Previous Article in Journal
The Effect of Oral GABA on the Nervous System: Potential for Therapeutic Intervention
Previous Article in Special Issue
Nutraceutical Aspects of Selected Wild Edible Plants of the Italian Central Apennines
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Melatonin Modulates Lipid Metabolism and Reduces Cardiovascular Risk in Apolipoprotein E-Deficient Mice Fed a Western Diet

by
Guillermo Santos-Sánchez
1,2,†,
Ana Isabel Álvarez-López
1,2,†,
Eduardo Ponce-España
1,2,
Ana Isabel Álvarez-Ríos
3,
Patricia Judith Lardone
1,2,
Antonio Carrillo-Vico
1,2,* and
Ivan Cruz-Chamorro
1,2,*
1
Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Seville, Spain
2
Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, 41009 Seville, Spain
3
Departamento de Bioquímica Clínica, Unidad de Gestión de Laboratorios, Hospital Universitario Virgen del Rocío, 41013 Seville, Spain
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutraceuticals 2024, 4(2), 260-272; https://doi.org/10.3390/nutraceuticals4020016
Submission received: 30 October 2023 / Revised: 25 April 2024 / Accepted: 6 May 2024 / Published: 9 May 2024
(This article belongs to the Special Issue Functional Foods as a New Therapeutic Strategy 2.0)

Abstract

:
Melatonin (MLT), a natural compound found in the animal and vegetable kingdom, participates in several physiological processes. MLT exerts antioxidant and anti-inflammatory activities, among others, but information about its action on lipid metabolism is still scarce. For this reason, mice deficient in apolipoprotein E (ApoE−/−) fed a Western diet (WD) were intragastrically treated with different concentrations of MLT (2 and 9 mg/kg) for 12 weeks. The lipid parameters were quantified, and, since links between cardiovascular risk and immune function and oxidative stress have been established, we also analyzed the population of leukocytes and the oxidative stress status. Although there was no change in the weight of the mice, a significant reduction in low-density lipoprotein cholesterol (LDL-C) was observed in mice treated with the higher concentration of MLT tested in this study. Additionally, an improvement in cardiovascular risk indexes was observed. A reduction in the hepatic total cholesterol (TC) and LDL-C levels was also observed in the treated mice. Finally, a decrease in leukocytes and lymphocytes in particular, as well as an increase in the antioxidant status, were shown in MLT-treated mice. In conclusion, MLT is a promising candidate that could be considered as a possible functional ingredient capable of preventing cardiovascular risk.

Graphical Abstract

1. Introduction

Cardiovascular diseases (CVDs), including coronary, rheumatic and congenital heart, cerebrovascular, and peripheral arterial diseases, are the leading cause of mortality worldwide (being responsible for 32% of all global deaths in 2019) and a major contributor to disability [1]. Furthermore, CVDs remain a major cause of rising health care costs, and their prevalence nearly doubled from 271 million in 1990 to 523 million in 2019 [2]. The relationship between cholesterol and CVDs has been reported by several epidemiological studies [3,4,5]. Low-density lipoprotein cholesterol (LDL-C) levels, also called ‘bad cholesterol’ as it is the main cholesterol-carrying particle in plasma, are directly associated with the subsequent risk of CVDs, due to LDL-C’s ability to be deposited in the intima vessels and thus cause obstruction [6]. In contrast, increasing high-density lipoprotein cholesterol (HDL-C) levels has been accepted as a therapeutic strategy to reduce the risk of death from CVDs because HDL-C transports cholesterol from tissues to the liver, reducing the serum values [7]. However, studies have shown that when conventional lipid parameters remain apparently normal or moderately high, lipid relationships such as the Castelli risk index (CRI) I (CRI-I, considered as the ratio between total cholesterol -TC- and HDL-C) and II (CRI-II, considered as the LDL-C/HDL-C ratio), and the atherogenic index of plasma (AIP, considered as the logarithm of the ratio between triglycerides -TGs- and HDL-C) are diagnostic alternatives for the prediction of cardiovascular events [8,9,10] and the effectiveness of therapy [11]. Specifically, the CRI-I has been associated with the formation of coronary plaques [12,13], and the CRI-II has been shown to be a predictor of cardiovascular risk [14]. Regarding the AIP, previous studies have correlated high values of this index to the risk of cardiovascular incidence and all-cause mortality in patients with coronary heart disease [15,16,17].
Alternatively, an elevated count of white blood cells serves as a robust and autonomous indicator of the risk of heart-related issues in individuals of all genders, whether they have coronary heart disease or not. Furthermore, an increased quantity of leukocytes is linked to the occurrence of coronary heart disease, peripheral arterial conditions, and strokes [18]. Furthermore, alterations in lipid metabolism are closely related to alterations in the immune system and prevailing chronic systemic inflammation in people with a pathological increase in body fat [19]. Furthermore, a rise in lymphocyte count has been observed in conditions like obesity, diabetes, and cardiovascular disease. Lymphocytes are notably elevated in visceral fat, playing a significant role as a key controller of insulin resistance. [20]. For this reason, the relationship between nutritional status and immune functions has been widely studied over the years, associating malnutrition with pathological conditions [21,22]. In such studies, overweight mice fed a Western diet (WD) have shown an increase in the total number of leukocytes and lymphocyte cell numbers [23,24].
Furthermore, oxidative stress has been widely highlighted as a major risk factor in the development of the main cardiovascular diseases. This is an imbalance between the production of reactive oxygen species (ROSs) and antioxidant defenses caused by certain cardiovascular risk factors, such as diabetes, hypertension, smoking, and obesity [25]. At the cardiovascular level, oxidative stress is highly implicated in myocardial infarction, ischemia/reperfusion, and heart failure due to damage to the vascular system through lipid peroxidation, membrane damage, immune cell activation (proteases, nucleases, and protein kinases), structural remodeling, or inflammation [26].
Melatonin (MLT) is considered a pleiotropic molecule due to its participation in various physiological processes. This particular indoleamine is primarily produced by the pineal gland in accordance with a daily cycle, reaching its highest levels at nighttime and the lowest levels during the daytime in humans [27]. In recent decades, this molecule has been shown to not be exclusively synthesized in the pineal gland but also in several peripheral tissues, such as the gastrointestinal tract, immune system cells, and skin cells [28,29]. Unlike studies on its antioxidant role, studies on the effect of MLT on the regulation of lipid metabolism are scarce. Previous studies have shown that MLT can reduce serum levels of TC, LDL-C, HDL-C, and TGs. However, the conclusions obtained are variable and depend on the methodology used [30,31,32,33]; thus, more studies are required to determine the benefits of MLT for the lipid profile [34].
Furthermore, the immunomodulatory role of MLT has been extensively studied in recent years, and its efficacy in controlling inflammatory processes has been demonstrated in different diseases [29,35,36,37]. Previous studies have reported that MLT therapy decreases the number of leukocytes in burn-induced Wistar rats [38] and controls the count of leukocytes and lymphocytes during intense effort in adolescent athletes [39]. However, there is no evidence of the anti-inflammatory role of MLT in an animal model of cardiovascular risk.
Given that growing evidence has shown that MLT might exert a cardiovascular protective effect through the control of lipid metabolism and anti-inflammatory and antioxidant status, the aim of this study was to evaluate the effect of MLT on (i) the plasmatic and hepatic lipid profile, (ii) white blood cell population, and (iii) anti-inflammatory and oxidative stress status in an experimental model of cardiovascular disease consisting of apolipoprotein E (ApoE) knockout mice (ApoE−/−) fed a WD.

