.F.; dos Santos Jaques, J.A.; Portugal, L.C.; Guimarães, R.C.A.; Favaro, S.P.; de Araújo Caldas, R.; Carvalho, C.M.E. Effect of Dietary Acrocomia aculeata Kernel Oil Rich in Medium Chain Fatty Acids on Type 2 Diabetic Rats. J. Funct. Foods 2020, 75, 104295. [Google Scholar] [CrossRef]
  155. da Silva, P.V.B.; Ramiro, M.M.; Iriguchi, E.K.K.; Corrêa, W.A.; Lowe, J.; Cardoso, C.A.L.; Arena, A.C.; Kassuya, C.A.L.; Muzzi, R.M. Antidiabetic, Cytotoxic and Antioxidant Activities of Oil Extracted from Acrocomia aculeata Pulp. Nat. Prod. Res. 2019, 33, 2413–2416. [Google Scholar] [CrossRef]
  156. Gomes, J.H.d.S.; Mbiakop, U.C.; Oliveira, R.L.; Stehmann, J.R.; de Pádua, R.M.; Cortes, S.F.; Braga, F.C. Polyphenol-Rich Extract and Fractions of Terminalia phaeocarpa Eichler Possess Hypoglycemic Effect, Reduce the Release of Cytokines, and Inhibit Lipase, α-Glucosidase, and α-Amilase Enzymes. J. Ethnopharmacol. 2021, 271, 113847. [Google Scholar] [CrossRef] [PubMed]
  157. Bastos, R.G.; Salles, B.C.C.; Bini, I.F.; Castaldini, L.P.; Silva, L.C.D.; Vilela, A.A.; Micheloni, A.L.C.; da Silva, G.M.; da Silva, P.H.C.; Maure, A.K.; et al. Phytochemical Composition, Antioxidant and in Vivo Antidiabetic Activities of the Hydroethanolic Extract of Eugenia florida DC. (Myrtaceae) Leaves. South Afr. J. Bot. 2019, 123, 317–332. [Google Scholar] [CrossRef]
  158. Mbiakop, U.C.; Gomes, J.H.S.; Pádua, R.M.; Lemos, V.S.; Braga, F.C.; Cortes, S.F. Oral Sub-Chronic Treatment with Terminalia phaeocarpa Eichler (Combretaceae) Reduces Liver PTP1B Activity in a Murine Model of Diabetes. J. Ethnopharmacol. 2023, 306, 116164. [Google Scholar] [CrossRef]
  159. e Silva, A.L.L.; dos Santos, D.C.; de Sousa, T.L.; Silva, F.G.; Egea, M.B. “Cerrado” Cashew (Anacardium othonianum Rizz.) Juice Improves Metabolic Parameters in Women: A Pilot Study. J. Funct. Foods 2020, 69, 103950. [Google Scholar] [CrossRef]
  160. Talpo, T.C.; Motta, B.P.; de Oliveira, J.O.; Figueiredo, I.D.; Pinheiro, C.G.; dos Santos, C.H.C.; de Carvalho, M.G.; Brunetti, I.L.; Baviera, A.M. Siolmatra Brasiliensis Stem Extract Ameliorates Antioxidant Defenses and Mitigates Glycoxidative Stress in Mice with High-Fat Diet-Induced Obesity. Obes. Res. Clin. Pract. 2022, 16, 130–137. [Google Scholar] [CrossRef]
  161. de Araujo, R.L.; Tomás-Barberán, F.A.; dos Santos, R.F.; Martinez-Blazquez, J.A.; Genovese, M.I. Postprandial Glucose-Lowering Effect of Cagaita (Eugenia dysenterica DC) Fruit Juice in Dysglycemic Subjects with Metabolic Syndrome: An Exploratory Study. Food Res. Int. 2021, 142, 110209. [Google Scholar] [CrossRef] [PubMed]
  162. de Souza, V.R.; Pereira, P.A.P.; Queiroz, F.; Borges, S.V.; de Deus Souza Carneiro, J. Determination of Bioactive Compounds, Antioxidant Activity and Chemical Composition of Cerrado Brazilian Fruits. Food Chem. 2012, 134, 381–386. [Google Scholar] [CrossRef]
  163. Morais, M.G.; Saldanha, A.A.; Azevedo, L.S.; Mendes, I.C.; Rodrigues, J.P.C.; Amado, P.A.; Farias, K.d.S.; Zanuncio, V.S.S.; Cassemiro, N.S.; da Silva, D.B.; et al. Antioxidant and Anti-Inflammatory Effects of Fractions from Ripe Fruits of Solanum lycocarpum St. Hil. (Solanaceae) and Putative Identification of Bioactive Compounds by GC–MS and LC-DAD-MS. Food Res. Int. 2022, 156, 111145. [Google Scholar] [CrossRef] [PubMed]
  164. Neves, C.; Rodrigues, T.; Sereno, J.; Simões, C.; Castelhano, J.; Gonçalves, J.; Bento, G.; Gonçalves, S.; Seiça, R.; Domingues, M.R.; et al. Dietary Glycotoxins Impair Hepatic Lipidemic Profile in Diet-Induced Obese Rats Causing Hepatic Oxidative Stress and Insulin Resistance. Oxidative Med. Cell. Longev. 2019, 2019, e6362910. [Google Scholar] [CrossRef]
