Next Article in Journal
Light Quality Potentiates the Antioxidant Properties of Brassica rapa Microgreen Extracts against Oxidative Stress and DNA Damage in Human Cells
Previous Article in Journal
β-Caryophyllene Inhibits Oxaliplatin-Induced Peripheral Neuropathy in Mice: Role of Cannabinoid Type 2 Receptors, Oxidative Stress and Neuroinflammation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Inflammation and Oxidative Stress Induced by Obesity, Gestational Diabetes, and Preeclampsia in Pregnancy: Role of High-Density Lipoproteins as Vectors for Bioactive Compounds

by
Angélica Saraí Jiménez-Osorio
1,
Elizabeth Carreón-Torres
2,
Emmanuel Correa-Solís
3,
Julieta Ángel-García
1,
José Arias-Rico
1,
Octavio Jiménez-Garza
1,
Lizbeth Morales-Castillejos
1,
Hugo Alexander Díaz-Zuleta
4,
Rosa María Baltazar-Tellez
1,
María Luisa Sánchez-Padilla
1,
Olga Rocío Flores-Chávez
1 and
Diego Estrada-Luna
1,*
1
Área Académica de Enfermería, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado Hida go, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico
2
Department of Molecular Biology, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Sección XVI, Tlalpan, Mexico City 14080, Mexico
3
Instituto de Farmacobiología, Universidad de la Cañada, Carretera Teotitlán-San Antonio Nanahuatipán Km 1.7 s/n., Paraje Titlacuatitla, Teotitlán de Flores Magón 68540, Oaxaca, Mexico
4
Facultad de Ciencias de la Salud, Universidad de Ciencias Aplicadas y Ambientales, Cl. 222 #54-21, Bogotá 111166, Colombia
*
Author to whom correspondence should be addressed.
Antioxidants 2023, 12(10), 1894; https://doi.org/10.3390/antiox12101894
Submission received: 6 September 2023 / Revised: 5 October 2023 / Accepted: 13 October 2023 / Published: 23 October 2023

Abstract

:
Inflammation and oxidative stress are essential components in a myriad of pathogenic entities that lead to metabolic and chronic diseases. Moreover, inflammation in its different phases is necessary for the initiation and maintenance of a healthy pregnancy. Therefore, an equilibrium between a necessary/pathologic level of inflammation and oxidative stress during pregnancy is needed to avoid disease development. High-density lipoproteins (HDL) are important for a healthy pregnancy and a good neonatal outcome. Their role in fetal development during challenging situations is vital for maintaining the equilibrium. However, in certain conditions, such as obesity, diabetes, and other cardiovascular diseases, it has been observed that HDL loses its protective properties, becoming dysfunctional. Bioactive compounds have been widely studied as mediators of inflammation and oxidative stress in different diseases, but their mechanisms of action are still unknown. Nonetheless, these agents, which are obtained from functional foods, increase the concentration of HDL, TRC, and antioxidant activity. Therefore, this review first summarizes several mechanisms of HDL participation in the equilibrium between inflammation and oxidative stress. Second, it gives an insight into how HDL may act as a vector for bioactive compounds. Third, it describes the relationships between the inflammation process in pregnancy and HDL activity. Consequently, different databases were used, including MEDLINE, PubMed, and Scopus, where scientific articles published in the English language up to 2023 were identified.

1. Introduction

Pregnancy is a physiological process that extends the development phase of the fetus in the uterus. In this period, women experience a series of adjustments to satisfy the metabolic, biochemical, endocrine, and cellular changes that fetuses demand [1]. Nevertheless, depending on the woman’s wellness, these adaptations may be tolerated or be a burden in the development of certain diseases [2], such as obesity, hypertension, hyperlipidemia, metabolic syndrome, diabetes mellitus, preeclampsia, and preterm birth, which in turn, increases the risk of cardiovascular diseases and may even produce death [3,4]. Also, these metabolic disorders are characterized by the presence of endothelial dysfunction, inflammatory, pro-oxidant, and lipid profile dysregulation processes that increase the levels of total cholesterol, triglycerides, low-density lipoprotein cholesterol (c-LDL), and dysfunctional high-density lipoproteins (HDL) [5,6]. Obesity, diabetes, and preeclampsia induce oxidative stress conditions on their own through a number of mechanisms or pathways, including superoxide anion generation, protein kinase-C activation, hyperglycemia, elevated cholesterol and triglyceride levels, mitochondrial dysfunction, low antioxidant system, chronic inflammation, and reactive oxygen species (ROS) generation, especially during the postprandial period [7,8,9,10]. In pregnancy, these mechanisms exacerbate oxidative stress (OS) and inflammation processes with deleterious effects on the fetus and child’s development, along with the damage to the mother.
Foods, fruits, herbs, and seeds are widely used in diverse cultures for the acute treatment of symptoms related to pain or inflammation, and in a long-term fashion to control the negative effects of some chronic diseases. Their method of consumption passes from one generation to the next, and it is part of several cultures and customs of diverse societies [11,12,13]. In some cultures, the consumption of plants during pregnancy is common. For example, plants are used to treat nausea, vomiting, or dizziness; nonetheless, their effectiveness in chronic degenerative diseases experienced by pregnant women has not been fully elucidated [14,15]. Therefore, the purpose of this review is to provide the most recent and relevant information concerning the role high-density lipoproteins play as vectors of bioactive compounds with a protective role in the processes of inflammation and OS involved in metabolic diseases that are frequently present during pregnancy, such as gestational diabetes, obesity, and preeclampsia.

2. Methods

This narrative review was carried out by considering scientific articles of interest for the topic, apprising those related to pregnancy and inflammation, and pregnancy and oxidative stress. These processes were considered factors for predisposition to chronic diseases for the mother and the fetus during the gestational stages such as childhood and adulthood. As selection criteria, the following descriptors were used: inflammation, oxidative stress, pregnancy, high-density lipoproteins, metabolic diseases, and bioactive compounds. To search for primary and secondary information sources, electronic databases such as MEDLINE, PubMed, and Scopus were used, finding original papers published from 1982 to August 2023. Selected articles for bibliographic research were categorized by clinical relevance, short-term (prematurity, preeclampsia, gestational diabetes) and long-term (diabetes mellitus, obesity, dyslipidemia) outcome term, type of population (animal models, humans) that was used, outcomes of the behavior of the inflammatory markers that were stated, and the effect of bioactive compounds on lipoproteins during pregnancy. Ultimately, a total of 207 references were selected, which were available in full text and in the English language.

3. Bioactive Compounds Contained in Foods Used during Pregnancy

Over the course of human history, some herbs, plants, and fruits have been used extensively in different cultures and societies to treat injuries, diseases, viruses, and bacterial infections, and even to enhance women’s health during pregnancy to facilitate childbirth [16]. In pregnancy, the particular use of plants and herbs is focused mostly on alleviating conditions, such as diarrhea, chills, nausea, constipation, vomiting, or to increase milk production and accelerate labor. Several studies have reported that the mentioned capabilities are attributed to the bioactive compounds contained in herbs, fruits, spices, and other foods possessing anti-inflammatory [17], antimicrobial [18], antioxidant [19], cardioprotective [20], hepatoprotective [21], hypoglycemic [22], neuroprotective [23], immunomodulatory [24], or hypolipidemic [25] properties. Some of these bioactive compounds include phytochemicals, phenolic compounds, polysaccharides, and unsaturated fatty acids. During pregnancy, the consumption and functionality of these compounds have been established [26], especially in the prevention of diseases and health promotion for both mother and fetus in the different phases of the gestational period [27,28,29].
The consumption of certain polyphenolic compounds in pregnancy improved levels of blood glucose in women with gestational diabetes [30], as well as insulin secretion in the fetus and in the mother [31]. Furthermore, it has been reported that some polyphenolic compounds regulate cell apoptosis, the secretion of adipokines, placentation, and arterial pressure. Such regulation occurs mainly through the activity of the matrix metalloproteinases (MMPs) [32,33] and some glucose transporter (GLUT) family members (GLUT1, GLUT4, and GLUT 9) [34].
A wide variety of secondary metabolites including flavonoids, quinones, flavones, tannins, and aromatic compounds are consumed in high frequency and quantity. Nonetheless, they could present a toxic effect mainly on the fetus because some of those metabolites are widely consumed during the first trimester. In fact, some of them are well known to be potentially dangerous because of the vulnerability of the fetus, for example, Japanese mint (Mentha arvensis), linden herb (Justicia pectoralis), verbena (Verbena officinalis), aloe (Aloe vera), chamomile (Matricaria recutita), oregano (Plecthranthus amboinicus), basil (Ocicum basilicum), caña santa (Cymbopoggon citratus), and wormwood (Artemisia abssinthium). Furthermore, some of these compounds could increase estrogenic activity by stimulating uterine contractility, increasing blood pressure, and having a hepatotoxic, mutagenic, or fetal malformation effect. Most of these agents may also have an effect on OS and inflammatory processes present in gestational diabetes mellitus (GDM), preeclampsia, or obesity during pregnancy because they contain phenolic compounds, flavonoids, tannins, omega-3, quercetin, kaempferol, tocopherols, and vitamin C [35,36,37,38,39,40].

