Next Article in Journal
Study of the Effect of Laser Radiation Parameters on the Efficiency of Lithotripsy
Previous Article in Journal
Understanding the Research Challenges in Low-Resource Language and Linking Bilingual News Articles in Multilingual News Archive
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Biological Properties in Relation to the Health-Promoting Effects of Independent and Combined Garcinia mangostana Pericarp and Curcuma in Lean Wistar Albino Rats

by
Hanan A. Alfawaz
1,
Ranyah Shaker M. Labban
1,2,
Ramesa Shafi Bhat
3 and
Afaf El-Ansary
4,*
1
Department of Food Science & Nutrition, College of Food Science & Agriculture, King Saud University, Riyadh 11495, Saudi Arabia
2
Deputyship for Therapeutic Services, General Administration of Nutrition, Ministry of Health, Riyadh 11595, Saudi Arabia
3
Biochemistry Department, College of Science, King Saud University, Riyadh 11495, Saudi Arabia
4
Central Research Laboratory, Female Centre for Scientific and Medical Studies, King Saud University, Riyadh 11495, Saudi Arabia
*
Author to whom correspondence should be addressed.
Appl. Sci. 2023, 13(15), 8567; https://doi.org/10.3390/app13158567
Submission received: 30 May 2023 / Revised: 12 July 2023 / Accepted: 12 July 2023 / Published: 25 July 2023
(This article belongs to the Section Food Science and Technology)

Abstract

:
With the increase in verifications and proofs highlighting the association between diet and health, several food products, such as phenolic rich fruits and vegetables, have emerged as possessing potential health benefits. Plants rich with fiber and phenolic content have long been considered as a health-promoting dietary intervention. Therefore, the present work aimed to test the independent and combined potential benefits of mangosteen pericarp extract (MPE) and curcuma rhizome extract (CRE) consumption through an in vivo study on Wister albino rats. The data demonstrated that the three nutritional interventions had no effect on serotonin or glutamate, but dopamine was considerably increased in response to the combined effects of MPE and curcuma (p ˂ 0.025). The anti-inflammatory potency of independent MPE and curcuma, as well as their combined effects, recorded lower levels of IL12 in all groups compared to controls (p ˂ 0.05), and only a considerably lower IL-6 in combination treatment only (p ˂ 0.028). The three dietary interventions dramatically lowered leptin levels, with the combined treatment significantly lower than the healthy control group (p ˂ 0.001). The combined treatment significantly improved levels of malondialdehyde (MDA) and GSH as indicators of oxidative stress and antioxidant capability. Our data reported anti-dyslipidemic and anti-hyperglycemic effects of the three studied nutritional interventions, with the independent curcuma being the most effective anti-hyperglycemic compound (p ˂ 0.009). Collectively, the three used nutritional intervention strategies demonstrated promising health-promoting effects with no side effects.

1. Introduction

Numerous chronic disorders, such as obesity, cardiovascular disease, hypertension, stroke, type 2 diabetes, metabolic syndrome, several malignancies, and possibly some neurological diseases, are influenced by diet, which is frequently viewed as a lifestyle component. Plants are the primary source of natural medications utilized in traditional therapeutic modalities [1].
Nearly 72,000 blooming plants, or roughly 17% of the 422,000 total, have been found to have therapeutic benefits according to current research undertaken globally. Nearly 8000 plant species were methodically categorized by Carolus Linnaeus (1707–1778) in the 18th century, and this categorization not only aided naturalists’ research but also that of pharmaceutical chemists. The long-standing connection between people and plants gave rise to the field of ethnobotany, which conducts rigorous research that is now accepted by all of humanity [2].
Through trial and error over the course of time, man has progressively learned to use plants as sources of food and medicine, and he has developed the capacity to meet his requirements locally. As a result of scientific and technical advancements, these plants are being used more and more effectively. Medicinal plants play a significant role in the delivery of primary healthcare in many regions of the world, particularly in their use as crucial sources of pharmaceutical and therapeutic products. Approximately 80% of the world’s population relies on natural products for primary healthcare. For instance, curcu-ma may act as a preventative in people at risk of cardiovascular diseases (CVD) by enhancing serum lipid levels and may be used as a secure dietary supplement in conjunction with traditional medications [3].
One of the most significant and flexible facets of human health is diet. Since over- and under-nutrition have a significant impact on morbidity and mortality, nutritional interventions are necessary to reduce morbidity and mortality through dietary adjustments. [4]. With an increase in research directly relating food and health, phenolic-rich vegetables and fruits have emerged as possibly health-beneficial. Plants have long been regarded to be the source of health promotion because of their high fiber and phenolic content, as well as their important biological potential [4,5].
In South and Southeast Asia, particularly Malaysia, Garcinia mangostana (GM) (Guttiferae family) is commonly available. Mangosteen is its indigenous name. For its sweetness and juiciness, as well as its significance in increasing one’s health, GM is referred to as “the queen of fruits” in many circles. They are white, soft, and juicy with a sweet, slightly acidic taste and have a kernel inside [6,7]. Mangosteens are often recognized as one of the most delectable fruits due to its lovely aroma [7]. Southeast Asia has long utilized mangosteen’s pericarp and the entire fruit to treat a number of diseases, including fever, cholera, mycosis, skin infections, diarrhea, dysentery, and dysentery. There are several therapeutic uses for the fruits. [8]. The GM has possible biological properties without causing significant negative effects [8]. It increases the immune system and reduces metabolic syndromes (MS) and its associated diseases, respectively [9]. By lowering oxidative stress, reducing inflammation, managing and controlling obesity and diabetes, inhibiting the growth of cancer cells, and exhibiting anti-allergic, anti-bacterial, anti-fungal, and anti-malarial properties, the active ingredient in GMs, xanthone, was found to have health-promoting effects [10].
Curcuma is a herbaceous perennial plant that can grow to be 1 m tall. It produces a large number of tasty rhizomes with yellow or orange interiors. These rhizomes are ground into a powder to make a curcuma rhizome extract (CRE) spice. Proteins, carbohydrates, and resins make up the remaining elements. The most extensive studies have focused on curcuma’s anti-inflammatory, antioxidant, anti-carcinogenic, antiviral, and anti-infectious qualities. Additionally, there is a lot of interest in CRE’s ability to detoxify and heal wounds [11].
Both in vivo and in vitro assessments are used in the identification of the biological activities of plant extracts [12]. All in vivo methods involve the use of micro-organisms, and tests are applied on animals (mice, rats etc.) with proper doses and methods, followed by the assessment of blood or tissue samples. In vitro methods use sub-cellular systems, such as enzymes and receptors, that are isolated from animal or cell cultures [13]. The most commonly used one among these is the evaluation of the oxidant states of plants. Identification of the antioxidant activity in plant extracts is both an easy and cost-efficient method to be used in the elimination of the possible effects of the free radicals [13,14].
This study aims to investigate the independent and combined health promoting effects of the mangosteen pericarp extract (MPE) and CRE through the use of Wister Albino rats as subjects for the nutritional interventions.