  165. Al-Aubaidy, H.A.; Jelinek, H.F. Oxidative DNA Damage and Obesity in Type 2 Diabetes Mellitus. Eur. J. Endocrinol. 2011, 164, 899–904. [Google Scholar] [CrossRef]
  166. Bao, W.; Song, F.; Li, X.; Rong, S.; Yang, W.; Zhang, M.; Yao, P.; Hao, L.; Yang, N.; Hu, F.B.; et al. Plasma Heme Oxygenase-1 Concentration Is Elevated in Individuals with Type 2 Diabetes Mellitus. PLoS ONE 2010, 5, e12371. [Google Scholar] [CrossRef]
  167. Chandrakumar, L.; Bagyánszki, M.; Szalai, Z.; Mezei, D.; Bódi, N. Diabetes-Related Induction of the Heme Oxygenase System and Enhanced Colocalization of Heme Oxygenase 1 and 2 with Neuronal Nitric Oxide Synthase in Myenteric Neurons of Different Intestinal Segments. Oxidative Med. Cell. Longev. 2017, 2017, e1890512. [Google Scholar] [CrossRef]
  168. Dong, Q.Y.; Cui, Y.; Chen, L.; Song, J.; Sun, L. Urinary 8-Hydroxydeoxyguanosine Levels in Diabetic Retinopathy Patients. Eur. J. Ophthalmol. 2008, 18, 94–98. [Google Scholar] [CrossRef]
  169. Pradeep, A.R.; Agarwal, E.; Bajaj, P.; Rao, N.S. 4-Hydroxy-2-Nonenal, an Oxidative Stress Marker in Crevicular Fluid and Serum in Type 2 Diabetes with Chronic Periodontitis. Contemp. Clin. Dent. 2013, 4, 281–285. [Google Scholar] [CrossRef]
  170. Toyokuni, S.; Yamada, S.; Kashima, M.; Ihara, Y.; Yamada, Y.; Tanaka, T.; Hiai, H.; Seino, Y.; Uchida, K. Serum 4-Hydroxy-2-Nonenal-Modified Albumin Is Elevated in Patients with Type 2 Diabetes Mellitus. Antioxid. Redox Signal. 2000, 2, 681–685. [Google Scholar] [CrossRef] [PubMed]
  171. Giacco, F.; Brownlee, M. Oxidative Stress and Diabetic Complications. Circ Res 2010, 107, 1058–1070. [Google Scholar] [CrossRef]
  172. Brownlee, M. Biochemistry and Molecular Cell Biology of Diabetic Complications. Nature 2001, 414, 813–820. [Google Scholar] [CrossRef]
  173. Okeleji, L.O.; Ajayi, A.F.; Adebayo-Gege, G.; Aremu, V.O.; Adebayo, O.I.; Adebayo, E.T. Epidemiologic Evidence Linking Oxidative Stress and Pulmonary Function in Healthy Populations. Chronic Dis. Transl. Med. 2021, 7, 88–99. [Google Scholar] [CrossRef]
  174. Barbosa, K.B.F.; Costa, N.M.B.; Alfenas, R.d.C.G.; De Paula, S.O.; Minim, V.P.R.; Bressan, J. Estresse oxidativo: Conceito, implicações e fatores modulatórios. Rev. Nutr. 2010, 23, 629–643. [Google Scholar] [CrossRef]
  175. Neha, K.; Haider, M.R.; Pathak, A.; Yar, M.S. Medicinal Prospects of Antioxidants: A Review. Eur. J. Med. Chem. 2019, 178, 687–704. [Google Scholar] [CrossRef] [PubMed]
  176. Fasseas, M.K.; Mountzouris, K.C.; Tarantilis, P.A.; Polissiou, M.; Zervas, G. Antioxidant Activity in Meat Treated with Oregano and Sage Essential Oils. Food Chem. 2008, 106, 1188–1194. [Google Scholar] [CrossRef]
  177. Furukawa, S.; Fujita, T.; Shimabukuro, M.; Iwaki, M.; Yamada, Y.; Nakajima, Y.; Nakayama, O.; Makishima, M.; Matsuda, M.; Shimomura, I. Increased Oxidative Stress in Obesity and Its Impact on Metabolic Syndrome. J. Clin. Investig. 2004, 114, 1752–1761. [Google Scholar] [CrossRef] [PubMed]
  178. Lee, H.; Lee, Y.J.; Choi, H.; Ko, E.H.; Kim, J.-W. Reactive Oxygen Species Facilitate Adipocyte Differentiation by Accelerating Mitotic Clonal Expansion. J. Biol. Chem. 2009, 284, 10601–10609. [Google Scholar] [CrossRef]
  179. Kaneto, H.; Matsuoka, T. Involvement of Oxidative Stress in Suppression of Insulin Biosynthesis under Diabetic Conditions. Int. J. Mol. Sci. 2012, 13, 13680–13690. [Google Scholar] [CrossRef]