2. Materials and Methods

2.1. Study Design

Male ApoE−/− mice (kindly donated by Dr. Antonio Ordoñez and Dr. Raquel del Toro), at the age of four weeks, were kept in the animal facility at the Instituto de Biomedicina de Sevilla (IBiS) under typical conditions, which included a 12 h light and 12 h dark cycle, a temperature of 22 ± 2 °C, and humidity levels below 55%. These mice were provided with unrestricted access to both water and a Western diet (Test Diet 58v8, containing 45% energy from fat, as detailed in Supplementary Table S1). When the mice turned 6 weeks old, they were randomly divided into three groups and treated intragastrically with MLT daily (Sigma Aldrich, MO, USA) 2 mg/kg (WD + MLT (2 mg/kg), n = 12), 9 mg/kg (WD + MLT (9 mg/kg), n = 12) or the vehicle (=ethanol; WD, n = 10) for 12 weeks (Figure 1). The mice were closely monitored, controlled, and observed by the researchers themselves and by the technical staff of the IBiS Animal Facility. In addition, the veterinary manager of the animal facility checked the health status of the animals every week, not recording any side effects.
The MLT doses used in this study were chosen because they are equivalent doses of 10 mg and 50 mg MLT/day, respectively, in humans, calculated according to [40] (Figure 2). Daily food intake and individual body weight were measured weekly and recorded. At the endpoint, fasted animals were euthanized, and blood was collected in Minicollect EDTA tubes (Greiner Bio-one, Kremsmünster, Austria) by cardiac puncture. Subsequently, plasma was obtained by centrifugation (3000× g, 4 °C, 10 min) and stored at −20 °C until use. The animals were then perfused with phosphate-buffered saline (PBS) for 5 min using an FH100 peristaltic pump (Thermo Scientific, Vantaa, Finland), and the liver was collected and stored until use.
Every experimental process adhered to the regulations established by Spanish law and conformed to the guidelines outlined in the EU Directive 2010/63/EU regarding animal experiments. Additionally, these procedures received approval from the Ethics Committee at the Virgen Macarena and Virgen del Rocío University Hospitals, with reference number 21/06/2016/105.

2.2. Plasma and Hepatic–Lipid Profile

Plasma lipid parameters (including TC, TGs, LDL-C, and HDL-C) were measured with chemiluminescence immunoassay techniques using the COBAS e601 modular analyzer (Roche Diagnostic, Basel, Switzerland). In addition, the cardiovascular disease risk indexes CRI-I, CRI-II, and AIP were calculated according to [41]. On the other hand, 100 mg liver tissues were homogenized with a TissueRuptor (Qiagen, Hilden, Germany), and the hepatic TC, HDL-C, LDL-C, and TG concentrations were measured in the supernatants produced by Cobas Integra 400 (Roche Diagnostics, Indianapolis, IN, USA) at the ‘Estación Biológica de Doñana’ (EBD-CSIC, Seville, Spain).

2.3. White Blood Cell (WBC) Count

White blood cells were quantified in blood samples using the SYSMEX XE 5000 Hematology Analyzer (Sysmex Europe GmbH, Norderstedt, Germany) fluorescence flow cytometer.

2.4. Plasma ELISA

To confirm that MLT could act as an anti-inflammatory molecule, TNF was quantified by a commercial enzyme-linked immunosorbent assay (BD OptEIA™ Mouse TNF (Mono/Mono) ELISA Set, BD Biosciences, San Jose, CA, USA).
Briefly, the plasma of mice was incubated overnight with anti-mouse TNF antibody in a precoated 96-well plate. A biotinylated anti-mouse TNF antibody and streptavidin–HRP conjugate enzyme were used to detect the TNF cytokine. The addition of tetramethylbenzidine (TMB; Sigma-Aldrich, Saint Louis, MO, USA) led to the development of a color that was read at 450 nm with a CLARIOstar Plus microplate reader (BMG Labtech, Ortenberg, Germany) once the reaction was stopped by HCl.

2.5. Plasma Antioxidant Capacity

To test the role of MLT in the antioxidant capacity, the Trolox equivalent antioxidant capacity (TEAC) assay was performed. Briefly, 140 µL of 2,2-azino-bis-(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS, Sigma-Aldrich) radical solution was mixed with 10 µL of plasma. After 30 min of incubation at 30 °C, the ABTS radical content was quantified using a CLARIOstar Plus microplate reader (BMG Labtech) at 730 nm. Then, the values were extrapolated by a Trolox (Sigma-Aldrich) standard curve.

2.6. Statistical Analysis

The data are presented as the mean value accompanied by the standard error of the mean (SEM). Statistical analysis involved the use of non-parametric Mann–Whitney U tests or two-way ANOVA, followed by post hoc corrections, and statistical significance was established at p-values equal to or less than 0.05. The data underwent analysis using GraphPad Prism v.8 (GraphPad Software, San Diego, CA, USA).

3. Results

3.1. Melatonin Does Not Alter the Body Weight of Mice

As shown in Figure 3A, no significant differences in body weight (p = 0.815) were observed between the three experimental groups throughout the experiment. No differences were shown in basal or final body weight (Figure 3B,C) between the control group (basal: 21.29 ± 0.48 g; final: 26.32 ± 0.45 g) and the groups given 2 mg/kg MLT (basal: 20.38 ± 0.84 g, p = 0.594; final: 26.16 ± 0.82 g, p = 0.987) or 9 mg/kg MLT (basal: 20.37 ± 0.82 g, p = 0.571; final: 26.57 ± 0.87 g, p = 0.959). Additionally, the mice did not show significant differences in body weight gain when treated with MLT at 2 mg/kg (5.78 ± 0.39 g, p = 0.288) and 9 mg/kg (6.23 ± 0.39 g, p = 0.118) compared to the control group (5.03 ± 0.49 g) (Figure 3D).

3.2. MLT Improves the Plasmatic Lipid Profile and Reduces the Risk of Cardiovascular Disease

To investigate the lipid-lowering effect of MLT, the plasma lipid profile and the main cardiovascular risk indexes were analyzed. As shown in Table 1, a significant reduction was observed in LDL-C values (−24.6%, p = 0.008) in mice treated with 9 mg/kg MLT, compared to the control group. No significant differences were observed in the values of TC, TGs, or HDL-C between the MLT groups compared to the WD group (p > 0.05).
In addition, cardiovascular risk was evaluated in each group by calculating the indexes CRI I, CRI II, and AIP. Although no differences in CRI I or II were observed at the lowest concentration (2 mg/kg) (104.50 ± 12.27% and 110.80 ± 15.81%, respectively, p > 0.05), they were reduced at the highest concentration of MLT (9 mg/kg) (CRI I: 80.35 ± 7.04%, p = 0.016 and CRI II: 74.93 ± 6.27%, p = 0.016) (Figure 4). In addition, mice treated with MLT showed a reduction in the AIP of 51.74% and 42.31% at both concentrations of MLT tested (p < 0.05), respectively (Figure 4).

3.3. MLT Treatment Decreases Hepatic Lipids

As shown in Figure 5, the levels of TC (Figure 5A) and LDL-C (Figure 5C) were reduced by 21.57 and 14.17%, respectively, after treatment with 9 mg/kg MLT, without significant differences in 2 mg/kg MLT-treated mice. On the other hand, TG (Figure 5B) and HDL-C (Figure 5D) levels remained unchanged for both groups treated with MLT.

3.4. MLT Reduces Lymphocytosis

To determine the immune status after MLT treatment, the total number of leukocytes, as well as their subpopulations (lymphocytes, monocytes, and granulocytes) was quantified in the plasma sample.
As shown in Table 2, although there were no significant differences in the number of leukocytes in the mice treated with 2 mg/kg MLT compared to the control group (p > 0.05), the daily ingestion of 9 mg/kg MLT significantly reduced the number of leukocytes (−50.36%, p = 0.011). In particular, MLT treatment reduced the lymphocyte populations by 56% (p = 0.030), while the monocyte and granulocyte subpopulations were not altered by the MLT treatment.