2. Materials and Methods

2.1. Plant Materials

Fresh G. mangostana (mangosteen) fruits were purchased in Riyadh, Saudi Arabia, in neighbourhood hypermarkets in February while curcuma longa rhizomes were purchased in October 2019. Dr. Mohamed Yousef of the King Saud University College of Pharmacy validated the taxonomic identification of plant species. The herbarium of the same department now houses the voucher specimens (GA-6-2019) and (CL-7-2019). The plant sample was adequately cleaned, free of extraneous matter, and washed twice with double-distilled water. The pericarp of the mangosteen fruits was manually harvested and dried for 10 days at room temperature after they had been peeled. On the other hand, curcuma rhizomes were peeled and dried for 3–4 days at 50 °C in a drying oven with an air fan (Advantec FG-220 forced convection oven), a product of Carbolite Gero (Germany), a part of Verder Scientific group, London, UK. Following drying, the biomaterials were individually weighed, processed into a coarse powder using a commercial mill, and kept in the ultra-low temperature freezer (Product of New Brunswick—Eppendorf Company, Germany—Eppendorf Middle East and Africa FZ-LLC, Dubai, United Arab Emirates) at −80 °C until further use. All regional, national, and international regulations and laws were adhered to in the creation of this study.

2.2. Preparation of Extracts

Mangosteen pericarps and individual curcuma rhizomes underwent cold percolation. Each powdered plant material was steeped for 3 days at room temperature in 1.5 L of absolute methanol with continuous stirring. The suspended particles were centrifuged out of the mixture after the supernatant was filtered using the Whatman No. 1 filter paper. The residues were subjected to a second and third extraction under comparable conditions. Every day, the dissolved fraction was filtered and stored in a glass bottle. After the third extraction, the three organic filtrates were mixed and concentrated in vacuo using a rota-evaporator at 50 °C under reduced pressure in order to produce a crude white (21.23 g) and dark yellow (23.12 g) viscus residue for MP and CR, respectively. Both extracts were stored in glass vials in a refrigerator at a temperature of 80 °C prior to use.

2.3. Animals and Experimental Arrangements

The Ethical Committee of Scientific Research at KSU approved all experimental procedures conducted at the Experimental Surgery and Animal Laboratory Prince Naif Health Research Center (PNHRC). All animals were housed in polypropylene cages in a clean, controlled environment with a temperature of (25 °C ± 1 °C), a 12-h light–dark cycle, and a relative humidity of 50 ± 5%. Twenty male Rattus norvegicus Wistar albino rats weighing 100 ± 20 g were kept individually in polypropylene cages under controlled environmental conditions at the age of 4 weeks. Control and nutritionally treated groups were fed on standard laboratory animal feed pellets (ain-93 diet purchased from Riyadh Grain Silos) and filtered tap water using an MPE (400 mg/kg/day) (i.e., a stock solution of 400 mg of MPE was dissolved in 5 mL water, and then 0.5 mL was orally administered/100 g body weight rat) and CRE (80 mg/kg/day) (i.e., a stock solution of 80 mg CRE was dissolved in 5 mL water, and then 0.5 mL was orally administered/100 g body weight rat). Both extracts were given throughout the 6-week experiment. Body weight was assessed on a weekly basis until the completion of the experiment. Animals were anaesthetized with 5.0% sevoflurane and 100% oxygen at the end of the experiment. The flow rate of sevoflurane was calculated using the formula: weight (g) = 0.5 flow rate (mL/min). Each animal’s blood and brain tissue samples were collected. The biochemical assays were repeated 5 times per each group. All experiments were carried out in compliance with the National Institutes of Health Guidelines for the Care and Use of Laboratory Animals and were authorized by King Saud University’s Animal Ethics Committee (reference no: KSU-SE-18-17). Our research was conducted in accordance with the ARRIVE criteria.