4. Pregnancy–Oxidative Stress–Inflammation Association

A significant increase in inflammatory processes and OS markers during pregnancy happens because of biological actions that are carried out to maintain body homeostasis. The first reports showed an elevation in OS and inflammatory markers in plasma and urine during the physiological course of pregnancy [41,42]. The significant increase in inflammatory and pro-oxidant molecules can trigger negative conditions such as preeclampsia, endothelial dysfunction, pre-diabetes, poor fetal development, or premature birth [43,44].
One of the reasons for the increase in OS is the development of the placenta, which contains high quantities of mitochondria, leading to an increase in the concentration of ROS, such as superoxide dismutase (SOD), xanthine oxidase (XO), or NADPH oxidase (NOX). The high activity of these prooxidant enzymes is associated with the onset of preeclampsia, which is one of the most common negative conditions during pregnancy, as well as an increase in cholesterol, triglycerides, and c-LDL levels [45,46]. Nevertheless, a number of external factors are related to OS augmentation during pregnancy, such as smoking, consumption of foods rich in saturated fatty acids, carbohydrates, alcohol consumption, ultraviolet radiation, obesity, and consumption of non-steroidal anti-inflammatory drugs like paracetamol, ibuprofen, or diclofenac [47,48].
ROS play a crucial role throughout the entire process of pregnancy. It starts with the generation of steroid hormones in the ovary, oocyte maturation, ovulation, luteolysis, implantation, follicular growth, and maintenance of pregnancy until childbirth [49]. Elevated ROS production leads to a decreased ovarian blood flow and tissue damage, polycystic ovarian syndrome, or endometriosis. In addition, inflammatory processes are important and play an important role in both the positive and negative aspects, alongside important functions of the adaptive immune system; during the first and third trimester, there is a presence of pro-inflammatory processes, triggered by implantation and placentation, as well as labor and delivery, whereas, in the second trimester, anti-inflammatory processes proliferate [50,51], ensuring proper fetal growth at this stage.
During gestation, women’s bodies possess diverse hormones, cytokines, nutrients, and enzymes that help maintain the balance of antioxidant and inflammatory processes (Figure 1) [52]. Advanced oxidation protein products (AOPPs) and C-reactive protein (CRP) levels in the maternal serum of pregnant women were significantly higher in the first and second trimesters compared to non-pregnant women. However, higher levels of AOPP were found in control men than in control non-pregnant women and were similar to pregnant women. Moreover, the CRP concentrations increased gradually from the first to the second trimester [38]. In contrast, an increase in OS was observed in the second trimester. The activity of the antioxidant enzyme glutathione peroxidase (GPx) was lower in pregnant women than non-pregnant women and postpartum women. Also, the oxidative damage marker to DNA, 8-hydroxy-2′-deoxyguanosine (8-OHdG), was higher in pregnant women compared to non-pregnant women [53].
During the third gestational stage, healthy pregnant women showed higher lipid peroxidation markers, such as malondialdehyde (MDA) in plasma and erythrocytes, than non-pregnant women, as well as lower levels of erythrocyte glutathione (GSH) levels than non-pregnant women [41]. Furthermore, a longitudinal study consisting of pregnant women at 30 weeks of gestational age showed that, after delivery, their products decreased levels of Ne-carboxymethyl lysine. Also, a soluble receptor for advanced glycation end products (sRAGE) was observed without changes in tumor necrosis factor alpha (TNF-α), CRP, and MDA in blood samples. Their newborns were healthy, but the newborns’ levels of sRAGE, TNF-α, and MDA were higher than their mothers at delivery [54]. Additionally, factors such as age and race influence OS and inflammation. Older pregnant women (age > 40 years) with uncomplicated pregnancies had lower anti-inflammatory markers, such as interleukin-10 (IL-10) and interleukin-1 receptor antagonist (IL-RA), and an increase in total antioxidant capacity (TAC) than younger pregnant women [55]. In Nigerian pregnant women, levels of vitamin E, an endogenous antioxidant, were observed to be lower than in pregnant women from the USA. However, levels of vitamin E depend on the isoform. Thus, in the US population, higher maternal α-tocopherol levels were associated with birth length (>10th percentile), while higher cord levels of α-tocopherol were associated with birth length (<10th percentile); these results were not observed in the Nigerian population, although the levels of delta-tocopherol were higher than maternal samples from the USA [56].
Undoubtedly, OS and inflammation are major factors in the development of diseases that are considered the first cause of death and morbidity in women during pregnancy since they can exacerbate negative conditions for the development of cardiovascular diseases and diabetes [3,57,58].
The inflammatory response during pregnancy is caused by toll-like receptor (TLR) activation, while inducing the activation of the inflammasome complex, to perpetuate the production of cytokines and chemokines in placenta and endometrium, such as monocyte chemotactic protein 1 (MCP-1), IL-8, IL1-β, and IL-6. This TLR activation triggers immune cell recruitment and the production of prostaglandins and MMPs, which leads to cervical cell activation and uterine contractions [59]. According to the stage of pregnancy, in vaginal epithelium, exocervix, endocervix, endometrium, and fallopian tubes, TLRs 1–5 induce the release of cytokines, chemokines, and antimicrobial peptides that modulate both proinflammatory and anti-inflammatory responses. Hence, it can promote or attenuate the onset of chronic diseases or adverse clinical conditions (Table 1) [60].
The mechanisms of inflammation suggested at this stage of life involve the role of type 1 and 2 T Helper (Th) cells, where Th1 cells are responsible for increased levels of interferon gamma (IFN-γ), interleukin 2 (IL-2), and tumor necrosis factor beta (TNF-β), which are responsible for phagocyte-dependent inflammation, as well as protection against intracellular pathogens. On the other hand, Th2 cells produce IL-4, IL-5, IL-6, IL-9, IL-10, and IL-13 and induce an antibody response by downregulating B cells and activating eosinophils, thus inhibiting phagocytic cell activity [51].

5. OS and Inflammation during Pregnancy, Metabolic Changes in Chronic Diseases

Currently, gestational obesity and diabetes are associated with fetal disorders affecting the proteomic profile. An increase in markers of inflammation and OS is observed, as well as a decrease in the antioxidant system or impairment of vasorelaxation [68]. All of these characteristics could be linked to the appearance of neonatal hypoglycemia, respiratory distress syndrome, fetal macrosomia, platelet hyperaggregability, or the development of cardiovascular disease early in life [69,70]. Also, obesity is associated with a 1.3- to 3.8-fold increase in the incidence of preeclampsia or GDM [59], short-term hemorrhage, low APGAR score, congenital birth defects, and admission to neonatal intensive care [51]. Meanwhile, in the long term, an increased risk of cardiovascular disease and diabetes mellitus type 2 (DM2) in children [51,63,71,72].