3.5. MLT Reduces the Number of TNF Pro-Inflammatory Cytokines and Improves the Antioxidant Capacity

The immunomodulatory effect of MLT was corroborated by quantifying the pro-inflammatory cytokine TNF in the plasma of mice. As shown in Figure 6A, MLT treatment reduced the TNF concentration in a dose-dependent manner, with a statistical difference for the treatment with MLT 9 mg/kg being observed (a reduction of 52.6%, p = 0.044, with respect to the WD group). The antioxidant capacity of MLT was measured by the ABTS radical scavenging assay in plasma. Figure 6B shows a significant increase in the TEAC values in plasma of mice treated with MLT 9mg/kg. Specifically, the treatment increased the TEAC values by 58.5 ± 29.70% compared to the control group (WD). No differences were observed for 2 mg/kg MLT-treated mice.

4. Discussion

This study highlights the cardioprotective effect of MLT therapy in an ApoE−/− mouse model characterized by a greater susceptibility to cardiovascular accidents. In fact, ApoE−/− mice display poor lipoprotein clearance with subsequent accumulation of cholesterol ester-enriched particles in the blood, which promotes the development of atherosclerotic plaques [42].
The present study shows, for the first time, that treatment with MLT for 12 weeks reduces the LDL-C concentration and cardiovascular risk indexes in WD-fed ApoE−/− mice. Additionally, MLT treatment decreases the plasma levels of lymphocytes and improves the plasma antioxidant status. The effects of MLT were not related to the body weight gain of the mice, which remained unchanged for the three experimental groups.
High plasma concentrations of TC, TG, and LDL-C, as well as low plasma concentrations of HDL-C, among other things, are risk factors for the onset and progression of CVDs [25,41]. Similarly, a previous study showed that an increase of 1.0 mmol/L in LDL-C was associated with an increased absolute risk of myocardial infarction in individuals aged 70–100 years [43]. On the contrary, an increase of 1 mg/dL in HDL-C reduces the risk of coronary heart disease by 2% in men and 3% in women [44]. It is remarkable that in the present study, 12 weeks of MLT treatment at 9 mg/kg reduced plasma LDL-C by −24.6%, while HDL-C levels were not altered. Furthermore, a strong association has been shown between CVDs, metabolic dysfunction-associated fatty liver disease (MAFLD), and the accumulation of liver fat (steatosis) [45]. In this sense, the present study shows for the first time that treatment with 9 mg/kg MLT reduced the liver total cholesterol and LDL-C in WD-fed ApoE−/− mice, which is in agreement with previous studies performed in rats [46,47]. Although these previous studies showed the effect of MLT treatment on serum cholesterol and LDL-C concentration [48,49,50], none of these were performed in a specific model of hypercholesterolemia and cardiovascular disease. In fact, these previous studies were focused on the effect of MLT on a specific increase in LDL-C caused by nicotine administration [51], cigarette smoke [52], diabetes induction [53], or a diet modification [31,54].
In addition, MLT reduces the CRI I, CRI II, and AIP, which are used as optimal indicators of cardiovascular risk [41]. Bhardwaj et al. have reported that CRIs can contribute significantly to the estimation of the risk of coronary artery disease, especially when the absolute values of the plasma lipid parameters do not change markedly [55]. Furthermore, Quispe and colleagues proposed that the CRI I should be considered for additional risk assessment in the primary prevention population, specifically in high-CV-risk individuals, such as patients with diabetes [56]. Regarding the AIP ratio, this is a novel indicator of dyslipidemia, and patients with coronary artery disease (CAD) have significantly higher values compared to healthy controls [57]. Furthermore, the AIP has been described as an independent predictor of CAD [57]. Although it was previously shown that daily treatment of rats fed a high-cholesterol diet with intraperitoneal MLT at 12.5 mg/kg decreased the CRI II [31], there are no previous studies related to the effect of MLT on the CRI I and AIP. Thus, to our knowledge, this is the first study to describe the potential of this molecule to reduce the CRI I and II, and AIP in a mouse model with hypercholesterolemia and cardiovascular risk.
Taking into account the present results, together with previous studies, we suggest that MLT could be a good candidate to prevent the development of CVDs and treat pathologies that cause an imbalance in the lipid profile, such as metabolic syndrome and obesity, among others.
In addition, the role of MLT in the modulation of the immune system has been extensively studied over the last years in different contexts, such as in autoimmune diseases [58], infections [59], and even pathologies associated with metabolic syndrome, including neuroinflammation [60]. Mice fed with WD have an increased number of blood leukocytes and lymphocytes [23]. According to these data, elevated cholesterol levels are widely known to predispose individuals to a pro-inflammatory state through a systemic increase in leukocytes and, to a greater extent, the lymphocytes and soluble mediators secreted by these cells [61]. Furthermore, a previous study has shown that body fat affects the number of circulating leukocytes and lymphocytes in children [24]. In numerous studies, the capacity of MLT to modulate the immune response in different diseases has been described, as well as the association of this molecule with low blood levels of leukocytes and/or lymphocytes [29,62]. Winklewski et al. showed that MLT treatment significantly decreases the number of leukocytes and lymphocytes in ethanol-intoxicated mice [63], and other authors have shown that MLT treatment reduces the number of lymphocytes in animals with zymosan-induced peritonitis [64]. Furthermore, recent studies show the ability of MLT to not only control the number of immune cells such as leukocytes and/or lymphocytes in various pathological situations, but also to modify its pro/anti-inflammatory profile, favoring resolution of the disease [35,58,65]. However, there is no evidence of the effect of MLT on blood leukocyte and/or lymphocyte levels in mice fed a high-fat diet. This study is the first to report the effect of MLT on the levels of lymphocytes and leukocytes in ApoE−/− mice fed with a WD. Furthermore, since a decrease in the population of lymphocytes in the blood, as well as in the plasma levels of TNF, was observed in this study, we could say that MLT contributes to the decrease in the subpopulation of Th1 lymphocytes, being characterized by the production of TNF and involved in inflammatory processes [66]. Also, M1 pro-inflammatory macrophages are responsible for the production of TNF. Our results indicate a slight decrease in monocytes, which could contribute to the decrease in M1-phenotype macrophages and therefore to the decrease in TNF. This is in agreement with previous results which demonstrated that MLT promotes the polarization of macrophages to an M2-type anti-inflammatory profile [67]. In this way, MLT has been shown to reduce TNF levels in the blood in women with polycystic ovary syndrome, patients with COVID-19, and anemic patients with chronic kidney disease, among other populations [68,69,70].
Finally, oxidative stress is widely known for its role in the generation and development of cardiovascular diseases. ApoE−/− mice have been shown to have a high basal pro-oxidative status compared to C57BL/6 mice due to the antioxidant role of ApoE [71]. In the present work, oxidative stress was also increased through the intake of a WD. Furthermore, it has been widely demonstrated that the consumption of fat-rich diets increases oxidative stress, and in turn, increases the risk of developing cardiovascular disease [72]. In the present investigation, the intake of 9 mg/kg MLT daily for 12 weeks was capable of alleviating the oxidative effects caused by the consumption of the WD, with no improvements being observed when the daily concentration consumed was 2 mg/kg MLT. Due to the close relationship between oxidative stress and cardiovascular diseases, it is concluded that the consumption of MLT, in addition to leading to the previously mentioned effects, would reduce the risk of cardiovascular disease, through the reduction in oxidative stress. The fact that MLT controls the number of systemic lymphocytes as well as the lipid parameters in WD-fed ApoE−/− mice, a murine model of hypercholesterolemia, atherosclerosis, and metabolic syndrome, indicates that this molecule could be used to restore the lipid and anti-inflammatory imbalance generated in the development of cardiovascular events.
In addition, it is important to note that chronobiotic MLT is not the same as MLT, which is administered in high doses to treat different diseases. Both have different treatment guidelines to obtain the greatest effectiveness without harming the patient’s health. Specifically, therapy with high doses of MLT has been tested in patients with Charcot–Marie–Tooth neuropathy (70 mg/day for 6 months) [73], in multiple sclerosis (25 mg/day for 6 months) [74], and neoplastic cachexia (20 mg/day for 3 months) [75], among other diseases, and in all of them, there was evidence of its effectiveness as an anti-inflammatory or antioxidant without showing side effects that could compromise the patients’ health. Furthermore, a series of articles reinforce the hypothesis that increasing inflammatory responses leads to the suppression of nocturnal MLT production and that MLT administration to control inflammatory processes could at the same time compensate for this loss of nocturnal MLT [76,77,78,79]. In line with this, it would also be interesting to delve deeper into the possibilities offered by chronotherapy in MLT treatment in the future, the objective of which is to understand the impact that biological rhythms have on the response to a given therapy to optimize its action, maximize the benefits, and minimize possible adverse effects.
As a limitation of our study, the effect of MLT globally on the number of leukocytes and lymphocytes was analyzed, but not the effect that MLT could have on the different immune subpopulations. In fact, different subsets of white blood cells play different roles, and some are even opposite [80,81]. However, there are previous studies in which MLT therapy in an inflammatory context decreases the number of CD4 cells and macrophages, and more specifically the potentially pathogenic Th1 cells (characterized by the production of TNF), while promoting the regulatory responses mediated by Tregs [35,58]. Regarding macrophage subpopulations, MLT favors polarization from M1 (characterized by the production of TNF) towards the M2 profile, promoting an anti-inflammatory environment [67]. Our results support the hypothesis that MLT could reduce these pro-inflammatory subpopulations (Th1 cells and M1 macrophages) due to a decrease in TNF levels.
Given all the above and that MLT is a pleiotropic molecule with a wide range of functions, we suggest that the consumption of MLT could also control other key risk factors, such as oxidative stress, involved in these pathologies.