2.4. Biochemical Assay

2.4.1. Preparation of Brain Tissue Homogenates

After collection, the brain tissue was rinsed using cold normal saline and homogenized in ten volumes/weight of phosphate-buffered saline (PBS). The homogenate was then centrifuged at 1500× g for 10 min, and the clear supernatant was collected and used for the measurements of neurotransmitters as described subsequently. We used the enzyme-linked Immunosorbent Assay (ELISA) instrument, a product of Tecan trading AG, 8708 Männedorf, Switzerland. Measurements were carried out in accordance with the manual’s directions. In general, the ELISA validation was carried out using the given limit of detection and the assessment of specificity (using quality control samples).

2.4.2. Measurement of Brain Neurotransmitters

A Competitive ELISA kit from MyBioSource was used to measure the levels of dopamine and glutamate. Using the MyBioSource Quantitative Sandwich ELISA kit, the level of serotonin was measured. The sensitivity was 1.0 ng/mL. for the 3 measured brain neurotransmitters.

2.4.3. Measurement of Serum Pro-Inflammatory Cytokines

The concentrations of IL-6 and IL-12 in the serum samples were determined using a quantitative sandwich enzyme ELISA kit from MyBioSource. The sensitivity was 1.0 pg/mL for both cytokines.

2.4.4. Leptin and Dyslipidemia-Related Markers

Using a quantitative sandwich ELISA kit from MyBio-Source, leptin levels in serum were measured. The sensitivity was 1.0 ng/mL. Using kits from the United Diagnostics Industry, quantitative estimations of cholesterol, high-density lipoprotein (HDL-C), and low-density lipoprotein (LDL-C) were assayed.

2.4.5. Measurement of Oxidative Stress Related Markers

By the usage of spectrophotometer, a product of Bioevopeak, Jinan, Shandong, China, lipid oxidation was assessed using the technique described by Ruiz-Larrea et al. [15]. Lipid peroxidation was expressed as nmol of malondialdehyde (MDA) per mL of serum, taking into account the molar extinction coefficient of MDA (ε = 156 mM−1·cm−1). The method of Jagota and Dani [16] was used to measure vitamin C. Using the 5,5′-dithiobis 2-nitrobenzoic acid and sulfhydryl compounds described by Beutler and Kelly [16], a relatively persistent yellow color was produced in order to estimate glutathione (GSH). By monitoring the reaction between the GST substrate, GSH, and 1-chloro-2,4-dinitrobenzene, GST activity was measured [17].

2.5. Statistical Analysis

Data are expressed as means ± standard deviation (SD). To statistically compare the results between groups, a one-way analysis of variance (ANOVA) with a Tukey post hoc test was performed. Significance was assigned at the level of p < 0.05. The IBM SPSS version 24 (IBM Corp., Armonk, NY, USA) was used.

3. Results

The results are presented as Mean ± S.D. of the different measured variables. The percentage increase or decrease was also presented within the same tables. Table 1 presents the concentrations of brain serotonin, dopamine, and glutamate as three important brain neurotransmitters. While serotonin and glutamate were not affected by the three nutritional interventions, dopamine was significantly elevated in response to the combined effects of MPE and CRE (p ˂ 0.025).
Table 2 demonstrates the anti-inflammatory effects of the independent and combined MPE and CRE. It can be easily noticed that there was significantly lower IL-12 in all groups (p ˂ 0.005) compared to controls, and only a significantly lower IL-6 in the MPE + CRE combined-treated group (p ˂ 0.028).
Table 3 demonstrates the concentrations of serum leptin in control, MPE, CRE and synergistically-treated rats. It was clear that the three dietary interventions induced remarkably lower levels of leptin with the combined treatment, in which it recorded a significantly lower level compared to healthy control group.
Levels of malondialdehyde (MDA) and GSH as markers of oxidative stress and antioxidant capacity were considerably improved with the combined treatment of MPE + CRE, in which it recorded a tendency to significance at p ˂ 0.071 and 0.06, respectively (Table 4).
Table 5 demonstrates the remarkable anti-dyslipidemic and anti-hyperglycemic effects of the three studied nutritional interventions, with the independent CRE being the most effective anti-hyperglycemic compound (p ˂ 0.009).