5.1. Preeclampsia

Among the prominent hypertensive disorders in pregnancy is preeclampsia. It is a multifactorial disorder, one of the most common conditions during pregnancy on which hypertension (≥140/90 mmHg) and proteinuria (≥300 mg of protein in 24 h) are predominant, leading to dysfunction in several organs, poor placental development, and even maternal or fetal death [73,74]. Preeclampsia regularly develops from the placenta, and its early stages are known as placental syndrome. This syndrome is characterized by endothelial damage and dysfunction in different tissues accompanied by excessive free radical formation [75,76,77]. Hence, some potential OS markers such as ischemia-modified albumin (IMA), uric acid (UA), MDA, and metals like zinc (Zn), copper (Cu), and selenium (Se) have been used as diagnostic biomarkers for preeclampsia [10]. Inflammatory processes exacerbate the effects of preeclampsia, being IL-1α and IL-1β mainly secreted and expressed in the placenta. However, the involvement of other members of the interleukin family such as IL-18, IL-33, IL-37, and IL-38 has also been described [78]. Leakage of trophoblasts into the uterine wall accentuates inflammation and OS, leading to the presence of hypoxia and an increased placental ROS concentration [79]
Obesity, preeclampsia, and GDM pregnancy have an altered lipid metabolism, specifically related to low concentrations of lipoproteins and their lipid composition (free and esterified cholesterol, phospholipids, and triglycerides). Therefore, women with these conditions become susceptible to OS, and this results in an inhibition in the production of nitric oxide (NO). NO is an important vasodilator at the endothelial level, and it can lead to hypertensive disorders, ischemia, or stroke [80].
Increased inflammatory processes are observed from the first trimester in pregnant women at high risk of hypertension (Table 2), although, in the third trimester, a significant increase in proinflammatory cytokines and oxidative markers are observed in women who develop preeclampsia [81,82,83,84]. In late pregnancy, the inflammatory processes are characterized by elevated cytokines such as IL-1Ra, IL2, IL4, IL6, IL8, IL10, IL12p40, IL12p70, IL18, TNF-α [85,86,87,88], increased CRP [87,89,90,91], and increased adhesion molecules, such as the vascular cell adhesion molecule (VCAM-1), intracellular adhesion molecule (ICAM) [86,87,88], and proteins associated with endothelial dysfunction like the soluble L-selectin (sL-selectin) in the umbilical cord [87], and in obesity-associated inflammatory processes characterized by increased leptin [92,93].
OS during early pregnancy is characterized by increased thiobarbituric acid reactive substances (TBARS) and 8 isoprostane [82,83]. The increase in the lipid peroxidation marker, MDA, was observed from the second trimester, accompanied by a decrease in antioxidant defense measured by GSH levels as well as SOD and CAT activity [94]. However, during the third trimester and delivery, there is increased MDA [85,88,91,95], TBARS [87,96], xanthine oxidase activity, 8-isoprostane [89,92,97,98], and maintenance of CAT by SOD and GPx activities [95,99,100]. Enhanced antioxidant system activity reflects impaired redox state maintenance in the third trimester, and it is observed by increased lipid peroxidation and GSH/GSSG ratio.
Table 2. Markers of OS and inflammation in pregnant women with preeclampsia.
Table 2. Markers of OS and inflammation in pregnant women with preeclampsia.
Study GroupsOxidative and Inflammatory ChangesEffects in Mothers/NeonatesRef.
Follow-up from 1st to 3rd trimester
1st–2nd trim = 22
3rd trim = 11
>35 GW = 9
↑ HIF1A mRNA since 1st trimester in PW who later developed PE
↑ MIF, ENG, FLT1, and BACE2 mRNA at week 24–30, and only ENG remains high at 31–34 weeks in PW who developed PE
ND[81]
Group I
(risk of HAT without PE) = 82
Group II
(risk of HAT developing PE) = 43
1st trimester in Group II vs. Group I:
↑ IL-6, TNF-α, IFNγ, TBARS, TAC
2nd trimester in Group II vs. Group I:
↑ hsCRP, TNF-α, IFNγ, TBARS, TAC
3rd trimester in Group II vs. Group I and 3rd vs. 2nd trimester:
↑ IL-6, hsCRP, TNF-α, IFNγ, TBARS, TAC
All parameters correlated with baroreflex sensitivity except BMI and TAC. TBARS had a significant independent contribution to BRS.[82]
Follow up from 1st trimester to 3rd trimester n = 441↑ CRP, TNF-α, and 8-isoprostane early in pregnancy (median 10 weeks)
CRP of PE is similar to non-PE at the end of pregnancy
↑ TNF-α in PE across pregnancy and ↑ 8-isoprostane in PE in early gestation.
No changes in IL-1, IL-6, IL-10 and 8-OHdG
50 PW developed PE
The strongest associations were observed at approximately 18 weeks of pregnancy. TNF-α and 8-isoprostane were consistently elevated at all four time points in pregnancy. 8-OHdG was found as a protective factor against PE.
[83]
Follow up of 96 PW with high risk of PE at 16th week and next at 36th week↑ MDA, hsCRP, and IL-6 at 36th week compared to 16th week of pregnancy
↓ Baroreflex sensitivity and NO
Decreased BRS and increased interleukin-6 are associated with reduction in NO[84]
Second trimester
PE = 49
Ctrl = 50
↑ MDA, Cardiac-specific troponin-I, and
↓ CAT, SOD, and GSH in PE vs. Ctrl
Association between SOD and MDA, and CAT with MDA. CAT correlated with hsCRP[94]
Third and delivery
PE = 20
Ctrl = 18
↑ 8-isoprostane in plasma only in post-partum
↓ urinary excretion of 8-iso-prostane in PE
The variation of urinary 8-iso-prostane depends on creatinine clearance and plasmatic endothelin-1[97]
PE = 10
Ctrl = 11
↑ AOPP in PE vs. non-pregnant women but not with Ctrl
↑ CRP of PE vs. Ctrl
ND[42]
PE = 19
Ctrl = 18
Non PW = 20
↑ Plasmatic and urinary 8-iso-prostane in PE and Ctrl vs. non-PW but not in PE vs. Ctrl
↓ α-tocopherol in PE and Ctrl vs. Non PW
γ-tocopherol decreased in PE vs. Ctrl and vs. non-PW
ND[98]
Mild PE = 47
Severe PE = 36
Ctrl = 50
↑ sFlt-1/PlGF, hs-CRP, 8-isoprostane and leptin and lower adiponectin in PE vs. Ctrl.Significant positive correlations in sFlt-1/PlGF and hs-CRP or leptin.
A weak inverse correlation emerged between sFlt-1/PlGF and adiponectin.
[92]
Non-pregnant controls = 10
PE = 28
Ctrl = 59
↑ Superoxide and ferritin in PE vs. controlDuring HP, superoxide concentrations correlate significantly with arterial stiffness, while in PE, superoxide is significantly correlated to microvascular endothelial function.[101]
PE = 15
Ctrl = 28
↑ AGEs in PE vs. Ctrl+ Correlation between circulating levels of AGEs and gamma-glutamyl transpeptidase, uric acid, glucose, insulin, and HOMA-IR in PE[102]
Non-PW = 20
PW = 20
Ctrl = 20
↑ Serum resistin, vaspin, MDA, and IL-8 in PE vs. non-PE and CtrlOnly resistin and vaspin were reduced in PE after four weeks of postpartum[85]
Mild PE = 45
Severe PE = 40
Ctrl = 48
↓ TAC and ↑ TBARS, IL-6, TNF-α and IFN-γ in mild and severe PE groups compared to Ctrl+ Correlation of circulating beta-human chorionic gonadotropin and OS and inflammatory markers[86]
PE = 27
Ctrl = 43
MPO levels in placenta higher than the levels in normal control subjectsND[103]
PE = 5
Ctrl = 5
In preeclamptic women, intense xanthine oxidase immunoreactivity was present within the epidermis.ND[104]
PE = 67
Ctrl = 70
↑ CRP, fetal DNA, MDA and Hsp70 levelIn PE serum, Hsp70 levels showed significant correlations with serum CRP, aspartate aminotransferase, and LDH activities MDA[86]
PE = 53
Ctrl = 20
Mild PE = 32
Severe PE = 21
↑ hsCRP, IL-6, TNF-α and 8-isoprostane in PE vs. Ctrl and in severe PE vs. mild PE
No changes in MDA
Plasma levels of 8-isoprostane were significantly correlated with the plasma levels of hs-CRP, IL-6, and TNF-α in patients with PE[89]
PE = 71
Ctrl = 71
In amniotic fluid: ↑ sFLT1, sEndoglin, endothelin 1, and leptin in PE.
sFLT1, sEndoglin, leptin, and adiponectin ↑ in PE IUGR than those without IUGR.
Leptin has the largest area under the curve (0.753). Amniotic proteins are involved in the inflammatory process of the human placenta.[93]
PE = 44
Ctrl = 44
↑ serum CRP, plasma MDAin PE vs. CtrlPlasma OPN concentrations are increased in
preeclamptic patients with extensive endothelial injury.
[95]
PE = 60
Ctrl = 60
↑ Hsp70, IL-1Ra, IL2, IL4, IL6, IL8, IL, IL10, IL12p40, IL12p70, IL18, INF&, IP10, MCP-1, ICAM-1, VCAM-1, sFlt-1, P1GF in PE vs. CtrlElevated serum Hsp70 level and sFlt-1/PlGF ratio had a synergistic (joint) effect in the risk of preeclampsia[86]
PE = 46
Ctrl = 42
↑ TBARS, TBARS/TAS, IL-6, TNF-a, a1-antitrypsin, CRP in plasma of PE vs. Ctrl
↑ CRP and a1-Antitrypsin in umbilical cord of PE vs. Ctrl
↑ sVCAM, sL-selectin in leucocytes of PE vs. Ctrl
+ Correlation between maternal and UCB TAS, IL-6, CRP in Ctrl and PE[87]
PE = 33
Ctrl = 33
↑ IL6, MDA, CP, in plasma and placenta of PE vs. Ctrl
↑ TLR-4 and NF-kB in the placenta
ND[88]
PE = 50 (17 mild and 33 severe)
Ctrl = 33
↑ MDA in PE mild vs. PE severe
↑ IL-6, IL-10, TNF-a in PE vs. Ctrl
↑ IL-6/IL-10 ratio in PE vs. Ctrl
ND[91]
PE = 18
Ctr = 18
↑ CAT, Il-6, TNF-α in PE vs. Ctrl
Plasma Mg was positively correlated with CAT and GPx activities with concentrations of IL-6 and TNF-α
Plasma magnesium and urinary 8-isoprostane were associated with PE[99]
PE = 100
Ctr = 50
↑ SOD, GSH/GSSG ratio, IL-6, and ↓ GSSG in placenta of PE vs. Ctrl+ Associations between placental GSH levels with weight, head, and chest circumference and gestational age at birth[100]
Abbreviations: 8-OHdG, 8-hydroxy-2′-deoxyguanosine; AOPP, advanced oxidation protein products; AGE, advanced glycation end products; BACE2, β-site APP-cleaving enzyme-2; BRS, baroreflex sensitivity; CAT, catalase; Ctrl, control group (pregnant women without preeclampsia); ENG, endoglin; FLT1, fms-related tyrosine kinase-1; GPx, glutathione peroxidase; GSH, glutathione; GSSG, oxidized GSH; GW, gestational week; HIF1a, Hypoxia-inducible factor 1-alpha; hsCRP, high sensitivity C Reactive Protein; Hsp70, heat shock proteins; ICAM-1, intracellular adhesion molecule; IFNγ, interferon gamma; IL, interleukin; IUGR, fetal intrauterine growth restriction; MCP1, monocyte chemotactic protein 1; MDA, malondialdehyde; MIF, macrophage migration inhibitory factor; MPO, myeloperoxidase; NF-kB, nuclear factor kappa B; NO, nitric oxide; OPN, osteoponin; PE, preeclampsia; PlGF, placental growth factor; PW, pregnant women; sFlt-1, soluble fms-like tyrosine kinase receptor-1; SOD, superoxide dismutase; TAC, total antioxidant capacity; TBARS, thiobarbituric acid reactive substances; TLR-4, toll-like receptor; TNF-α, tumor necrosis factor alpha; trim, trimester; VCAM-1, vascular cell adhesion molecule; VEGF, vascular endothelial growth factor.

5.2. Obesity

A common condition observed is the accumulation of lipids during the first trimester that decreases towards the end of the third trimester. Conversely, lipolysis decreases in the first trimester and increases by the third trimester. Similarly, glucose tolerance and insulin resistance behave in the same way, respectively [105]. Moreover, there is a high variability in lipid, protein, and carbohydrate metabolism, which are regulated by immunometabolic and immunoinflammatory processes, including proinflammatory cytokines and ROS production [106,107]. Nevertheless, when antioxidant–ROS balance is disturbed, extreme OS occurs, leading to abnormal placentation with increased vascular resistance inside the placenta, thereby allowing OS to overwhelm antioxidants, to shorten telomeres, and, ultimately, to lead to cellular senescence [108]. Also, when pregnant women develop preeclampsia, the processes of apoptosis, autophagy, and cellular senescence, which play a critical role in placental and fetal homeostasis and growth, appear disrupted, increasing susceptibility to negative conditions in the fetus and the mother [109]. Among the conditions that present an alteration in metabolic and inflammatory processes is obesity, which is a chronic disease considered a risk factor for the development of preeclampsia, GDM, anemia, complications or death in childbirth in women, increased cardiovascular risk, insulin resistance, hyperinsulinemia, and prevalence of inflammatory processes in the neonate [110,111,112]. Usually, pregnant women with obesity at pre-pregnancy or becoming obese have a lipotoxic placental environment leading to increased OS and elevated concentrations of TNF-α, IL-1, IL-1β, IL-3, IL-4 and IL-6, and IFN-γ. These cytokines also regulate the activity and expression of placental fatty acid transporters, and, during the second or third trimester, they downregulate some metabolic pathways then increase the levels of total cholesterol, triglycerides, and very low-density lipoproteins (VLDL) [112,113,114].
Clinical studies show increased lipid peroxidation (MDA, LOOH) [115,116,117,118,119] and significant changes in antioxidant status (total antioxidant capacity and antioxidant enzyme activity) in pregnant women with obesity [119,120,121,122,123] during the third trimester of pregnancy (Table 3). Another study has shown an increase in lipid peroxidation and protein carbonylation (PC) in pregnant women with obesity versus controls, only in the third trimester, but not in the first and second trimesters of pregnancy. However, an increase in TNF-α was observed at 12–13 weeks of gestation, also in women with obesity who developed GDM. The ratio of reduced oxidized glutathione (GSH/GSSG) is an indirect marker of redox status [115]. In addition, increased antioxidant activity of SOD and CAT was observed in the placenta [122] and umbilical cord [119] of obese women compared to non-obese women, and compared to plasma activities during the third trimester of pregnancy. These results reveal necessary changes to maintain the homeostatic redox state in the placenta and the umbilical cord at the expense of maternal plasma redox. Inflammatory conditions have also been associated with reduced adiponectin levels in the third trimester [117,118,119,120,121,122,123,124]. Maternal BMI has been shown to have a significant positive correlation with increased levels of CRP, PC, GSSG/GSH, IL-6 ratio, and salivary CT [117,120,121]. These associations could be affected by hyperglycemia [113,116,119] and maternal age [115]. The association of mother and newborn in inflammatory and oxidative statuses revealed a positive correlation in the cord SOD activity of newborns with an increased SOD activity of normoglycemic ones. MDA levels in newborns were lower in term neonates and those with mothers who consumed vitamin supplements [119].