  180. Hang, Y.; Yamamoto, T.; Benninger, R.K.P.; Brissova, M.; Guo, M.; Bush, W.; Piston, D.W.; Powers, A.C.; Magnuson, M.; Thurmond, D.C.; et al. The MafA Transcription Factor Becomes Essential to Islet β-Cells Soon After Birth. Diabetes 2014, 63, 1994–2005. [Google Scholar] [CrossRef]
  181. Zhang, C.; Moriguchi, T.; Kajihara, M.; Esaki, R.; Harada, A.; Shimohata, H.; Oishi, H.; Hamada, M.; Morito, N.; Hasegawa, K.; et al. MafA Is a Key Regulator of Glucose-Stimulated Insulin Secretion. Mol. Cell. Biol. 2005, 25, 4969–4976. [Google Scholar] [CrossRef] [PubMed]
  182. Matafome, P.; Sena, C.; Seiça, R. Methylglyoxal, Obesity, and Diabetes. Endocrine 2013, 43, 472–484. [Google Scholar] [CrossRef] [PubMed]
  183. Monteiro-Alfredo, T.; Matafome, P. Gut Metabolism of Sugars: Formation of Glycotoxins and Their Intestinal Absorption. Diabetology 2022, 3, 596–605. [Google Scholar] [CrossRef]
  184. Bianchi, M.d.L.P.; Antunes, L.M.G. Radicais livres e os principais antioxidantes da dieta. Rev. Nutr. 1999, 12, 123–130. [Google Scholar] [CrossRef]
  185. de Giffoni de Carvalho, J.T.; da Silva Baldivia, D.; Leite, D.F.; de Araújo, L.C.A.; de Toledo Espindola, P.P.; Antunes, K.A.; Rocha, P.S.; de Picoli Souza, K.; dos Santos, E.L. Medicinal Plants from Brazilian Cerrado: Antioxidant and Anticancer Potential and Protection against Chemotherapy Toxicity. Oxidative Med. Cell. Longev. 2019, 2019, e3685264. [Google Scholar] [CrossRef] [PubMed]
  186. Gao, S.; Hu, M. Bioavailability Challenges Associated with Development of Anti-Cancer Phenolics. Mini Rev. Med. Chem. 2010, 10, 550–567. [Google Scholar] [CrossRef]
  187. Souza, J.E.D.; Casanova, L.M.; Costa, S.S. Bioavailability of Phenolic Compounds: A Major Challenge for Drug Development? Rev. Fitos 2015, 9, 55–67. [Google Scholar] [CrossRef]
Figure 1. Main classes of molecules found in Cerrado plants and their potential therapeutic targets in obesity.
Figure 1. Main classes of molecules found in Cerrado plants and their potential therapeutic targets in obesity.
Ijms 24 15588 g001
Figure 2. Summary of the main mechanisms involved in the pathophysiology of obesity and its metabolic complications, and the role of Cerrado plants in improving such mechanisms. The role of Cerrado plants in regulating several mechanisms is not yet know, as it is also not known which compounds are responsible for the therapeutic effects of the plants.
Figure 2. Summary of the main mechanisms involved in the pathophysiology of obesity and its metabolic complications, and the role of Cerrado plants in improving such mechanisms. The role of Cerrado plants in regulating several mechanisms is not yet know, as it is also not known which compounds are responsible for the therapeutic effects of the plants.
Ijms 24 15588 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Monteiro-Alfredo, T.; Macedo, M.L.R.; de Picoli Souza, K.; Matafome, P. New Therapeutic Strategies for Obesity and Its Metabolic Sequelae: Brazilian Cerrado as a Unique Biome. Int. J. Mol. Sci. 2023, 24, 15588. https://doi.org/10.3390/ijms242115588

AMA Style

Monteiro-Alfredo T, Macedo MLR, de Picoli Souza K, Matafome P. New Therapeutic Strategies for Obesity and Its Metabolic Sequelae: Brazilian Cerrado as a Unique Biome. International Journal of Molecular Sciences. 2023; 24(21):15588. https://doi.org/10.3390/ijms242115588

Chicago/Turabian Style

Monteiro-Alfredo, Tamaeh, Maria Lígia Rodrigues Macedo, Kely de Picoli Souza, and Paulo Matafome. 2023. "New Therapeutic Strategies for Obesity and Its Metabolic Sequelae: Brazilian Cerrado as a Unique Biome" International Journal of Molecular Sciences 24, no. 21: 15588. https://doi.org/10.3390/ijms242115588

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