4. Discussion

Overall, the current study supported the independent and combined health-promoting effects of MPE and CRE as nutritional treatments that may be preventive against the development of neurological and other chronic disorders.
While serotonin and glutamate levels were unaffected by single or combined treatments with MPE and CRE, dopamine levels were significantly higher in the three treated groups compared to the control, with the combined effects having the greatest effect.
It is well accepted that besides eating a balanced diet, several potential dietary interventions may help increase dopamine levels, including prebiotics, probiotics, fish oil, and vitamin D. This, in turn, could help mend brain function and mental health. Based on this, we could suggest that independent or combined MPE and CRE can induce healthier brain functions. This is in good agreement with the recent work of Do and Cho [18], which reported that MPE and its bioactive xanthones are promising candidates for the treatment of Alzheimer’s disease (AD), Parkinson’s disease (PD), and depression. Moreover, it could find support in the previous work of Ramaholimihaso et al. [19], which proved that CRE has demonstrated—a potency in modulating neurotransmitter levels—inflammatory signaling and excitotoxicity in the brain. This may support the hypothesis that MPE and curcumin, taken alone or in combination, may prevent the onset of dementia with Lewy bodies (DLB), the second-most common kind of late-onset degenerative dementia that is frequently associated with dopamine deficiency [20].
Under normal physiological conditions, a typical inflammatory response is characterized by the temporally limited elevation of inflammatory activity that occurs when a threat is present and resolves once the threat has passed [20,21,22]. However, the presence of certain social, psychological, environmental, and biological factors has been linked to the prevention of the resolution of acute inflammation and, in turn, the promotion of a state of low-grade systemic chronic inflammation (SCI) characterized by having higher circulating levels of cytokines. Transitioning from a short-lived to a long-lived inflammatory response can decrease immunological tolerance, cause major alterations in all tissues and organs, as well as normal cellular physiology, and increase the risk of a variety of noncommunicable diseases in both young and old people. Clinical consequences of SCI-induced damage can be severe, including an increased risk of metabolic syndrome (MS), which includes the triad of hypertension, hyperglycemia, and dyslipidemia [23,24], type 2 diabetes [25], cardiovascular disease (CVD) [26,27], chronic kidney disease, various types of cancer, and depression [28].
Table 2 demonstrated the significant anti-inflammatory effects of the three used nutritional interventions. A significantly lower IL-12 was recorded in MPE and CRE—either in independently or combined treated groups—while IL-6 was significantly reduced in response to the combined treatment only. Isogarcinol, as an ingredient of GM, presented anti-inflammatory activity by suppressing the CD4 T-cells regulation in the murine model [29,30]. The combination of Garcinia with other prebiotics significantly reduced the lipid content from the serum and liver in a dose-dependent manner. It also improved the homeostasis model by assessing the insulin resistance index and pro-inflammatory cytokines (tumor necrosis factor α TNF, IL-6) [29]. The reported anti-inflammatory effects could support the recent recommendation of [30] that natural xanthones as major components of MPE are potential lead compounds to be further industrialized into pharmaceutical means for the treatment of inflammatory diseases. Their mechanisms of action usually comprise of the modulation of different pro-inflammatory cytokines, such as interleukin (IL)-1β, IL-6, IL-12, IL-8, and TNF-α, as well as anti-inflammatory cytokines, such as IL-10. MPE is also effective in reducing inflammation through numerous pathways, such as the inhibition of the conversion of omega-6 arachidonic acid to prostaglandin (PG) E2 by Cyclooxygenase (COX) and the down-regulation of the COX2 gene transcription [31,32]. This could help to suggest the use of MPE as an alternative or complementary strategy to block cyclo-oxygenase activity and prevent arachidonic acid from being converted to prostaglandin for the prevention of PGE2-driven tumorigenesis [33]. The significant reduction of IL-6 only in the MPE + CRE treated group could support the phenomenon that the inclusion of natural products in combination therapies could be more effective in treating diseases. This could find great support through, considering the work of Yousaf et al. [34], showing that natural product combinations improved endothelial function and, thus, was linked to multiple targeted effects or better active ingredient absorption in the body. The endothelium was shown to be protected against hyperlipidemia, hypertension, DM, platelet activation, oxidative stress, and hyperhomocysteinemia by natural product combinations in 17 preclinical studies [35]. Reactive oxygen species, Nrf2-HO-1, p38MAPK, P13K/Akt, and NF-B were among the molecules targeted. This could support the antioxidant and anti-inflammatory effects of MPE and CRE as health-promoting nutritional intervention.
Table 3 demonstrated the significant reductions in serum leptin in the three nutritional studied interventions. In the table, it was clear that MPE had much higher leptin-lowering effects compared to CRE, while the combination of both was the most potent. While leptin is an essential factor of metabolic alterations that happen as a function of weight loss and may initiate the restoration of body weight [35], it may be counterintuitive to consider that lowering serum leptin concentration would improve weight loss. However, according to the evidence-based model suggested by Zhao et al. [36], a decline of leptin levels may give rise to central and peripheral enhancements of leptin sensitivity and leptin action. Therefore, the adiposity- and leptin-lowering effects of MPE and curcumin as dietary interventions (Table 3) could help to suggest their health promoting effects through the increase of sensitivity to this hormone by modulating biological mechanisms involved in leptin resistance [37].
Table 4 demonstrated the anti-oxidant effects of the independent and combined MPE and CRE. It was clear that the three dietary interventions induced considerably lower lipid peroxides as marker of oxidative stress (p ˂ 0.193; 0.067, and 0.071), and together with enhanced antioxidant activity, presented as much higher glutathione levels in the MPE + CRE-combined treated group compared to the control (p ˂ 0.060). This can find multiple support through considering previously published work related to the health-promoting effects of MPE and CRE [38]. They found that drinking 245 mL of a mangosteen-contained beverage by healthy people boosted the plasma anti-oxidant capacity by 60 % after 1 h. A clinical study reported that oral administration of polar fraction extract from MP to human subjects for 24 weeks enhanced the anti-oxidant activity without producing potential side effects [39], which again supports the MP health-promoting effects.
Table 5 demonstrated the lipid—metabolism-related health-promoting effects of the independent or combined MPE and CRE. While independently or combined, the three nutritional interventions did not demonstrate any change in CHOL/HDL-C as a marker of CVD development; the combined effects of the MPE + CRE demonstrated a much higher HDL-C/LDL-C as marker of low risk/protection against CVD occurrence. This was interesting, as presently existing drugs used for the treatment of dyslipidemia usually are only effective in reducing total- and LDL-C and triglycerides levels, with little or no effect observed in HDL-C levels [40]. The promising results we have, although failing to show a significant difference compared to control, suggest a possible role of the combined MPE + CRE in selectively increasing HDL-C, with their known anti-atherogenic effects. This was in good agreement with the previous works of Qin et al. [1] and Labban et al. [41], in which there were anti-dyslipidemia effects of CRE, MPE, and the combination of both. It was hopeful to report that CRE could be protective in individuals at risk of CVD through improving serum lipid levels and may be used as a safe dietary supplement with conventional drugs [1].
Table 5 also demonstrated the regulatory effects of the three nutritional interventions on serum glucose as markers related the development of DM. MPE and CRE either independently or in combination showed a satisfactory glucose-lowering effect. This could be attributed to the insulin sensitizing effect of mangosteen extract previously reported by [2], in which CRE could be protective in individuals at risk of CVD through improving serum lipid levels and may be used as a safe dietary supplement with conventional drugs [1].
Table 5 also demonstrated the regulatory effects of the three nutritional interventions on serum glucose as markers related the development of DM. MPE and CRE, either independently or in combination, showed a satisfactory glucose lowering effect. This could be attributed to the insulin sensitizing effect of mangosteen extract previously reported by Watanabe et al. [42,43].