5.3. Gestational Diabetes Mellitus

The most frequent endocrinological complication in pregnancy, worldwide, is obesity, and it plays an important role in the development of GDM. Also, it is related to negative neonatal and obstetric conditions in the mother and infant at both cardiovascular and neurological levels [125,126]. Commonly, GDM is defined as an abnormal glucose tolerance that results in the development of hyperglycemia during pregnancy, and it can be triggered by obesity, a variation in adipokine production, and other external conditions, including maternal age, a diet rich in saturated fatty acids, family history of diabetes, and hypertensive processes during pregnancy [127]. The molecular pathophysiology of GDM has been extensively investigated. Proteomic studies in plasma from women with GDM have revealed changes in inflammation and OS markers, insulin resistance, blood coagulation, and lipid homeostasis during the second trimester of pregnancy, as well as elevated CRP in the first trimester of women who developed GDM [128] (Table 4).
Inflammatory processes are a key factor in the development of GDM, as increased activity or expression of inflammatory markers negatively alters insulin receptors, resulting in insulin resistance [129]. In addition to TNF-α, IL-6, adiponectin, resistin, or leptin that are involved in obesity, other biomarkers are included, such as the retinol-binding protein-4 (RBP-4), visfatin, adipocyte fatty acid-binding protein (AFAABP), and some novel proteins like visceral adipose tissue-derived serpin A12 (vaspin) [130], apelin [131], and omentin, which may be involved in the development of GDM and in the progression of obesity [132].
During the second trimester of pregnancy, OS and inflammation are established in GDM by an increase in F2 isoprostanes [133,134], ROS in lymphocytes [135], total OS, and OS index in serum [134], as well as an increase in the acute phase reactants, which are independent determinants of GDM after adjustment for BMI [135,136] (Table 4). At the end of pregnancy, there is an increase in antioxidant enzymes, like CAT and GSR in placenta [137]. In contrast, other studies did not show an increase in protein carbonylation [138] or TBARS [139]. However, in studies with a bigger sample size, an increase in MDA, hsCRP, and IL-6 in the plasma of pregnant women with GDM was observed to be associated with stress and postnatal depression [140].
Both GDM and obesity complications constitute independent risk factors that increase problems during delivery and that are associated with an increase in monocyte count in cord blood and with the expression of several genes like the silent information regulator sirtuin 1 (SIRT1) and uncoupling protein 1 (UCP1) in obese pregnant women [139], while growth/differentiation factor 15 (GDF-15) is increased in GDM [141]. Also, an increase in mitochondrial DNA levels, but dysfunctional syncytiotrophoblast mitochondria with morphological abnormalities [142], has been observed in the placenta of obese pregnant women who developed GDM. These physical abnormalities could lead to impaired pregnancy outcomes and future hypertensive [143,144] and neurological disorders [145].
Table 4. Inflammation and OS markers in pregnant women with GDM.
Table 4. Inflammation and OS markers in pregnant women with GDM.
Study GroupsOxidative and Inflammatory MarkersOxidative Stress ChangesInflammation ChangesEffects in Mothers and NewbornRef.
GDM = 22
Ctrl = 22
Proteome profiling in plasma↓ PON/LAC3 in GDM vs. Ctrl↑ CRP and ↓ IGFBP2 in GDM vs. CtrlStrong correlations among the inflammation-related proteins and proteins of blood coagulation, lipid homeostasis membrane, and antioxidative enzymes[128]
GDM = 11
Ctrl = 23
Six F2-isoprostanes isomers↑ 8-iso-15(R)-PGF2α levels in GDM vs. CtrlNDDelta-6-desaturase (D6D) activity index, calculated using fatty acid ratios, was 9% lower in pre-existing diabetes than in controls[146]
GDM = 48
Ctrl = 46
Total oxidative stress (TOS), total antioxidant status (TAS), and oxidative stress index (OSI) in serum↑ Serum OGTT, OSI, TOS in GDM vs. CtrlNDND[134]
GDM = 60
Ctrl = 75
CRP, NLR and PLR, PCT, and VAP-1ND↑ CRP, NLR, PLR, PCT and VAP-1 in GDM vs. Ctrl+ Correlation of VAP-1 with glucose, HbA1c, PLR, and CRP[136]
GDM = 114
Ctrl = 114
ROS (DCFH-DA) and DNA damage (comet assay) in lymphocytesNLR and MVP in blood↑ ROS and DNA damage in GDM vs. Ctrl↑ NLR and mean platelet volume (MPV) in GDM vs. CtrlThe elevated parameters are independent determinants of GDM after adjustment for BMI[135]
GDM = 30CRP, ferritin, TNF-α, methylglyoxal, glycated albuminND↑ PAF and TNF-α after diet
↑ MGO in GDM vs. non-pregnant healthy people
+ Correlation of MGO levels with HbA1c, pregnancy weight and HOMA-IR at GDM diagnosis and after 12 weeks[147]
GDM = 40
Ctrl = 40
GDF-15 in serumND↑ Serum GDF-15 levels in patients with GDM vs. Ctrl+ Correlation of GDF-15 and BMI for both GDM and Ctrl
↓ Apgar scores at 1 min and 5 min in the GDM vs. Ctrl
[141]
GDM = 176
Ctrl = 164
MDA, hsCRP, IL-6, and NO in plasma↑ MDA and ↓ NO in GDM vs. Ctrl↑ hsCRP and IL-6 in GDM vs. CtrlSignificant association of HbA1C, MDA, and interleukin-6 with maternal stress and postnatal depression[140]
GDM = 17
Ctrl = 23
CAT, GPX, GSR, and SOD gene expression in placenta and omental and subcutaneous adipose tissue↑ CAT and GSR in placenta of GDM vs. Ctrl
No changes in GPx or SOD in placenta and no changes in adipose tissue
Treatment with hypoxanthine/xanthine oxidase-stimulated cytokine release (IL1B, MIP1b, and TNF-α) and TNF-α mRNA expressionND[137]
GDM = 8
T2D = 3
T1D = 12
Ctrl = 13
IL-8, MCP-1, and CP in plasma and umbilical cordNo changes in CP in mothers with DM vs. CtrlNo changes in MCP-1 and IL-8 in mothers with DM vs. Ctrl
↑ MCP-1 and CP in umbilical cords from mothers with DM
MCP-1 correlated with ketone bodies and acetoacetate[138]
GDM = 69
PWOb = 44
PWOw = 48
Ctrl = 104
TBARS in placenta and UCP2, SIRT1, PPARα, TLR4, and GR-a gene expression in placenta tissue↑ UCP2 expression in PWOw and PWOb vs. Ctrl↑ SIRT1 expression in PWOw and PWOb vs. Ctrl
↑ GR-a expression in GDM vs. Ctrl
↑ Monocyte count was higher in the cord blood PWOb with normal glucose tolerance
↑ Serum leptin PWOb compared to Ctrl and their offspring
[139]
Additional abbreviations: GDF, growth differentiation factor-15; GSR, glutathione reductase; IGFBP2, insulin-like growth factor-binding protein 2; MGO, methylglyoxal; MVP, mean platelet volume; NLR, neutrophil to lymphocyte ratio; PAF, platelet-activating factor; PCT, procalcitonin; PLR, platelet to lymphocyte ratio; PON, paraoxonase; PPARα, peroxisome proliferator-activated receptors; SIRT1, silent mating type information regulation 2 homolog; UCP2, uncoupling protein 2; VAP-1, vascular adhesion protein-1.

5.4. Risk of Preterm Delivery

OS and inflammation induce impairment of placental permeability, producing a hypoxic placenta, which usually leads to activation of the maternal systemic inflammatory response [148]. As a consequence, it affects both maternal and placental functions, thereby causing intrauterine growth restriction (IUGR), preterm delivery, gestational diabetes, preeclampsia, and aortic dissection during pregnancy [61,149]. Likewise, there is dysregulation in lipid profiles such as sphingomyelin and phosphatidylcholine, which have been associated with inflammatory biomarkers such as TNF-α, IL-6, and CRP [150].

5.5. Non-Alcoholic Fatty Liver Disease (NAFLD)

NAFLD is regarded as a hepatic manifestation of metabolic syndrome (MS), which is characterized by elevated levels of saturated fatty acids, polyunsaturated fatty acids, and reduced levels of phospholipids in both the serum and HDL, specifically according to their PUFA fraction.
In both animals and humans, studies have established a relationship between adaption to maternal malnutrition (over or undernutrition), environmental factors, maternal stress, pathology genetics, and epigenetic modifications and early-life and later development of components of MS in offspring, including obesity, insulin resistance, impaired glucose tolerance, dyslipidemia, low HDL-C, increased VLDL-TG, and abnormal liver function such as NAFLD [151,152]. Moreover, recent studies on young adults revealed that being born to an obese mother induced perturbations in adipose tissue function and lipid homeostasis, increasing the risk for developing NAFLD in adulthood three-fold [153].
Declining adipose tissue function is a key characteristic in the transition to metabolically unhealthy, hypertrophic obesity. Dysfunctional adipose tissue is characterized by chronic inflammation that may promote injury, increasing oxidative stress, insulin resistance, dysregulated lipolysis, and eventually lipotoxicity combined with ectopic deposition in peripheral organs such as the liver. Therefore, adiposopathy is a key trigger for the onset of obesity-associated hepatic steatosis [154,155].
In this context, exposure to an abnormal intrauterine milieu may be an important risk factor for the development of cardiometabolic diseases in childhood and adulthood. This adaptation to the intrauterine environment has been explained with Barker’s theory, also known as the intrauterine programming effect, where the intrauterine environment alters the metabolism of the fetus, redistributes its blood flows to protect important organs such as the brain, and even adapts to slower growth to decrease its substrate demands. On the other hand, those changes can be permanent in the structure and function of the offspring. According to this theory, the growth and development of the fetus are determined by three factors: first, the mother’s nutritional status; second, placental function; and third, the ability of the fetus to utilize nutrients [156].