5. Conclusions

MLT could be considered a functional ingredient to prevent or treat the development of CVDs derived from high cholesterol intake, since in addition to controlling plasma and liver TC and LDL-C levels and reducing CV risk indexes, it decreases systemic inflammation and oxidative stress due to excessive fat consumption. However, more research is needed to decipher the molecular mechanisms by which MLT exerts these actions and to develop clinical trials to determine the effect of MLT in patients with cardiovascular risk.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/nutraceuticals4020016/s1, Table S1: Western diet composition.

Author Contributions

The following are the authors’ contributions: Conceptualization: A.C.-V., I.C.-C. and P.J.L.; Methodology: G.S.-S., I.C.-C., A.I.Á.-R., A.I.Á.-L. and E.P.-E.; Resources: A.C.-V.; Formal analysis: G.S.-S., I.C.-C., A.I.Á.-L. and E.P.-E.; Drafting of the manuscript: G.S.-S., A.I.Á.-L. and A.C.-V.; Supervision: A.C.-V., I.C.-C. and P.J.L.; Funding acquisition: A.C.-V. and P.J.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Andalusian Government Ministry of Health PC-0111-2016-0111, PEMP-0085-2020 (co-financed with FEDER funds, Resolution of 7 July 2021 of the General Secretary for Research, Development and Innovation in Health, which called for grants to finance research, development, and innovation in biomedicine and health sciences in Andalusia by 2021) and the PAIDI Program from the Andalusian Government (CTS160). G.S.-S. was supported by FPU grants from the Spanish Ministerio de Educación, Cultura y Deporte, (FPU16/02339). I.C.-C. was supported by a postdoctoral fellowship from the Andalusian Government Ministry of Economy, Knowledge, Business, and University (DOC_00587/2020). A.I.Á.-L. was funded by the Andalusian Government Ministry of Health (PI-0136-2019 and PEMP-0085-2020). E.P.-E. was supported by the VI Program of Inner Initiative for Research and Transfer of University of Seville (VI PPIT-US).

Institutional Review Board Statement

All experimental procedures were conducted under the Spanish legislation and under the EU Directive 2010/63/EU for animal experiments and was approved by the Virgen Macarena and Virgen del Rocío University Hospitals Ethical Committee (reference 21/06/2016/105).

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are included within the article and Supplementary Materials.