5. Conclusions

Recently, growing concerns about the negative effects of nutrition and diets on obesity, the development of chronic diseases such as neurological disorders, CVD, DM, cancer, and other diseases has made nutritional intervention using polyphenolic compounds (e.g., xanthone, lignans, phenolic acids, etc.)—rich natural products—highly recommended and promising. MPE and CRE, either alone or in combination, have potential biological qualities without causing obvious negative effects. The three nutritional interventional strategies studied were found to have health-beneficial effects by lowering oxidative stress, reducing inflammation, protecting against dyslipidemia, and increasing insulin sensitivity as a preventative strategy against the development of diabetes. However, more research on the health effects of MPE and CRE on human subjects is required to clarify concerns regarding dosage form, safety, dose, and treatment frequency. ection is not mandatory, but can be added to the manuscript if the discussion is unusually long or complex.

Author Contributions

H.A.A.: co-drafted the manuscript; R.S.M.L.: acquired the data; R.S.B.: acquired the data; and A.E.-A.: suggested the topic and drafted the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded through the deputyship for Research and Innovation, “Ministry of Education” in Saudi Arabia (IFKSUOR3-123-2).

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Institutional Review Board of the College of Medicine at King Saud University.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The datasets of the current study are available from the corresponding author on reasonable request.