6. Lipids and Lipoproteins in Pregnancy

Lipid metabolism during pregnancy becomes relevant since lipid concentrations change according to maternal requirements and fetal growth; subsequently, dysregulation of lipid metabolism is associated with endothelial dysfunction or immunological changes [150], while major alterations are found in the concentration of triglycerides and cholesterol and in the number of LDL and HDL particles [157], hyperlipidemia being a common condition even in normal pregnancy that allows glucose and calories to be utilized by the fetus. Nevertheless, it has been reported that maternal lipid levels during pregnancy are significantly correlated with the lipid profile of children during the first years of life [158]. Additionally, it is well known that lipid dysregulation is an important risk factor associated with the development of preeclampsia and cardiovascular disease in pregnancy.

6.1. Lipoproteins

Lipoproteins are macromolecular complexes composed of hydrophobic lipids such as triglycerides and cholesterol esters on the inside, whereas their surface is formed by amphipathic lipids like phospholipids and free cholesterol. Moreover, there are proteins, known as the apolipoproteins (Apo), providing stability to the surface and conferring part of their own properties [5] (Figure 2). In recent decades, the role that certain lipoproteins play in different chronic-degenerative diseases has sparked interest, especially high-density lipoproteins (HDL).

6.2. HDL

High-density lipoproteins (HDL) are complex and heterogeneous structures constituting a lipid transport mechanism. Different components (lipids and proteins) of HDL are continuously being exchanged. As a result, they modify the composition, charge, and size of these particles. Currently, it has been described that HDL particles can similarly transport other compounds (about 250), such as sphingosine-1-phosphate, paraoxonase-1 (PON1), acute phase proteins (SAA) [159], platelet-activating factor acetylhydrolase (PFA-AH) enzymes and proteins, such as cholesterol ester transporter protein (CETP) and phospholipid transporter protein (PLTP), among many other components [160,161].
In this context, HDL has been attributed to exert some cardioprotective properties, including reverse cholesterol transport and antioxidant, anti-inflammatory, and antiatherogenic activities (Table 5) [162,163,164,165]. Many of these functions are important for a healthy pregnancy and good neonatal outcomes [166,167].
These positive effects are explained by the structure and chemical composition of these particles. However, it has been shown that these lipoproteins can lose or reduce their cardioprotective capacity, giving rise to prooxidant, proinflammatory, and proatherogenic lipoproteins, contributing to the process of atherosclerosis; this phenomena has been termed “dysfunctional HDL” [173,174].
Some study groups proposed the hypothesis that HDL delivers lipids to cells. For instance, Pérez-Mendez et al. demonstrated that HDL delivers cholesterol and sphingomyelin to endothelial cells in vitro [175]. Therefore, the possibility of the regulation of these lipoproteins on cell function after internalization and the delivery of their content is extremely high. Hence, the lipid delivery of HDL to cells becomes of particular importance when cell membranes should be intensively synthesized or re-structured, i.e., during fetal growth. HDL-C plasma levels and composition may change drastically during inflammatory processes.
It has been described that HDL can inhibit the oxidation of other molecules, such as LDL through free radical damage, which results in the generation of oxidized lipids with pro-inflammatory activity [176]. Nonetheless, in certain conditions such as obesity, diabetes, and other cardiovascular diseases, it has been observed that HDL loses its protective properties, becoming dysfunctional HDL [168,169,170], and leads to an increase in inflammatory processes and OS in several conditions, including pregnancy (Figure 3) [166,167].
Given that this is of great relevance in chronic degenerative diseases, the use of bioactive compounds from fruits, vegetables, foods of animal origin, and plants has become an alternative for improving the functionality and chemical composition of HDL. Thus, they are able to regulate the negative effects caused by OS and inflammatory processes.

6.3. HDL Role and Upregulation Contribution in Inflammatory Processes and OS

Pregnant women normally experience physiological changes, involving carbohydrate and lipid metabolism, insulin resistance, inflammation, coagulation, and OS, all of them causing endothelial damage [170]. Despite this unfavorable environment, pregnant women have better vascular function. Likewise, during embryogenesis and fetal development, the levels of apolipoproteins, lipoproteins, and lipids increase significantly. HDL-C levels change during pregnancy: in the first trimester, changes are insignificant, but, in the second trimester, these changes increase and then slightly decrease in the third trimester. In chronic inflammatory processes, the functional activities of HDL are reduced, the formation of new particles decreases, and catabolism increases. Also, structural changes occur at the protein level, such as the replacement of PON1 or Apo A1 molecules by proinflammatory proteins, including ceruloplasmin and SAA [177,178] that converts HDL to HDL-proinflammatory and results in increased chemoattractant activity, oxidation of LDL, and the release of additional proinflammatory molecules [179,180]. In this chronic inflammation, HDL-proinflammatory may accelerate immune responses toward pathogens, due to HDL remodeling. It is well known that immunological changes occurring in pregnancy for improved fetal tolerance lead to an increased susceptibility to infections. In the acute phase response, HDL levels decrease constantly, with an increase in SAA and ceruloplasmin concentration and a respective decrease in PON1 and Apo A1. Consequently, this could be one of the major risk factors during pregnancy (in trimesters of increased inflammatory processes) for the development of diseases or negative conditions.
Furthermore, an important role of HDL in pregnancy has been reported in the reducing of OS levels, both at placental level and in umbilical cord blood, which are mainly associated with PON1 activity [181]. An important factor in pregnancy is the higher activity of lipoprotein-associated phospholipase A2 (LpPla2) (mainly LDL and HDL), which is an enzyme synthesized predominantly by macrophages and associated with inflammatory processes and higher triglyceride levels, as well as in conditions of elevated OS such as GDM concentration of LpPla2, which is highly elevated compared to healthy women. However, this enzyme can be associated with HDL because it improves the antioxidant and anti-inflammatory functions of HDL, thereby reducing OS levels in plasma [182].
Another important complication of pregnancy caused by the increase in OS is preeclampsia, which affects both pregnant women and newborns and presents oxidative alterations in both LDL and HDL, caused by lipoperoxidation and inactivation of PON1, potentially leading to improper placentation [183].
It has been shown that a maternal diet rich in saturated and trans fatty acids causes harmful changes in the bacteria that colonize the intestine of the offspring, and these in turn produce metabolites that can subsequently affect different organs. Organic acids produced by intestinal bacteria may be involved in inflammatory mechanisms and play a key role in changes in the metabolism and develop neonatally or in adulthood. A high-fat diet during perinatal life predisposes greater expression of the NF-κB gene, which is a transcription factor of multiple biological processes, including immune and inflammatory responses and cell growth and survival [184,185].
Several studies have suggested that HDL dysfunction is a common pathological factor that connects the metabolic syndrome to NAFLD and cardiovascular disease development. The composition and structure of HDL particles seem to be characterized by the depletion of polyunsaturated fatty acid phospholipids and enrichment of saturated fatty acid ceramides [151,186]. In this context, preclinical studies have provided mechanistic insights as to how PUFA (especially essential fatty acids, EFA) deficiency promotes hepatic steatosis. EFA can negatively modulate the hepatic de novo lipogenesis machinery toward the negative modulation of the liver X receptor (LXR) of SREBP-1 and/or of the carbohydrate response element binding protein (ChREBP). Also, PUFA can activate the peroxisome proliferator, activated receptor-alpha (PPARα), and may promote fatty acid oxidation [186]. This is strong evidence for the role of PUFAs in modulating hepatic lipid metabolism [151].

6.4. Bioactive Compounds and HDL Functionality

Bioactive compounds have been widely studied as mediators of inflammation and OS in several conditions and diseases. Nevertheless, their mechanism of action remains unclear. Some studies describe that they are an important part of the secretion of inflammatory molecules (cytokines, adipokines, etc.), of the mediation of metabolic pathways, or of the regulation of gene expression at the muscle or adipose tissue level [187]. Currently, the most commonly studied bioactive compounds are folates [188], polyphenolic compounds [189], polyunsaturated fatty acids [190], prebiotics [191], and probiotics [192], along with their derivatives.
Studies have described that part of the functionality of HDL is linked to its chemical composition. This, in turn, depends on the appearance of some diseases causing HDL dysfunction [193], especially chronic degenerative diseases, which may also change HDL size as well as the number of circulating particles. In DM and coronary heart disease patients, it has been reported that, in addition to Apo A concentration modifications, the presence of OX increases glycoxidation and peroxidation of protein and lipid fractions of HDL, respectively [194,195,196,197].
An alternative way to reverse the mentioned effects is via bioactive compounds from functional foods. It is well documented that foods, for example, fruits and vegetables, fish, legumes, cereals, red wine, and elements of the Mediterranean diet, increase the concentration of HDL, TRC, and antioxidant activity at the same time, increasing the activity and/or expression of paraoxonase-1 (PON1), an atheroprotective enzyme that is bound to HDL [198,199,200]. Likewise, it has been reported that foods rich in polyphenols, hydrolysable tannins, and polyunsaturated fatty acids (PUFAs) modify the protein [201] c-HDL, Tg-HDL, and Phos-HDL content of HDL [197,198].
Alternative dietary modifications such as Mediterranean diet [202] seek to enhance HDL functionality via regulation of RCT. On the other hand, olive oil consumption [203] seems to have a similar effect but is attributable to its effect on the size and stability of HDL. For instance, a study based on red yeast rice extract and additional compounds shown to reduce cardiovascular risk in humans, a significant decrease in the lipid profile associated with cardiovascular risk, mainly c-LDL, and an increase in Apo A1 were found in 102 participants. However, there was no significant difference in the levels of c-HDL [204]. In contrast, a pilot study of 167 patients with metabolic syndrome features, bioactive compounds such as docosahexaenoic acid, β-glucans, and anthocyanins were proved as components of fortified functional foods, and a significant decrease in triglycerides and an increase in LDL-C were observed [205]. Moreover, in a study that evaluated the structure and function of HDL in adults with overweight, obesity, and cardiovascular risk, it was observed that there is a relationship between the decrease in inflammation markers such as IL-6 with sphingosine 1 phosphate (SP1) of HDL under a diet based on a Mediterranean diet [206].
Studies by our research group have demonstrated this: in an animal model as well as in women with acute coronary ischemic syndrome (ACS), by using a microencapsulated product enriched in antioxidants and PUFAs, mainly punicic acid, it was observed that this treatment improved the lipid profile, PON1 activity, and endothelial function mediated by HDL. In ACS women, the dysfunctionality of HDL was reverted by regulating the protein and lipid composition of the smallest subclasses (HDL3) [201,207]. Two possible explanations for these results suggest that bioactive compounds present in this microencapsulated product could remodel alterations in HDL under conditions of dyslipidemia, OS, and inflammation. Also, another possible explanation is that HDL acts as a vector for bioactive compounds, enhancing its bioavailability and potentially increasing its health benefits.