Acknowledgments

We thank all the staff from the IBiS Animal Facility for their valuable assistance.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. WHO Organization. Cardiovascular Diseases (CVDs). Available online: https://www.who.int/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds) (accessed on 20 February 2024).
  2. Roth, G.A.; Mensah, G.A.; Johnson, C.O.; Addolorato, G.; Ammirati, E.; Baddour, L.M.; Barengo, N.C.; Beaton, A.Z.; Benjamin, E.J.; Benziger, C.P. Global burden of cardiovascular diseases and risk factors, 1990–2019: Update from the GBD 2019 study. J. Am. Coll. Cardiol. 2020, 76, 2982–3021. [Google Scholar] [CrossRef]
  3. Anderson, K.M.; Castelli, W.P.; Levy, D. Cholesterol and mortality: 30 years of follow-up from the Framingham study. JAMA 1987, 257, 2176–2180. [Google Scholar] [CrossRef]
  4. Berger, S.; Raman, G.; Vishwanathan, R.; Jacques, P.F.; Johnson, E.J. Dietary cholesterol and cardiovascular disease: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2015, 102, 276–294. [Google Scholar] [CrossRef]
  5. Targher, G.; Bonapace, S.; Byrne, C. Does high LDL-cholesterol cause cardiovascular disease? Expert Rev. Clin. Pharmacol. 2019, 12, 91. [Google Scholar] [CrossRef] [PubMed]
  6. Trinick, T.; Duly, E. Hyperlipidemia: Overview; Caballero, B., Ed.; Academic Press: Waltham, MA, USA, 2013. [Google Scholar]
  7. Bardagjy, A.S.; Steinberg, F.M. Relationship between HDL functional characteristics and cardiovascular health and potential impact of dietary patterns: A narrative review. Nutrients 2019, 11, 1231. [Google Scholar] [CrossRef] [PubMed]
  8. Criqui, M.H.; Golomb, B.A. Epidemiologic aspects of lipid abnormalities. Am. J. Med. 1998, 105, 48s–57s. [Google Scholar] [CrossRef] [PubMed]
  9. Akpınar, O.; Bozkurt, A.; Acartürk, E.; Seydaoğlu, G. A new index (CHOLINDEX) in detecting coronary artery disease risk. Anadolu Kardiyol. Derg. 2013, 13, 315–319. [Google Scholar] [CrossRef]
  10. Edwards, M.K.; Blaha, M.J.; Loprinzi, P.D. Atherogenic Index of Plasma and Triglyceride/High-Density Lipoprotein Cholesterol Ratio Predict Mortality Risk Better Than Individual Cholesterol Risk Factors, Among an Older Adult Population. Mayo Clin. Proc. 2017, 92, 680–681. [Google Scholar] [CrossRef]
  11. Dobiásová, M.; Frohlich, J.; Sedová, M.; Cheung, M.C.; Brown, B.G. Cholesterol esterification and atherogenic index of plasma correlate with lipoprotein size and findings on coronary angiography. J. Lipid Res. 2011, 52, 566–571. [Google Scholar] [CrossRef]
  12. Cai, G.; Shi, G.; Xue, S.; Lu, W. The atherogenic index of plasma is a strong and independent predictor for coronary artery disease in the Chinese Han population. Medicine 2017, 96, e8058. [Google Scholar] [CrossRef]
  13. Olamoyegun, M.A.; Oluyombo, R.; Asaolu, S.O. Evaluation of dyslipidemia, lipid ratios, and atherogenic index as cardiovascular risk factors among semi-urban dwellers in Nigeria. Ann. Afr. Med. 2016, 15, 194–199. [Google Scholar] [CrossRef] [PubMed]
  14. Millán, J.; Pintó, X.; Muñoz, A.; Zúñiga, M.; Rubiés-Prat, J.; Pallardo, L.F.; Masana, L.; Mangas, A.; Hernández-Mijares, A.; González-Santos, P.; et al. Lipoprotein ratios: Physiological significance and clinical usefulness in cardiovascular prevention. Vasc. Health Risk Manag. 2009, 5, 757–765. [Google Scholar] [PubMed]
  15. Hadaegh, F.; Khalili, D.; Ghasemi, A.; Tohidi, M.; Sheikholeslami, F.; Azizi, F. Triglyceride/HDL-cholesterol ratio is an independent predictor for coronary heart disease in a population of Iranian men. Nutr. Metab. Cardiovasc. Dis. 2009, 19, 401–408. [Google Scholar] [CrossRef] [PubMed]
  16. Bittner, V.; Johnson, B.D.; Zineh, I.; Rogers, W.J.; Vido, D.; Marroquin, O.C.; Bairey-Merz, C.N.; Sopko, G. The TG/HDL cholesterol ratio predicts all cause mortality in women with suspected myocardial ischemia a report from the Women’s ischemia syndrome evaluation (WISE). Am. Heart J. 2009, 157, 548. [Google Scholar] [CrossRef]
  17. Bertoluci, M.C.; Quadros, A.S.; Sarmento-Leite, R.; Schaan, B.D. Insulin resistance and triglyceride/HDLc index are associated with coronary artery disease. Diabetol. Metab. Syndr. 2010, 2, 11. [Google Scholar] [CrossRef] [PubMed]
  18. Madjid, M.; Fatemi, O. Components of the complete blood count as risk predictors for coronary heart disease: In-depth review and update. Tex. Heart Inst. J. 2013, 40, 17–29. [Google Scholar] [PubMed]
  19. Hubler, M.J.; Kennedy, A.J. Role of lipids in the metabolism and activation of immune cells. J. Nutr. Biochem. 2016, 34, 1–7. [Google Scholar] [CrossRef] [PubMed]
  20. Winer, S.; Chan, Y.; Paltser, G.; Truong, D.; Tsui, H.; Bahrami, J.; Dorfman, R.; Wang, Y.; Zielenski, J.; Mastronardi, F.; et al. Normalization of obesity-associated insulin resistance through immunotherapy. Nat. Med. 2009, 15, 921–929. [Google Scholar] [CrossRef]
  21. Keusch, G.T. The history of nutrition: Malnutrition, infection and immunity. J. Nutr. 2003, 133, 336S–340S. [Google Scholar] [CrossRef]
  22. Mustafa, A.; Ward, A.; Treasure, J.; Peakman, M. T lymphocyte subpopulations in anorexia nervosa and refeeding. Clin. Immunol. Immunopathol. 1997, 82, 282–289. [Google Scholar] [CrossRef]
  23. Maysami, S.; Haley, M.J.; Gorenkova, N.; Krishnan, S.; McColl, B.W.; Lawrence, C.B. Prolonged diet-induced obesity in mice modifies the inflammatory response and leads to worse outcome after stroke. J. Neuroinflammation 2015, 12, 140. [Google Scholar] [CrossRef] [PubMed]
  24. Zaldivar, F.; McMurray, R.; Nemet, D.; Galassetti, P.; Mills, P.; Cooper, D. Body fat and circulating leukocytes in children. Int. J. Obes. 2006, 30, 906–911. [Google Scholar] [CrossRef] [PubMed]
  25. Dubois-Deruy, E.; Peugnet, V.; Turkieh, A.; Pinet, F. Oxidative stress in cardiovascular diseases. Antioxidants 2020, 9, 864. [Google Scholar] [CrossRef] [PubMed]
  26. Mourino-Alvarez, L.; Sastre-Oliva, T.; Corbacho-Alonso, N.; Barderas, M.G. Oxidative Stress in Cardiovascular Diseases. In Importance of Oxidative Stress and Antioxidant System in Health and Disease; IntechOpen: London, UK, 2022. [Google Scholar]
  27. Zawilska, J.B.; Skene, D.J.; Arendt, J. Physiology and pharmacology of melatonin in relation to biological rhythms. Pharmacol. Rep. 2009, 61, 383–410. [Google Scholar] [CrossRef] [PubMed]
  28. Chen, C.-Q.; Fichna, J.; Bashashati, M.; Li, Y.-Y.; Storr, M. Distribution, function and physiological role of melatonin in the lower gut. World J. Gastroenterol. WJG 2011, 17, 3888. [Google Scholar] [CrossRef] [PubMed]
  29. Carrillo-Vico, A.; Lardone, P.J.; Álvarez-Sánchez, N.; Rodríguez-Rodríguez, A.; Guerrero, J.M. Melatonin: Buffering the immune system. Int. J. Mol. Sci. 2013, 14, 8638–8683. [Google Scholar] [CrossRef] [PubMed]
  30. Hussain, S.A.R. Effect of melatonin on cholesterol absorption in rats. J. Pineal Res. 2007, 42, 267–271. [Google Scholar] [CrossRef] [PubMed]
  31. Chan, T.; Tang, P. Effect of melatonin on the maintenance of cholesterol homeostasis in the rat. Endocr. Res. 1995, 21, 681–696. [Google Scholar] [CrossRef]
  32. Elias, A.; Nelson, B.; Obianime, W.A. Beneficial effects of melatonin and alpha lipoic acid on lopinavir/ritonavir-induced alterations in lipid and glucose levels in male albino rats. J. Med. Sci. 2016, 85, 46–53. [Google Scholar]
  33. Allagui, M.S.; Hachani, R.; Saidi, S.; Feriani, A.; Murat, J.C.; Kacem, K. Pleiotropic protective roles of melatonin against aluminium-induced toxicity in rats. General. Physiol. Biophys. 2015, 34, 415–424. [Google Scholar]
  34. Mohammadi-Sartang, M.; Ghorbani, M.; Mazloom, Z. Effects of melatonin supplementation on blood lipid concentrations: A systematic review and meta-analysis of randomized controlled trials. Clin. Nutr. 2018, 37, 1943–1954. [Google Scholar] [CrossRef] [PubMed]
  35. Álvarez-Sánchez, N.; Cruz-Chamorro, I.; Díaz-Sánchez, M.; Sarmiento-Soto, H.; Medrano-Campillo, P.; Martínez-López, A.; Lardone, P.J.; Guerrero, J.M.; Carrillo-Vico, A. Melatonin reduces inflammatory response in peripheral T helper lymphocytes from relapsing-remitting multiple sclerosis patients. J. Pineal Res. 2017, 63, e12442. [Google Scholar] [CrossRef] [PubMed]
  36. Carrillo-Vico, A.; Lardone, P.J.; Naji, L.; Fernández-Santos, J.M.; Martín-Lacave, I.; Guerrero, J.M.; Calvo, J.R. Beneficial pleiotropic actions of melatonin in an experimental model of septic shock in mice: Regulation of pro-/anti-inflammatory cytokine network, protection against oxidative damage and anti-apoptotic effects. J. Pineal Res. 2005, 39, 400–408. [Google Scholar] [CrossRef] [PubMed]