Acknowledgments

The authors extend their appreciation to the Deputyship for Research and Innovation, “Ministry of Education”, in Saudi Arabia for funding this research (IFKSUOR3-123-2).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Gropper, S.S. The Role of Nutrition in Chronic Disease. Nutrients 2023, 15, 664. [Google Scholar] [CrossRef]
  2. Capasso, R.; Izzo, A.A.; Pinto, L.; Bifulco, T.; Vitobello, C.; Mascolo, N. Phytotherapy and quality of herbal medicines. Fitoterapia 2000, 71 (Suppl. 1), S58–S65. [Google Scholar] [CrossRef]
  3. Qin, S.; Huang, L.; Gong, J.; Shen, S.; Huang, J.; Ren, H.; Hu, H. Efficacy and Safety of Turmeric and Curcumin in Lowering Blood Lipid Levels in Patients with Cardiovascular Risk Factors: A Meta-Analysis of Randomized Controlled Trials. Nutr. J. 2017, 16, 1–10. [Google Scholar]
  4. Ali, M.S.; Kassahun, C.W.; Wubneh, C.A. Overnutrition and Associated Factors: A Comparative Cross-Sectional Study between Government and Private Primary School Students in Gondar Town, Northwest Ethiopia. J. Nutr. Metab. 2020, 2020, 3670895. [Google Scholar] [CrossRef]
  5. Alebel, A.; Demant, D.; Petrucka, P.; Sibbritt, D. Effects of undernutrition on mortality and morbidity among adults living with HIV in sub-Saharan Africa: A systematic review and meta-analysis. BMC Infect. Dis. 2021, 21, 1–20. [Google Scholar] [CrossRef]
  6. Fernandes, A.; Mateus, N.; de Freitas, V. Polyphenol-Dietary Fiber Conjugates from Fruits and Vegetables: Nature and Biological Fate in a Food and Nutrition Perspective. Foods 2023, 12, 1052. [Google Scholar] [CrossRef]
  7. Rana, A.; Samtiya, M.; Dhewa, T.; Mishra, V.; Aluko, R.E. Health benefits of polyphenols: A concise review. J. Food Biochem. 2022, 46, e14264. [Google Scholar] [CrossRef]
  8. Obolskiy, D.; Pischel, I.; Siriwatanametanon, N.; Heinrich, M. Garcinia mangostana L.: A Phytochemical and Pharmacological Review. Phytother. Res. PTR 2009, 23, 1047–1065. [Google Scholar] [CrossRef]
  9. Ovalle-Magallanes, B.; Eugenio, D.; Pedraza-Chaverri, J. Medicinal properties of mangosteen (Garcinia mangostana L.): A comprehensive update. Food Chem. Toxicol. 2017, 109, 102–122. [Google Scholar] [CrossRef]
  10. Pedraza-Chaverri, J.; Cardenas, N.; Orozco-Ibarra, M.; Pérez-Rojas, J. Medicinal properties of mangosteen (Garcinia mangostana). Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2008, 46, 3227–3239. [Google Scholar] [CrossRef]
  11. Zhang, S.; Li, Z.; Wang, X.; An, L.; Bao, J.; Zhang, J.; Cui, J.; Li, Y.; Jin, D.-Q.; Tuerhong, M.; et al. Isolation, structural elucidation, and immunoregulation properties of an arabinofuranan from the rinds of Garcinia mangostana. Carbohydr. Polym. 2020, 246, 116567. [Google Scholar] [CrossRef]
  12. Yikna, B.B.; Yehualashet, A.S. Medicinal Plant Extracts Evaluated In Vitro and In Vivo for Antidiabetic Activities in Ethiopia: Bases for Future Clinical Trials and Related Investigations. Evid. Based Complement. Altern. Med. 2021, 2021, 9108499. [Google Scholar] [CrossRef]
  13. Zjawiony, J.K. Biologically active compounds from aphyllophorales (polypore) fungi. J. Nat. Prod. 2004, 67, 300–310. [Google Scholar] [CrossRef]
  14. Chikezie, P.C.; Ibegbulem, C.O.; Mbagwu, F.N. Bioactive principles from medicinal plants. Res. J. Phytochem. 2015, 9, 88–115. [Google Scholar] [CrossRef] [Green Version]
  15. Ruiz-Larrea, B.M.; Leal, A.M.; Liza, M.; Lacort, M.; de Groott, H. Antioxidant Effects of Estradiol and 2-Hydroxyestradiol on Iron-Induced Lipid Peroxidation of Rat Liver Microsomes. Steroids 1994, 59, 383–388. [Google Scholar]
  16. Jagota, S.K.; Dani, H.M. A New Calorimetric Technique for the Estimation of Vitamin C Using Folin Phenol Reagent. Anal. Biochem. 1982, 127, 178–182. [Google Scholar] [CrossRef]
  17. Beutler, E.; Kelly, B.M. The Effect of Sodium Nitrite on Red Cell GSH. Experientia 1963, 19, 96–97. [Google Scholar] [CrossRef]
  18. Do, H.T.T.; Cho, J. Mangosteen Pericarp and Its Bioactive Xanthones: Potential Therapeutic Value in Alzheimer’s Disease, Parkinson’s Disease, and Depression with Pharmacokinetic and Safety Profiles. Int. J. Mol. Sci. 2020, 21, 6211. [Google Scholar]
  19. Ramaholimihaso, T.; Bouazzaoui, F.; Kaladjian, A. Curcumin in Depression: Potential Mechanisms of Action and Current Evidence—A Narrative Review. Front. Psychiatry 2020, 11, 572533. [Google Scholar]
  20. Huber, M.; Beyer, L.; Prix, C.; Schönecker, S.; Palleis, C.; Rauchmann, B.S.; Morbelli, S.; Chincarini, A.; Bruffaerts, R.; Vandenberghe, R.; et al. Metabolic Correlates of Dopaminergic Loss in Dementia with Lewy Bodies. Mov. Disord. 2020, 35, 595–605. [Google Scholar] [CrossRef]