7. Conclusions

Health in pregnant women is of great relevance in preventing cardiometabolic diseases in offspring. In this context, the properties of HDL, like removing oxidized lipids, carrying bioactive compounds, inhibiting expression and activity of pro-inflammatory molecules, lipid transport or regulation of lipid metabolism, are important during pregnancy to progress to childbirth without complications, in addition to promoting placentation and healthy development of the fetus. Moreover, processes of lipoperoxidation, glycoxidation, and the release of pro-inflammatory molecules, which may possibly be reverted by the interaction and transport of bioactive compounds that are present in foods, characterize diseases such as preeclampsia, diabetes, and gestational obesity during pregnancy. However, more studies are needed in order to find therapies that regulate metabolic processes to prevent diseases and to ensure a healthy pregnancy.

Author Contributions

A.S.J.-O., E.C.-T., and D.E.-L. wrote the majority of the original draft of the paper; A.S.J.-O., E.C.-T., and D.E.-L. produced Figure 1, Figure 2 and Figure 3; O.J.-G., E.C.-T., A.S.J.-O., and D.E.-L. edited the manuscript and wrote part of Section 2; L.M.-C. and H.A.D.-Z. wrote part of Section 3, and J.Á.-G. and E.C.-S. wrote Section 4; R.M.B.-T., M.L.S.-P., and A.S.J.-O. wrote part of Section 5.1, Section 5.2, Section 5.3, and Section 5.4 and produced Table 2, Table 3 and Table 4; O.R.F.-C. and J.A.-R. wrote part of Section 5 and Section 6; E.C.-T. and D.E.-L. wrote part of Section 6.1 and Section 6.2; L.M.-C., H.A.D.-Z., J.Á.-G., and E.C.-S. wrote part of Section 6.3 and Section 6.4; A.S.J.-O., J.A.-R. and D.E.-L., participated in designing the content and the analysis of the information in the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Generic State Fund of Universidad Autónoma del Estado de Hidalgo, PAO-2023-0492.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kazma, J.M.; van den Anker, J.; Allegaert, K.; Dallmann, A.; Ahmadzia, H.K. Anatomical and Physiological Alterations of Pregnancy. J. Pharmacokinet. Pharmacodyn. 2020, 47, 271–285. [Google Scholar] [CrossRef] [PubMed]
  2. Kesikburun, S.; Güzelküçük, Ü.; Fidan, U.; Demir, Y.; Ergün, A.; Tan, A.K. Musculoskeletal Pain and Symptoms in Pregnancy: A Descriptive Study. Ther. Adv. Musculoskelet. Dis. 2018, 10, 229–234. [Google Scholar] [CrossRef] [PubMed]
  3. Sahu, A.K.; Harsha, M.M.; Rathoor, S. Cardiovascular Diseases in Pregnancy—A Brief Overview. Curr. Cardiol. Rev. 2021, 18, 77–88. [Google Scholar] [CrossRef]
  4. O’Kelly, A.C.; Michos, E.D.; Shufelt, C.L.; Vermunt, J.V.; Minissian, M.B.; Quesada, O.; Smith, G.N.; Rich-Edwards, J.W.; Garovic, V.D.; El Khoudary, S.R.; et al. Pregnancy and Reproductive Risk Factors for Cardiovascular Disease in Women. Circ. Res. 2022, 130, 652–672. [Google Scholar] [CrossRef]
  5. Estrada-Luna, D.; Ortiz-Rodriguez, M.A.; Medina-Briseño, L.; Carreón-Torres, E.; Izquierdo-Vega, J.A.; Sharma, A.; Cancino-Díaz, J.C.; Pérez-Méndez, O.; Belefant-Miller, H.; Betanzos-Cabrera, G. Current Therapies Focused on High-Density Lipoproteins Associated with Cardiovascular Disease. Molecules 2018, 23, 2730. [Google Scholar] [CrossRef]
  6. Stadler, J.T.; van Poppel, M.N.M.; Wadsack, C.; Holzer, M.; Pammer, A.; Simmons, D.; Hill, D.; Desoye, G.; Marsche, G. Obesity Affects Maternal and Neonatal HDL Metabolism and Function. Antioxidants 2023, 12, 199. [Google Scholar] [CrossRef] [PubMed]
  7. Monserrat-Mesquida, M.; Quetglas-Llabrés, M.; Bouzas, C.; Capó, X.; Mateos, D.; Ugarriza, L.; Tur, J.A.; Sureda, A. Peripheral Blood Mononuclear Cells Oxidative Stress and Plasma Inflammatory Biomarkers in Adults with Normal Weight, Overweight and Obesity. Antioxidants 2021, 10, 813. [Google Scholar] [CrossRef] [PubMed]
  8. Yaribeygi, H.; Sathyapalan, T.; Atkin, S.L.; Sahebkar, A. Molecular Mechanisms Linking Oxidative Stress and Diabetes Mellitus. Oxid. Med. Cell. Longev. 2020, 2020, 8609213. [Google Scholar] [CrossRef]
  9. Singh, A.; Kukreti, R.; Saso, L.; Kukreti, S. Mechanistic Insight into Oxidative Stress-Triggered Signaling Pathways and Type 2 Diabetes. Molecules 2022, 27, 950. [Google Scholar] [CrossRef]
  10. Afrose, D.; Chen, H.; Ranashinghe, A.; Liu, C.C.; Henessy, A.; Hansbro, P.M.; McClements, L. The Diagnostic Potential of Oxidative Stress Biomarkers for Preeclampsia: Systematic Review and Meta-Analysis. Biol. Sex Differ. 2022, 13, 26. [Google Scholar] [CrossRef]
  11. Sorrell, T.R. Mexican Traditional Medicine: Application of a Traditional and Complementary Medicine System to Improve Opioid Use Treatment in Latinos. J. Holist. Nurs. 2020, 38, 384–399. [Google Scholar] [CrossRef] [PubMed]
  12. Lucía, C.P.A.; Jacqueline, B.R.; Alberto, B.R.L.; David, B.A.; Beatriz, R.A. Actualized Inventory of Medicinal Plants Used in Traditional Medicine in Oaxaca, Mexico. J. Ethnobiol. Ethnomed. 2021, 17, 7. [Google Scholar] [CrossRef] [PubMed]
  13. Harris, P.E.; Cooper, K.L.; Relton, C.; Thomas, K.J. Prevalence of Complementary and Alternative Medicine (CAM) Use by the General Population: A Systematic Review and Update. Int. J. Clin. Pract. 2012, 66, 924–939. [Google Scholar] [CrossRef]
  14. Shewamene, Z.; Dune, T.; Smith, C.A. The Use of Traditional Medicine in Maternity Care among African Women in Africa and the Diaspora: A Systematic Review. BMC Complement. Altern. Med. 2017, 17, 382. [Google Scholar] [CrossRef] [PubMed]
  15. Okoth, K.; Chandan, J.S.; Marshall, T.; Thangaratinam, S.; Thomas, G.N.; Nirantharakumar, K.; Adderley, N.J. Association between the Reproductive Health of Young Women and Cardiovascular Disease in Later Life: Umbrella Review. BMJ 2020, 371, m3502. [Google Scholar] [CrossRef] [PubMed]
  16. Cabada-Aguirre, P.; López López, A.M.; Mendoza, K.C.O.; Garay Buenrostro, K.D.; Luna-Vital, D.A.; Mahady, G.B. Mexican Traditional Medicines for Women’s Reproductive Health. Sci. Rep. 2023, 13, 2807. [Google Scholar] [CrossRef]
  17. Yılmaz, İ.; Enver Dolar, M.; Özpınar, H. Effect of Administering Kefir on the Changes in Fecal Microbiota and Symptoms of Inflammatory Bowel Disease: A Randomized Controlled Trial. Turkish J. Gastroenterol. 2019, 30, 242–253. [Google Scholar] [CrossRef]
  18. Vaou, N.; Stavropoulou, E.; Voidarou, C.; Tsakris, Z.; Rozos, G.; Tsigalou, C.; Bezirtzoglou, E. Interactions between Medical Plant-Derived Bioactive Compounds: Focus on Antimicrobial Combination Effects. Antibiotics 2022, 11, 1014. [Google Scholar] [CrossRef]
  19. Liang, B.; Zhu, Y.C.; Lu, J.; Gu, N. Effects of Traditional Chinese Medication-Based Bioactive Compounds on Cellular and Molecular Mechanisms of Oxidative Stress. Oxid. Med. Cell. Longev. 2021, 2021, 3617498. [Google Scholar] [CrossRef]
  20. Chernukha, I.; Kotenkova, E.; Derbeneva, S.; Khvostov, D. Bioactive Compounds of Porcine Hearts and Aortas May Improve Cardiovascular Disorders in Humans. Int. J. Environ. Res. Public Health 2021, 18, 7330. [Google Scholar] [CrossRef]
  21. Li, H.Y.; Gan, R.Y.; Shang, A.; Mao, Q.Q.; Sun, Q.C.; Wu, D.T.; Geng, F.; He, X.Q.; Li, H. Bin Plant-Based Foods and Their Bioactive Compounds on Fatty Liver Disease: Effects, Mechanisms, and Clinical Application. Oxid. Med. Cell. Longev. 2021, 2021, 6621644. [Google Scholar] [CrossRef] [PubMed]