  37. Nabavi, S.M.; Nabavi, S.F.; Sureda, A.; Xiao, J.; Dehpour, A.R.; Shirooie, S.; Silva, A.S.; Baldi, A.; Khan, H.; Daglia, M. Anti-inflammatory effects of Melatonin: A mechanistic review. Crit. Rev. Food Sci. Nutr. 2019, 59, S4–S16. [Google Scholar] [CrossRef] [PubMed]
  38. Kurniawan, M.F.; Utami, S.B.; Fulyani, F.; Kresnoadi, E.; Wicaksono, S.A. Melatonin prevented the elevation of leukocyte count and the decreased of hematocrit levels in burn-induced Wistar Rats. Bali Med. J. 2021, 10, 668–672. [Google Scholar] [CrossRef]
  39. Cheikh, M.; Makhlouf, K.; Ghattassi, K.; Graja, A.; Ferchichi, S.; Kallel, C.; Houda, M.; Souissi, N.; Hammouda, O. Melatonin ingestion after exhaustive late-evening exercise attenuate muscle damage, oxidative stress, and inflammation during intense short term effort in the following day in teenage athletes. Chronobiol. Int. 2020, 37, 236–247. [Google Scholar] [CrossRef] [PubMed]
  40. Reagan-Shaw, S.; Nihal, M.; Ahmad, N. Dose translation from animal to human studies revisited. FASEB J. 2008, 22, 659–661. [Google Scholar] [CrossRef] [PubMed]
  41. Kalelioglu, T.; Genc, A.; Karamustafalioglu, N.; Emul, M. Assessment of cardiovascular risk via atherogenic indices in patients with bipolar disorder manic episode and alterations with treatment. Diabetes Metab. Syndr. Clin. Res. Rev. 2017, 11, S473–S475. [Google Scholar] [CrossRef] [PubMed]
  42. Lo Sasso, G.; Schlage, W.K.; Boué, S.; Veljkovic, E.; Peitsch, M.C.; Hoeng, J. The Apoe−/− mouse model: A suitable model to study cardiovascular and respiratory diseases in the context of cigarette smoke exposure and harm reduction. J. Transl. Med. 2016, 14, 146. [Google Scholar] [CrossRef]
  43. Mortensen, M.B.; Nordestgaard, B.G. Elevated LDL cholesterol and increased risk of myocardial infarction and atherosclerotic cardiovascular disease in individuals aged 70–100 years: A contemporary primary prevention cohort. Lancet 2020, 396, 1644–1652. [Google Scholar] [CrossRef]
  44. Gordon, D.J.; Probstfield, J.L.; Garrison, R.J.; Neaton, J.D.; Castelli, W.P.; Knoke, J.D.; Jacobs Jr, D.R.; Bangdiwala, S.; Tyroler, H.A. High-density lipoprotein cholesterol and cardiovascular disease. Four prospective American studies. Circulation 1989, 79, 8–15. [Google Scholar] [CrossRef] [PubMed]
  45. Roca-Fernandez, A.; Banerjee, R.; Thomaides-Brears, H.; Telford, A.; Sanyal, A.; Neubauer, S.; Nichols, T.E.; Raman, B.; McCracken, C.; Petersen, S.E. Liver disease is a significant risk factor for cardiovascular outcomes-a UK Biobank study. J. Hepatol. 2023, 79, 1085–1095. [Google Scholar] [CrossRef] [PubMed]
  46. Pan, M.; Song, Y.L.; Xu, J.M.; Gan, H.Z. Melatonin ameliorates nonalcoholic fatty liver induced by high-fat diet in rats. J. Pineal Res. 2006, 41, 79–84. [Google Scholar] [CrossRef] [PubMed]
  47. Sun, H.; Huang, F.-f.; Qu, S. Melatonin: A potential intervention for hepatic steatosis. Lipids Health Dis. 2015, 14, 75. [Google Scholar] [CrossRef] [PubMed]
  48. Shabani, A.; Foroozanfard, F.; Kavossian, E.; Aghadavod, E.; Ostadmohammadi, V.; Reiter, R.J.; Eftekhar, T.; Asemi, Z. Effects of melatonin administration on mental health parameters, metabolic and genetic profiles in women with polycystic ovary syndrome: A randomized, double-blind, placebo-controlled trial. J. Affect. Disord. 2019, 250, 51–56. [Google Scholar] [CrossRef] [PubMed]
  49. Cichoz-Lach, H.; Celinski, K.; Konturek, P.; Konturek, S.; Slomka, M. The effects of L-tryptophan and melatonin on selected biochemical parameters in patients with steatohepatitis. J. Physiol. Pharmacol. 2010, 61, 577. [Google Scholar] [PubMed]
  50. Agil, A.; Navarro-Alarcón, M.; Ruiz, R.; Abuhamadah, S.; El-Mir, M.Y.; Vázquez, G.F. Beneficial effects of melatonin on obesity and lipid profile in young Zucker diabetic fatty rats. J. Pineal Res. 2011, 50, 207–212. [Google Scholar] [CrossRef] [PubMed]
  51. Hendawy, A.K.; El-Toukhey, N.E.S.; AbdEl-Rahman, S.S.; Ahmed, H.H. Ameliorating effect of melatonin against nicotine induced lung and heart toxicity in rats. Environ. Sci. Pollut. Res. 2021, 28, 35628–35641. [Google Scholar] [CrossRef] [PubMed]
  52. Ma, B.; Chen, Y.; Wang, X.; Zhang, R.; Niu, S.; Ni, L.; Di, X.; Han, Q.; Liu, C. Cigarette smoke exposure impairs lipid metabolism by decreasing low-density lipoprotein receptor expression in hepatocytes. Lipids Health Dis. 2020, 19, 88. [Google Scholar] [CrossRef]
  53. Ebaid, H.; Bashandy, S.A.; Abdel-Mageed, A.M.; Al-Tamimi, J.; Hassan, I.; Alhazza, I.M. Folic acid and melatonin mitigate diabetic nephropathy in rats via inhibition of oxidative stress. Nutr. Metab. 2020, 17, 6. [Google Scholar] [CrossRef]
  54. Wang, L.; McFadden, J.W.; Yang, G.; Zhu, H.; Lian, H.; Fu, T.; Sun, Y.; Gao, T.; Li, M. Effect of melatonin on visceral fat deposition, lipid metabolism and hepatic lipo-metabolic gene expression in male rats. J. Anim. Physiol. Anim. Nutr. 2021, 105, 787–796. [Google Scholar] [CrossRef]
  55. Bhardwaj, S.; Bhattacharjee, J.; Bhatnagar, M.; Tyagi, S.; Delhi, N. Atherogenic index of plasma, castelli risk index and atherogenic coefficient-new parameters in assessing cardiovascular risk. Int. J. Pharm. Biol. Sci. 2013, 3, 359–364. [Google Scholar]
  56. Quispe, R.; Elshazly, M.B.; Zhao, D.; Toth, P.P.; Puri, R.; Virani, S.S.; Blumenthal, R.S.; Martin, S.S.; Jones, S.R.; Michos, E.D. Total cholesterol/HDL-cholesterol ratio discordance with LDL-cholesterol and non-HDL-cholesterol and incidence of atherosclerotic cardiovascular disease in primary prevention: The ARIC study. Eur. J. Prev. Cardiol. 2020, 27, 1597–1605. [Google Scholar] [CrossRef] [PubMed]
  57. Wu, T.T.; Gao, Y.; Zheng, Y.Y.; Ma, Y.T.; Xie, X. Atherogenic index of plasma (AIP): A novel predictive indicator for the coronary artery disease in postmenopausal women. Lipids Health Dis. 2018, 17, 197. [Google Scholar] [CrossRef] [PubMed]
  58. Álvarez-Sánchez, N.; Cruz-Chamorro, I.; López-González, A.; Utrilla, J.C.; Fernández-Santos, J.M.; Martínez-López, A.; Lardone, P.J.; Guerrero, J.M.; Carrillo-Vico, A. Melatonin controls experimental autoimmune encephalomyelitis by altering the T effector/regulatory balance. Brain Behav. Immun. 2015, 50, 101–114. [Google Scholar] [CrossRef]
  59. Begum, R.; Mamun-Or-Rashid, A.N.M.; Lucy, T.T.; Pramanik, M.K.; Sil, B.K.; Mukerjee, N.; Tagde, P.; Yagi, M.; Yonei, Y. Potential Therapeutic Approach of Melatonin against Omicron and Some Other Variants of SARS-CoV-2. Molecules 2022, 27, 6934. [Google Scholar] [CrossRef]
  60. Mansouri, S.; Salari, A.A.; Abedi, A.; Mohammadi, P.; Amani, M. Melatonin treatment improves cognitive deficits by altering inflammatory and neurotrophic factors in the hippocampus of obese mice. Physiol. Behav. 2022, 254, 113919. [Google Scholar] [CrossRef] [PubMed]
  61. Ellulu, M.S.; Patimah, I.; Khaza’ai, H.; Rahmat, A.; Abed, Y. Obesity and inflammation: The linking mechanism and the complications. Arch. Med. Sci. 2017, 13, 851–863. [Google Scholar] [CrossRef] [PubMed]
  62. Obayashi, K.; Saeki, K.; Kurumatani, N. Higher melatonin secretion is associated with lower leukocyte and platelet counts in the general elderly population: The HEIJO-KYO cohort. J. Pineal Res. 2015, 58, 227–233. [Google Scholar] [CrossRef]
  63. Kurhaluk, N.; Sliuta, A.; Kyriienko, S.; Winklewski, P.J. Melatonin restores white blood cell count, diminishes glycated haemoglobin level and prevents liver, kidney and muscle oxidative stress in mice exposed to acute ethanol intoxication. Alcohol Alcohol. 2017, 52, 521–528. [Google Scholar] [CrossRef]
  64. Kepka, M.; Szwejser, E.; Pijanowski, L.; Verburg-van Kemenade, B.L.; Chadzinska, M. A role for melatonin in maintaining the pro-and anti-inflammatory balance by influencing leukocyte migration and apoptosis in carp. Dev. Comp. Immunol. 2015, 53, 179–190. [Google Scholar] [CrossRef]
  65. Medrano-Campillo, P.; Sarmiento-Soto, H.; Álvarez-Sánchez, N.; Álvarez-Ríos, A.I.; Guerrero, J.M.; Rodríguez-Prieto, I.; Castillo-Palma, M.J.; Lardone, P.J.; Carrillo-Vico, A. Evaluation of the immunomodulatory effect of melatonin on the T-cell response in peripheral blood from systemic lupus erythematosus patients. J. Pineal Res. 2015, 58, 219–226. [Google Scholar] [CrossRef]
  66. Moss, R.B.; Moll, T.; El-Kalay, M.; Kohne, C.; Soo Hoo, W.; Encinas, J.; Carlo, D.J. Th1/Th2 cells in inflammatory disease states: Therapeutic implications. Expert. Opin. Biol. Ther. 2004, 4, 1887–1896. [Google Scholar] [CrossRef]
  67. Nardo, L.; Rezzani, R.; Facchetti, L.; Favero, G.; Franco, C.; Abdelhafez, Y.G.; Badawi, R.D.; Guindani, M.; Seo, Y.; Pampaloni, M. Beneficial effects of melatonin on apolipoprotein-E knockout mice by morphological and 18F-FDG PET/CT assessments. Int. J. Mol. Sci. 2020, 21, 2920. [Google Scholar] [CrossRef] [PubMed]