  21. Kotas, M.E.; Medzhitov, R. Homeostasis, Inflammation, and Disease Susceptibility. Cell 2015, 160, 816–827. [Google Scholar] [CrossRef] [Green Version]
  22. Fullerton, J.N.; Gilroy, D.W. Resolution of Inflammation: A New Therapeutic Frontier. Nat. Rev. Drug Discov. 2016, 15, 551–567. [Google Scholar] [CrossRef]
  23. Straub, R.H. The Brain and Immune System Prompt Energy Shortage in Chronic Inflammation and Ageing. Nat. Rev. Rheumatol. 2017, 13, 743–751. [Google Scholar] [CrossRef]
  24. Jin, C.; Henao-Mejia, J.; Flavell, R.A. Innate Immune Receptors: Key Regulators of Metabolic Disease Progression. Cell Metab. 2013, 17, 873–882. [Google Scholar] [CrossRef] [Green Version]
  25. Hotamisligil, G.S. Inflammation and Metabolic Disorders. Nature 2006, 444, 860–867. [Google Scholar] [CrossRef]
  26. Dregan, A.; Charlton, J.; Chowienczyk, P.J.; Gulliford, M.C. Chronic Inflammatory Disorders and Risk of Type 2 Diabetes Mellitus, Coronary Heart Disease, and Stroke: A Population-Based Cohort Study. Circulation 2014, 130, 837–844. [Google Scholar] [CrossRef] [Green Version]
  27. Gisterå, A.; Hansson, G.K. The Immunology of Atherosclerosis. Nat. Rev. Nephrol. 2017, 13, 368–380. [Google Scholar] [CrossRef]
  28. Ferrucci, L.; Fabbri, E. Inflammageing: Chronic Inflammation in Ageing, Cardiovascular Disease, and Frailty. Nat. Rev. Cardiol. 2018, 15, 505–522. [Google Scholar] [CrossRef]
  29. Furman, D.; Campisi, J.; Verdin, E.; Carrera-Bastos, P.; Targ, S.; Franceschi, C.; Ferrucci, L.; Gilroy, D.W.; Fasano, A.; Miller, G.W.; et al. Chronic Inflammation in the Etiology of Disease across the Life Span. Nat. Med. 2019, 25, 1822–1832. [Google Scholar] [CrossRef]
  30. Chang, C.W.; Hsu, Y.J.; Chen, Y.M.; Huang, W.C.; Huang, C.C.; Hsu, M.C. Effects of Combined Extract of Cocoa, Coffee, Green Tea and Garcinia on Lipid Profiles, Glycaemic Markers and Inflammatory Responses in Hamsters. BMC Complement. Altern. Med. 2015, 15, 1–11. [Google Scholar] [CrossRef] [Green Version]
  31. Gunter, N.V.; Teh, S.S.; Lim, Y.M.; Mah, S.H. Natural Xanthones and Skin Inflammatory Diseases: Multitargeting Mechanisms of Action and Potential Application. Front. Pharmacol. 2020, 11, 594202. [Google Scholar] [CrossRef]
  32. Nakatani, K.; Yamakuni, T.; Kondo, N.; Arakawa, T.; Oosawa, K.; Shimura, S.; Inoue, H.; Ohizumi, Y. γ-Mangostin Inhibits Inhibitor-ΚB Kinase Activity and Decreases Lipopolysaccharide-Lnduced Cyclooxygenase-2 Gene Expression in C6 Rat Glioma Cells. Mol. Pharmacol. 2004, 66, 667–674. [Google Scholar] [CrossRef] [Green Version]
  33. Yamakuni, T.; Aoki, K.; Nakatani, K.; Kondo, N.; Oku, H.; Ishiguro, K.; Ohizumi, Y. Garcinone B Reduces Prostaglandin E2 Release and NF-ΚB-Mediated Transcription in C6 Rat Glioma Cells. Neurosci. Lett. 2006, 394, 206–210. [Google Scholar] [CrossRef]
  34. Collard, T.J.; Fallatah, H.M.; Greenhough, A.; Paraskeva, C.; Williams, A.C. BCL–3 Promotes Cyclooxygenase–2/Prostaglandin E2 Signalling in Colorectal Cancer. Int. J. Oncol. 2020, 56, 1304–1313. [Google Scholar] [CrossRef]
  35. Yusuf, M.S.; Hassan, M.A.; Tag, H.M.; Sarivistava, K.; Reddy, P.G.; Hassan, A.M. Influence of Turmeric (Curcuma longa) On performance, histomorphology and microbiota of intestine in juvenile Tilapia (Oreochromis niloticus). Int. J. Agric. Sci. Vet. Med. 2017, 5, 7–16. [Google Scholar]
  36. Sumithran, P.; Prendergast, L.A.; Delbridge, E.; Purcell, K.; Shulkes, A.; Kriketos, A.; Proietto, J. Long-Term Persistence of Hormonal Adaptations to Weight Loss. N. Engl. J. Med. 2011, 365, 1597–1604. [Google Scholar] [CrossRef] [Green Version]
  37. Zhao, S.; Kusminski, C.M.; Elmquist, J.K.; Scherer, P.E. Leptin: Less Is More. Diabetes 2020, 69, 823–829. [Google Scholar] [CrossRef]
  38. Andreoli, M.F.; Donato, J.; Cakir, I.; Perello, M. Leptin Resensitisation: A Reversion of Leptin-Resistant States. J. Endocrinol. 2019, 241, R81–R96. [Google Scholar] [CrossRef] [Green Version]
  39. Xie, Z.; Sintara, M.; Chang, T.; Ou, B. Functional Beverage of Garcinia mangostana (Mangosteen) Enhances Plasma Antioxidant Capacity in Healthy Adults. Food Sci. Nutr. 2015, 3, 32–38. [Google Scholar] [CrossRef]
  40. Suthammarak, W.; Numpraphrut, P.; Charoensakdi, R.; Neungton, N.; Tunrungruangtavee, V.; Jaisupa, N.; Charoensak, S.; Moongkarndi, P.; Muangpaisan, W. Antioxidant-Enhancing Property of the Polar Fraction of Mangosteen Pericarp Extract and Evaluation of Its Safety in Humans. Oxidative Med. Cell. Longev. 2016, 2016, 1293036. [Google Scholar] [CrossRef] [Green Version]
  41. Chang, Y.; Robidoux, J. Dyslipidemia Management Update. Curr. Opin. Pharmacol. 2017, 33, 47–55. [Google Scholar] [CrossRef]