  22. Bocanegra, A.; Macho-González, A.; Alba Garcimartín, J.B.; Sánchez-Muniz, F.J. Whole Alga, Algal Extracts, and Compounds as Ingredients of Functional Foods: Composition and Action Mechanism Relationships in the Prevention and Treatment of Type-2 Diabetes Mellitus. Int. J. Mol. Sci. 2021, 22, 3816. [Google Scholar] [CrossRef] [PubMed]
  23. Mohd Sairazi, N.S.; Sirajudeen, K.N.S. Natural Products and Their Bioactive Compounds: Neuroprotective Potentials against Neurodegenerative Diseases. Evid.-Based Complement. Altern. Med. 2020, 2020, 6565396. [Google Scholar] [CrossRef] [PubMed]
  24. Ghosh; Rajarshi; Bryant; Daniel, L.; Farone, A.L. Polysaccharides as Immunomodulators : Current Research Status and Future Directions. Molecules 2020, 25, 5854. [Google Scholar] [CrossRef] [PubMed]
  25. Ge, P.Y.; Qi, Y.Y.; Qu, S.Y.; Zhao, X.; Ni, S.J.; Yao, Z.Y.; Guo, R.; Yang, N.Y.; Zhang, Q.C.; Zhu, H.X. Potential Mechanism of s. Baicalensis on Lipid Metabolism Explored via Network Pharmacology and Untargeted Lipidomics. Drug Des. Devel. Ther. 2021, 15, 1915–1930. [Google Scholar] [CrossRef] [PubMed]
  26. Davis, D.W.; Crew, J.; Planinic, P.; Alexander, J.M.; Basu, A. Associations of Dietary Bioactive Compounds with Maternal Adiposity and Inflammation in Gestational Diabetes: An Update on Observational and Clinical Studies. Int. J. Environ. Res. Public Health 2020, 17, 7528. [Google Scholar] [CrossRef]
  27. Ajanaku, C.O.; Ademosun, O.T.; Atohengbe, P.O.; Ajayi, S.O.; Obafemi, Y.D.; Owolabi, O.A.; Akinduti, P.A.; Ajanaku, K.O. Functional Bioactive Compounds in Ginger, Turmeric, and Garlic. Front. Nutr. 2022, 9, 1012023. [Google Scholar] [CrossRef]
  28. Nacka-Aleksić, M.; Pirković, A.; Vilotić, A.; Bojić-Trbojević, Ž.; Jovanović Krivokuća, M.; Giampieri, F.; Battino, M.; Dekanski, D. The Role of Dietary Polyphenols in Pregnancy and Pregnancy-Related Disorders. Nutrients 2022, 14, 5246. [Google Scholar] [CrossRef]
  29. Fogacci, S.; Fogacci, F.; Cicero, A.F.G. Nutraceuticals and Hypertensive Disorders in Pregnancy: The Available Clinical Evidence. Nutrients 2020, 12, 378. [Google Scholar] [CrossRef]
  30. Zhang, H.; Su, S.; Yu, X.; Li, Y. Dietary Epigallocatechin 3-Gallate Supplement Improves Maternal and Neonatal Treatment Outcome of Gestational Diabetes Mellitus: A Double-Blind Randomised Controlled Trial. J. Hum. Nutr. Diet. 2017, 30, 753–758. [Google Scholar] [CrossRef]
  31. Yuan, L.J.; Qin, Y.; Wang, L.; Zeng, Y.; Chang, H.; Wang, J.; Wang, B.; Wan, J.; Chen, S.H.; Zhang, Q.Y.; et al. Capsaicin-Containing Chili Improved Postprandial Hyperglycemia, Hyperinsulinemia, and Fasting Lipid Disorders in Women with Gestational Diabetes Mellitus and Lowered the Incidence of Large-for-Gestational-Age Newborns. Clin. Nutr. 2016, 35, 388–393. [Google Scholar] [CrossRef]
  32. Jaworsky, K.; DeVillez, P.; Basu, A. The Role of Phytochemicals and Plant-Based Diets in Gestational Diabetes: Evidence from Clinical Trials. Int. J. Environ. Res. Public Health 2023, 20, 4188. [Google Scholar] [CrossRef]
  33. Majed, B.H.; Khalil, R.A. Molecular Mechanisms Regulating the Vascular Prostacyclin Pathways and Their Adaptation during Pregnancy and in the Newborn. Pharmacol. Rev. 2012, 64, 540–582. [Google Scholar] [CrossRef] [PubMed]
  34. Salinas-Roca, B.; Rubió-Piqué, L.; Montull-López, A. Polyphenol Intake in Pregnant Women on Gestational Diabetes Risk and Neurodevelopmental Disorders in Offspring: A Systematic Review. Nutrients 2022, 14, 3753. [Google Scholar] [CrossRef]
  35. Parham, S.; Kharazi, A.Z.; Bakhsheshi-Rad, H.R.; Nur, H.; Ismail, A.F.; Sharif, S.; Ramakrishna, S.; Berto, F. Antioxidant, Antimicrobial and Antiviral Properties of Herbal Materials. Antioxidants 2020, 9, 1309. [Google Scholar] [CrossRef] [PubMed]
  36. Ozkur, M.; Benlier, N.; Takan, I.; Vasileiou, C.; Georgakilas, A.G.; Pavlopoulou, A.; Cetin, Z.; Saygili, E.I. Ginger for Healthy Ageing: A Systematic Review on Current Evidence of Its Antioxidant, Anti-Inflammatory, and Anticancer Properties. Oxid. Med. Cell. Longev. 2022, 2022, 4748447. [Google Scholar] [CrossRef] [PubMed]
  37. Jeong, Y.Y.; Ryu, J.H.; Shin, J.H.; Kang, M.J.; Kang, J.R.; Han, J.; Kang, D. Comparison of Anti-Oxidant and Anti-Inflammatory Effects between Fresh and Aged Black Garlic Extracts. Molecules 2016, 21, 430. [Google Scholar] [CrossRef]
  38. Sah, A.; Naseef, P.P.; Kuruniyan, M.S.; Jain, G.K.; Zakir, F.; Aggarwal, G. A Comprehensive Study of Therapeutic Applications of Chamomile. Pharmaceuticals 2022, 15, 1284. [Google Scholar] [CrossRef]
  39. Tafrihi, M.; Imran, M.; Tufail, T.; Gondal, T.A.; Caruso, G.; Sharma, S.; Sharma, R.; Atanassova, M.; Atanassov, L.; Fokou, P.V.T.; et al. The Wonderful Activities of the Genus Mentha: Not Only Antioxidant Properties. Molecules 2021, 26, 1118. [Google Scholar] [CrossRef]
  40. Sánchez, M.; González-Burgos, E.; Iglesias, I.; Lozano, R.; Gómez-Serranillos, M.P. The Pharmacological Activity of Camellia sinensis (L.) Kuntze on Metabolic and Endocrine Disorders: A Systematic Review. Biomolecules 2020, 10, 603. [Google Scholar] [CrossRef]
  41. Arikan, S.; Konukog, D.; Tülay, A.; Davas, I. Lipid Peroxidation and Antioxidant Status in Maternal and Cord Blood. Gynecol. Obs. Investig. 2001, 51, 145–149. [Google Scholar] [CrossRef]
  42. Fialová, L.; Malbohan, I.; Kalousová, M.; Soukupová, J.; Krofta, L.; Štípek, S.; Zima, T. Oxidative Stress and Inflammation in Pregnancy. Scand. J. Clin. Lab. Investig. 2006, 66, 121–128. [Google Scholar] [CrossRef]
  43. Guerby, P.; Tasta, O.; Swiader, A.; Pont, F.; Bujold, E.; Parant, O.; Vayssiere, C.; Salvayre, R.; Negre-Salvayre, A. Role of Oxidative Stress in the Dysfunction of the Placental Endothelial Nitric Oxide Synthase in Preeclampsia. Redox Biol. 2021, 40, 101861. [Google Scholar] [CrossRef] [PubMed]
  44. Sultana, Z.; Maiti, K.; Aitken, J.; Morris, J.; Dedman, L.; Smith, R. Oxidative Stress, Placental Ageing-Related Pathologies and Adverse Pregnancy Outcomes. Am. J. Reprod. Immunol. 2017, 77, e12653. [Google Scholar] [CrossRef]
  45. Myatt, L. Review: Reactive Oxygen and Nitrogen Species and Functional Adaptation of the Placenta. Placenta 2010, 31, S66–S69. [Google Scholar] [CrossRef] [PubMed]
  46. Hussain, T.; Murtaza, G.; Metwally, E.; Kalhoro, D.H.; Kalhoro, M.S.; Rahu, B.A.; Sahito, R.G.A.; Yin, Y.; Yang, H.; Chughtai, M.I.; et al. The Role of Oxidative Stress and Antioxidant Balance in Pregnancy. Mediat. Inflamm. 2021, 2021, 9962860. [Google Scholar] [CrossRef]
  47. Aouache, R.; Biquard, L.; Vaiman, D.; Miralles, F. Oxidative Stress in Preeclampsia and Placental Diseases. Int. J. Mol. Sci. 2018, 19, 1496. [Google Scholar] [CrossRef]
  48. Parrettini, S.; Caroli, A.; Torlone, E. Nutrition and Metabolic Adaptations in Physiological and Complicated Pregnancy: Focus on Obesity and Gestational Diabetes. Front. Endocrinol. 2020, 11, 1–19. [Google Scholar] [CrossRef] [PubMed]
  49. Lu, J.; Wang, Z.; Cao, J.; Chen, Y.; Dong, Y. A Novel and Compact Review on the Role of Oxidative Stress in Female Reproduction. Reprod. Biol. Endocrinol. 2018, 16, 80. [Google Scholar] [CrossRef]
  50. Mor, G.; Aldo, P.; Alvero, A.B. The Unique Immunological and Microbial Aspects of Pregnancy. Nat. Rev. Immunol. 2017, 17, 469–482. [Google Scholar] [CrossRef]
  51. Denizli, M.; Capitano, M.L.; Kua, K.L. Maternal Obesity and the Impact of Associated Early-Life Inflammation on Long-Term Health of Offspring. Front. Cell. Infect. Microbiol. 2022, 12, 1–17. [Google Scholar] [CrossRef] [PubMed]