  68. Mousavi, R.; Alizadeh, M.; Asghari Jafarabadi, M.; Heidari, L.; Nikbakht, R.; Babaahmadi Rezaei, H.; Karandish, M. Effects of melatonin and/or magnesium supplementation on biomarkers of inflammation and oxidative stress in women with polycystic ovary syndrome: A randomized, double-blind, placebo-controlled trial. Biol. Trace Elem. Res. 2022, 200, 1010–1019. [Google Scholar] [CrossRef] [PubMed]
  69. Esmaeili Gouvarchin Ghaleh, H.; Hosseini, A.; Aghamollaei, H.; Fasihi-Ramandi, M.; Alishiri, G.; Saeedi-Boroujeni, A.; Hassanpour, K.; Mahmoudian-Sani, M.-R.; Farnoosh, G. NLRP3 inflammasome activation and oxidative stress status in the mild and moderate SARS-CoV-2 infected patients: Impact of melatonin as a medicinal supplement. Z. Naturforsch. C 2022, 77, 37–42. [Google Scholar] [CrossRef]
  70. Hameed, E.N.; Tukmagi, H.A.; Haydar, F.; Allami, H.C. Melatonin improves erythropoietin hyporesponsiveness via suppression of inflammation. Rev. Recent. Clin. Trials 2019, 14, 203–208. [Google Scholar] [CrossRef]
  71. Pereira, S.S.; Teixeira, L.G.; Aguilar, E.C.; Matoso, R.O.; Soares, F.L.; Ferreira, A.V.; Alvarez-Leite, J.I. Differences in adipose tissue inflammation and oxidative status in C57BL/6 and ApoE−/− mice fed high fat diet. Anim. Sci. J. 2012, 83, 549–555. [Google Scholar] [CrossRef] [PubMed]
  72. Jiang, S.; Liu, H.; Li, C. Dietary regulation of oxidative stress in chronic metabolic diseases. Foods 2021, 10, 1854. [Google Scholar] [CrossRef]
  73. Chahbouni, M.; López, M.D.S.; Molina-Carballo, A.; De Haro, T.; Muñoz-Hoyos, A.; Fernández-Ortiz, M.; Guerra-Librero, A.; Acuña-Castroviejo, D. Melatonin treatment reduces oxidative damage and normalizes plasma pro-inflammatory cytokines in patients suffering from Charcot-Marie-Tooth neuropathy: A pilot study in three children. Molecules 2017, 22, 1728. [Google Scholar] [CrossRef]
  74. Sánchez-López, A.L.; Ortiz, G.G.; Pacheco-Moises, F.P.; Mireles-Ramírez, M.A.; Bitzer-Quintero, O.K.; Delgado-Lara, D.L.; Ramírez-Jirano, L.J.; Velázquez-Brizuela, I.E. Efficacy of melatonin on serum pro-inflammatory cytokines and oxidative stress markers in relapsing remitting multiple sclerosis. Arch. Med. Res. 2018, 49, 391–398. [Google Scholar] [CrossRef] [PubMed]
  75. Lissoni, P.; Paolorossi, F.; Tancini, G.; Barni, S.; Ardizzoia, A.; Brivio, F.; Zubelewicz, B.; Chatikhine, V. Is there a role for melatonin in the treatment of neoplastic cachexia? Eur. J. Cancer 1996, 32, 1340–1343. [Google Scholar] [CrossRef]
  76. Fernandes, P.A.; Cecon, E.; Markus, R.P.; Ferreira, Z.S. Effect of TNF-α on the melatonin synthetic pathway in the rat pineal gland: Basis for a ‘feedback’of the immune response on circadian timing. J. Pineal Res. 2006, 41, 344–350. [Google Scholar] [CrossRef] [PubMed]
  77. Pontes, G.N.; Cardoso, E.C.; Carneiro-Sampaio, M.M.; Markus, R.P. Injury switches melatonin production source from endocrine (pineal) to paracrine (phagocytes)–melatonin in human colostrum and colostrum phagocytes. J. Pineal Res. 2006, 41, 136–141. [Google Scholar] [CrossRef] [PubMed]
  78. Pontes, G.N.; Cardoso, E.C.; Carneiro-Sampaio, M.M.; Markus, R.P. Pineal melatonin and the innate immune response: The TNF-α increase after cesarean section suppresses nocturnal melatonin production. J. Pineal Res. 2007, 43, 365–371. [Google Scholar] [CrossRef] [PubMed]
  79. Tamura, E.K.; Fernandes, P.A.; Marçola, M.; Cruz-Machado, S.d.S.; Markus, R.P. Long-lasting priming of endothelial cells by plasma melatonin levels. PLoS ONE 2010, 5, e13958. [Google Scholar] [CrossRef] [PubMed]
  80. Zielinski, C.E. T helper cell subsets: Diversification of the field. Eur. J. Immunol. 2023, 53, 2250218. [Google Scholar] [CrossRef]
  81. Hou, P.; Fang, J.; Liu, Z.; Shi, Y.; Agostini, M.; Bernassola, F.; Bove, P.; Candi, E.; Rovella, V.; Sica, G. Macrophage polarization and metabolism in atherosclerosis. Cell Death Dis. 2023, 14, 691. [Google Scholar] [CrossRef]
Figure 1. Experimental design of the study. ApoE−/−, apolipoprotein E knockout mice; MLT, melatonin; WD, Western diet.
Figure 1. Experimental design of the study. ApoE−/−, apolipoprotein E knockout mice; MLT, melatonin; WD, Western diet.
Nutraceuticals 04 00016 g001
Figure 2. Chemical structure of melatonin ((A), CAS number: 73-31-4) and equivalent doses of it in mice and humans (B). MW, molecular weight.
Figure 2. Chemical structure of melatonin ((A), CAS number: 73-31-4) and equivalent doses of it in mice and humans (B). MW, molecular weight.
Nutraceuticals 04 00016 g002
Figure 3. Body weight monitored over time (A), basal body weight (B), final weight (C), and body weight gain (D) in the in vivo experiments. Data were represented as mean ± SEM. n.s., not significant. MLT, melatonin; WD, Western diet.
Figure 3. Body weight monitored over time (A), basal body weight (B), final weight (C), and body weight gain (D) in the in vivo experiments. Data were represented as mean ± SEM. n.s., not significant. MLT, melatonin; WD, Western diet.
Nutraceuticals 04 00016 g003
Figure 4. Evaluation of cardiovascular disease risk through Castelli risk index I (TC/HDL-C) (A) and II (LDL-C/HDL-C) (B) and atherogenic index of plasma (Log(TG/HDL-C)) (C). Results are expressed as a percentage of the control group and represent the mean ± SEM of each group. * p ≤ 0.05, ** p ≤ 0.01 vs. WD group; HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol; MLT, melatonin; TC, total cholesterol; TGs, triglycerides; WD, Western diet.
Figure 4. Evaluation of cardiovascular disease risk through Castelli risk index I (TC/HDL-C) (A) and II (LDL-C/HDL-C) (B) and atherogenic index of plasma (Log(TG/HDL-C)) (C). Results are expressed as a percentage of the control group and represent the mean ± SEM of each group. * p ≤ 0.05, ** p ≤ 0.01 vs. WD group; HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol; MLT, melatonin; TC, total cholesterol; TGs, triglycerides; WD, Western diet.
Nutraceuticals 04 00016 g004
Figure 5. Hepatic TC (A), TG (B), LDL-C (C), and HDL-C (D) content in the three experimental groups. Results are expressed as a percentage of the control group and represent the mean ± SEM of each group. * p ≤ 0.05 vs. WD. n.s., not significant. HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol; MLT, melatonin; TC, total cholesterol; TGs, triglycerides; WD, Western diet.
Figure 5. Hepatic TC (A), TG (B), LDL-C (C), and HDL-C (D) content in the three experimental groups. Results are expressed as a percentage of the control group and represent the mean ± SEM of each group. * p ≤ 0.05 vs. WD. n.s., not significant. HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol; MLT, melatonin; TC, total cholesterol; TGs, triglycerides; WD, Western diet.
Nutraceuticals 04 00016 g005
Figure 6. Effect of MLT treatment on plasma anti-inflammatory and antioxidant status evaluated by an enzyme-linked immunosorbent assay (ELISA) (A) and the Trolox equivalent antioxidant capacity (TEAC) assay (B). Results are expressed as a percentage of the control group and represent the mean ± SEM of each group. * p ≤ 0.05 with respect to the Western diet (WD) group. MLT, melatonin.
Figure 6. Effect of MLT treatment on plasma anti-inflammatory and antioxidant status evaluated by an enzyme-linked immunosorbent assay (ELISA) (A) and the Trolox equivalent antioxidant capacity (TEAC) assay (B). Results are expressed as a percentage of the control group and represent the mean ± SEM of each group. * p ≤ 0.05 with respect to the Western diet (WD) group. MLT, melatonin.
Nutraceuticals 04 00016 g006
Table 1. Plasma lipid profile.
Table 1. Plasma lipid profile.
Biochemical
Parameter
WD
(mg/dL)
WD + MLT
(2 mg/kg)
(% of Control)
p-ValueWD + MLT
(9 mg/kg)
(% of Control)
p-Value
TC506.10 ± 12.34106.20 ± 8.810.54887.12 ± 6.590.151
TG99.38 ± 0.72110.80 ± 14.890.706105.20 ± 9.370.683
LDL-C373.20 ± 36.81112.20 ± 12.100.64375.43 ± 4.390.008
HDL-C113.70 ± 33.59103.40 ± 5.410.548111.00 ± 11.160.691
Results are expressed as a percentage of the control group and represent the mean ± SEM of each group. TC, total cholesterol; TGs, triglycerides; LDL-C, low-density lipoprotein cholesterol; HDL-C, high-density lipoprotein cholesterol; MLT, melatonin.
Table 2. Hemogram.
Table 2. Hemogram.
Cells (×103/µL)WDMLT
(2 mg/kg)
MLT
(9 mg/kg)
Leukocytes1.49 ± 0.211.14 ± 0.160.74 ± 0.15 *
Lymphocytes0.99 ± 0.180.76 ± 0.140.43 ± 0.13 *
Monocytes0.069 ± 0.0240.067 ± 0.0210.062 ± 0.032
Granulocytes0.37 ± 0.060.31 ± 0.030.33 ± 0.04
Results are expressed as the number of cells ± SEM of each experimental group. *, p ≤ 0.05 with respect to the control group. MLT, melatonin; WD, Western diet.
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