  42. Labban, R.S.M.; Alfawaz, H.A.; Almnaizel, A.T.; Al-Muammar, M.N.; Bhat, R.S.; El-Ansary, A. Garcinia mangostana Extract and Curcumin Ameliorate Oxidative Stress, Dyslipidemia, and Hyperglycemia in High Fat Diet-Induced Obese Wistar Albino Rats. Sci. Rep. 2021, 11, 7278. [Google Scholar] [CrossRef]
  43. Watanabe, M.; Gangitano, E.; Francomano, D.; Addessi, E.; Toscano, R.; Costantini, D.; Tuccinardi, D.; Mariani, S.; Basciani, S.; Spera, G.; et al. Mangosteen Extract Shows a Potent Insulin Sensitizing Effect in Obese Female Patients: A Prospective Randomized Controlled Pilot Study. Nutrients 2018, 10, 586. [Google Scholar] [CrossRef] [Green Version]
Table 1. Levels of brain serotonin, dopamine, and glutamate neurotransmitters in the control, independent MPE- or CRE-treated groups, and the combined-treated group.
Table 1. Levels of brain serotonin, dopamine, and glutamate neurotransmitters in the control, independent MPE- or CRE-treated groups, and the combined-treated group.
ParametersGroupsMean ± S.D.Percent Changep Value
Serotonin (ng/mL)Control77.40 ± 14.50100.00
MPE64.20 ± 2.17−17.050.111
CRE80.60 ± 13.32+4.130.726
MPE + CRE62.40 ± 16.04−19.380.159
Dopamin (ng/mL)Control34.60 ± 11.37100.00
MPE40.40 ± 5.46+16.760.345
CRE43.40 ± 11.55+25.430.259
MPE + CRE57.20 ± 14.36+65.320.025
Glutamate (ng/mL)Control36.20 ± 8.79100.00
MPE38.60 ± 3.36+6.630.584
CRE36.20 ± 6.72100.001.000
MPE + CRE42.60 ± 8.62+17.680.278
Table 2. Levels of serum IL-12 and IL-6 in the control, independent MPE or CRE, and the combined-treated rats. is a table. Tables should be placed in the main text near to the first time they are cited.
Table 2. Levels of serum IL-12 and IL-6 in the control, independent MPE or CRE, and the combined-treated rats. is a table. Tables should be placed in the main text near to the first time they are cited.
ParametersGroupsMean ± S.D.Percent Changep Value
(IL12) pg/mLControl44.14 ± 6.68100.00
MPE34.50 ± 5.20−21.830.034
CRE35.14 ± 3.46−20.390.028
MPE + CRE34.86 ± 3.75−21.030.027
IL6 (pg/mL)Control84.50 ± 6.22100.00
MPE81.17 ± 4.55−3.940.362
CRE85.59 ± 4.30+1.300.754
MPE + CRE76.02 ± 3.40−10.130.028
Table 3. Levels of serum leptin in the control, independent MPE or CRE, and combined-treated rats.
Table 3. Levels of serum leptin in the control, independent MPE or CRE, and combined-treated rats.
ParametersGroupsMean ± S.D.Percent Changep Value
Leptin (ng/mL)Control0.89 ± 0.14100.00
MPE0.62 ± 0.09−30.040.007
CRE0.74 ± 0.14−17.340.123
MPE + CRE0.56 ± 0.05−37.220.001
Table 4. Levels of serum MDA, GSH, GST and vit. C in the control, independent MPE or CRE, and combined-treated rats.
Table 4. Levels of serum MDA, GSH, GST and vit. C in the control, independent MPE or CRE, and combined-treated rats.
ParametersGroupsMean ± S.D.Percent Changep Value
MDA
µm/ml
Control108.95 ± 12.54100.00
MPE100.18 ± 0.98−8.050.193
CRE94.77 ± 8.12−13.010.067
MPE + CRE93.17 ± 11.36−14.480.071
GSH
µg/ml
Control148.31 ± 36.42100.00
MPE123.86 ± 15.53−16.480.205
CRE125.11 ± 16.23−15.640.230
MPE + CRE162.51 ± 47.20+9.570.060
Table 5. Levels of serum CHOL/HDL-C, HDL-C/LDL-C, and GLU in the control, independent MPE or CRE, and combined-treated rats.
Table 5. Levels of serum CHOL/HDL-C, HDL-C/LDL-C, and GLU in the control, independent MPE or CRE, and combined-treated rats.
ParametersGroupsMean ± S.D.Percent Changep Value
CHOL/HDL-C
mg/dL
Control1.52 ± 0.28100.00
MPE1.49 ± 0.24−1.710.877
CRE1.73 ± 0.40+14.230.352
MPE + CRE1.36 ± 0.32−10.670.416
HDL-C/LDL-CControl4.61 ± 1.21100.00
MPE2.54 ± 1.02−44.900.016
CRE1.95 ± 0.40−57.710.009
MPE + CRE5.35 ± 1.43+16.070.465
GLU (mg/dL)Control232.55 ± 74.67100.00
MPE154.43 ± 34.27−33.590.076
CRE121.12 ± 8.90−47.910.009
MPE + CRE162.08 ± 39.89−30.300.117
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

Alfawaz, H.A.; Labban, R.S.M.; Bhat, R.S.; El-Ansary, A. Biological Properties in Relation to the Health-Promoting Effects of Independent and Combined Garcinia mangostana Pericarp and Curcuma in Lean Wistar Albino Rats. Appl. Sci. 2023, 13, 8567. https://doi.org/10.3390/app13158567

AMA Style

Alfawaz HA, Labban RSM, Bhat RS, El-Ansary A. Biological Properties in Relation to the Health-Promoting Effects of Independent and Combined Garcinia mangostana Pericarp and Curcuma in Lean Wistar Albino Rats. Applied Sciences. 2023; 13(15):8567. https://doi.org/10.3390/app13158567

Chicago/Turabian Style

Alfawaz, Hanan A., Ranyah Shaker M. Labban, Ramesa Shafi Bhat, and Afaf El-Ansary. 2023. "Biological Properties in Relation to the Health-Promoting Effects of Independent and Combined Garcinia mangostana Pericarp and Curcuma in Lean Wistar Albino Rats" Applied Sciences 13, no. 15: 8567. https://doi.org/10.3390/app13158567

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