Santos-Sánchez, G.; Álvarez-López, A.I.; Ponce-España, E.; Álvarez-Ríos, A.I.; Lardone, P.J.; Carrillo-Vico, A.; Cruz-Chamorro, I. Melatonin Modulates Lipid Metabolism and Reduces Cardiovascular Risk in Apolipoprotein E-Deficient Mice Fed a Western Diet. Nutraceuticals 2024, 4, 260-272. https://doi.org/10.3390/nutraceuticals4020016

AMA Style

Santos-Sánchez G, Álvarez-López AI, Ponce-España E, Álvarez-Ríos AI, Lardone PJ, Carrillo-Vico A, Cruz-Chamorro I. Melatonin Modulates Lipid Metabolism and Reduces Cardiovascular Risk in Apolipoprotein E-Deficient Mice Fed a Western Diet. Nutraceuticals. 2024; 4(2):260-272. https://doi.org/10.3390/nutraceuticals4020016

Chicago/Turabian Style

Santos-Sánchez, Guillermo, Ana Isabel Álvarez-López, Eduardo Ponce-España, Ana Isabel Álvarez-Ríos, Patricia Judith Lardone, Antonio Carrillo-Vico, and Ivan Cruz-Chamorro. 2024. "Melatonin Modulates Lipid Metabolism and Reduces Cardiovascular Risk in Apolipoprotein E-Deficient Mice Fed a Western Diet" Nutraceuticals 4, no. 2: 260-272. https://doi.org/10.3390/nutraceuticals4020016

Article Metrics

Back to TopTop