  52. Puttabyatappa, M.; Banker, M.; Zeng, L.; Goodrich, J.M.; Domino, S.E.; Dolinoy, D.C.; Meeker, J.D.; Pennathur, S.; Song, P.X.K.; Padmanabhan, V. Maternal Exposure to Environmental Disruptors and Sexually Dimorphic Changes in Maternal and Neonatal Oxidative Stress. J. Clin. Endocrinol. Metab. 2020, 105, 492–505. [Google Scholar] [CrossRef] [PubMed]
  53. Gümüş, P.; Emingil, G.; Öztürk, V.Ö.; Belibasakis, G.N.; Bostanci, N. Oxidative Stress Markers in Saliva and Periodontal Disease Status: Modulation during Pregnancy and Postpartum. BMC Infect. Dis. 2015, 15, 261. [Google Scholar] [CrossRef] [PubMed]
  54. Quintanilla-García, C.V.; Uribarri, J.; Fajardo-Araujo, M.E.; Barrientos-Romero, J.J.; Romero-Gutiérrez, G.; Reynaga-Ornelas, M.G.; Garay-Sevilla, M.E. Changes in Circulating Levels of Carboxymethyllysine, Soluble Receptor for Advanced Glycation End Products (SRAGE), and Inflammation Markers in Women during Normal Pregnancy. J. Matern. Neonatal Med. 2019, 32, 4102–4107. [Google Scholar] [CrossRef]
  55. Lean, S.C.; Jones, R.L.; Roberts, S.A.; Heazell, A.E.P. A Prospective Cohort Study Providing Insights for Markers of Adverse Pregnancy Outcome in Older Mothers. BMC Pregnancy Childbirth 2021, 21, 706. [Google Scholar] [CrossRef]
  56. Cave, C.; Hanson, C.; Schumacher, M.; Lyden, E.; Furtado, J.; Obaro, S.; Delair, S.; Kocmich, N.; Rezac, A.; Izevbigie, N.I.; et al. A Comparison of Vitamin E Status and Associated Pregnancy Outcomes in Maternal–Infant Dyads between a Nigerian and a United States Population. Nutrients 2018, 10, 1300. [Google Scholar] [CrossRef]
  57. Merlo, A.C.; Rosa, G.M.; Porto, I. Pregnancy-Related Acute Myocardial Infarction: A Review of the Recent Literature. Clin. Res. Cardiol. 2022, 111, 723–731. [Google Scholar] [CrossRef] [PubMed]
  58. Mao, Y.; Hu, W.; Xia, B.; Liu, L.; Han, X.; Liu, Q. Association between Gestational Diabetes Mellitus and the Risks of Type-Specific Cardiovascular Diseases. Front. Public Health 2022, 10, 1–10. [Google Scholar] [CrossRef]
  59. Grønberg, M.; Flatmark, T. Inhibition of the H+-ATPase in Bovine Adrenal Chromaffin Granule Ghosts by Diethylstilbestrol Evidence for a Tight Coupling between ATP Hydrolysis and Proton Translocation. FEBS Lett. 1988, 229, 40–44. [Google Scholar] [CrossRef]
  60. Zvolsky, P.; Dvorakova, M.; Grof, P. Genealogic Study of Patients with Admission Diagnosis of Functional Psychosis. Arch. Psychiatr. Nervenkr. 1982, 232, 157–166. [Google Scholar] [CrossRef]
  61. Kalagiri, R.R.; Carder, T.; Choudhury, S.; Vora, N.; Ballard, A.R.; Govande, V.; Drever, N.; Beeram, M.R.; Uddin, M.N. Inflammation in Complicated Pregnancy and Its Outcome. Am. J. Perinatol. 2016, 33, 1337–1356. [Google Scholar] [CrossRef] [PubMed]
  62. Catalano, P.M.; Shankar, K. Obesity and Pregnancy: Mechanisms of Short Term and Long Term Adverse Consequences for Mother and Child. BMJ 2017, 356, j1. [Google Scholar] [CrossRef]
  63. Pantham, P.; Aye, I.L.M.H.; Powella, T.L. Inflammation in Maternal Obesity and Gestational Diabetes Mellitus. Placenta 2015, 176, 139–148. [Google Scholar] [CrossRef] [PubMed]
  64. Humberg, A.; Fortmann, I.; Siller, B.; Kopp, M.V.; Herting, E.; Göpel, W.; Härtel, C.; Neonatal, G. Preterm Birth and Sustained Inflammation: Consequences for the Neonate Network, German Center for Lung Research and Priming Immunity at the Beginning of Life (PRIMAL) Consortium. Semin. Immunopathol. 2020, 42, 451–468. [Google Scholar] [CrossRef] [PubMed]
  65. Musa, E.; Salazar-Petres, E.; Arowolo, A.; Levitt, N.; Matjila, M.; Sferruzzi-Perri, A.N. Obesity and Gestational Diabetes Independently and Collectively Induce Specific Effects on Placental Structure, Inflammation and Endocrine Function in a Cohort of South African Women. J. Physiol. 2023, 7, 1287–1306. [Google Scholar] [CrossRef] [PubMed]
  66. Green, E.S.; Arck, P.C. Pathogenesis of Preterm Birth: Bidirectional Inflammation in Mother and Fetus. Semin. Immunopathol. 2020, 42, 413–429. [Google Scholar] [CrossRef] [PubMed]
  67. Alston, M.C.; Redman, L.M.; Sones, J.L. An Overview of Obesity, Cholesterol, and Systemic Inflammation in Preeclampsia. Nutrients 2022, 14, 2087. [Google Scholar] [CrossRef]
  68. Schneider, D.; Hernández, C.; Farías, M.; Uauy, R.; Krause, B.J.; Casanello, P. Oxidative Stress as Common Trait of Endothelial Dysfunction in Chorionic Arteries from Fetuses with IUGR and LGA. Placenta 2015, 36, 552–558. [Google Scholar] [CrossRef]
  69. Kamath, U.; Rao, G.; Raghothama, C.; Rai, L.; Rao, P. Erythrocyte Indicators of Oxidative Stress in Gestational Diabetes. Acta Paediatr. Int. J. Paediatr. 1998, 87, 676–679. [Google Scholar] [CrossRef]
  70. Jarmuzek, P.; Wielgos, M.; Bomba-Opon, D.A. Placental Pathologic Changes in Gestational Diabetes Mellitus. Neuroendocrinol. Lett. 2015, 36, 101–105. [Google Scholar]
  71. van den Bosch, W.J.H.M. The Practice Guideline “Problematic Alcohol Consumption” (Second Revision) from the Dutch College of General Practitioners. Ned. Tijdschr. Geneeskd. 2006, 150, 2523–2524. [Google Scholar]
  72. Alexander, B.T.; Dasinger, J.H.; Intapad, S. Fetal Programming and Cardiovascular Pathology. Compr. Physiol. 2017, 176, 139–148. [Google Scholar]
  73. Phipps, E.A.; Thadhani, R.; Benzing, T.; Karumanchi, S.A. Pre-Eclampsia: Pathogenesis, Novel Diagnostics and Therapies. Nat. Rev. Nephrol. 2019, 15, 275–289. [Google Scholar] [CrossRef]
  74. MacDonald, T.M.; Walker, S.P.; Hannan, N.J.; Tong, S.; Kaitu’u-Lino, T.J. Clinical Tools and Biomarkers to Predict Preeclampsia. eBioMedicine 2022, 75, 103780. [Google Scholar] [CrossRef]
  75. Hutcheon, J.A.; Lisonkova, S.; Joseph, K.S. Epidemiology of Pre-Eclampsia and the Other Hypertensive Disorders of Pregnancy. Best Pract. Res. Clin. Obstet. Gynaecol. 2011, 25, 391–403. [Google Scholar] [CrossRef] [PubMed]
  76. Offer, E.R.E.Z.; Romero, R.; Eunjung, J.U.N.G.; Chaemsaithong, P.; Bosco, M.; Suksai, M.; Gotsch, F. Preeclampsia/Eclampsia: The Conceptual Evolution of a Syndrome. Am. J. Obs. Gynecol. 2022, 226, 1–42. [Google Scholar] [CrossRef]
  77. Bartsch, E.; Medcalf, K.E.; Park, A.L.; Ray, J.G.; Al-Rubaie, Z.T.A.; Askie, L.M.; Berger, H.; Blake, J.; Graves, L.; Kingdom, J.C.; et al. Clinical Risk Factors for Pre-Eclampsia Determined in Early Pregnancy: Systematic Review and Meta-Analysis of Large Cohort Studies. BMJ 2016, 353, i1753. [Google Scholar] [CrossRef] [PubMed]
  78. Southcombe, J.H.; Redman, C.W.G.; Sargent, I.L.; Granne, I. Interleukin-1 Family Cytokines and Their Regulatory Proteins in Normal Pregnancy and Pre-Eclampsia. Clin. Exp. Immunol. 2015, 181, 480–490. [Google Scholar] [CrossRef]
  79. Wang, Y.; Li, B.; Zhao, Y. Inflammation in Preeclampsia: Genetic Biomarkers, Mechanisms, and Therapeutic Strategies. Front. Immunol. 2022, 13, 1–14. [Google Scholar] [CrossRef] [PubMed]
  80. Gil-Acevedo, L.; Ceballos, G.; Torres-Ramos, Y. Foetal Lipoprotein Oxidation and Preeclampsia. Lipids Health Dis. 2022, 21, 51. [Google Scholar] [CrossRef] [PubMed]
  81. Galbiati, S.; Inversetti, A.; Causarano, V.; Stenirri, S.; Soriani, N.; Ambrosi, A.; Valsecchi, L.; Candiani, M.; Cremonesi, L.; Ferrari, M.; et al. HIF1A and MIF as Potential Predictive MRNA Biomarkers of Pre-Eclampsia: A Longitudinal Prospective Study in High Risk Population. Clin. Chem. Lab. Med. 2015, 53, 1339–1347. [Google Scholar] [CrossRef] [PubMed]
  82. Subha, M.; Pal, P.; Pal, G.K.; Habeebullah, S.; Adithan, C.; Sridhar, M.G. Decreased Baroreflex Sensitivity Is Linked to Sympathovagal Imbalance, Low-Grade Inflammation, and Oxidative Stress in Pregnancy-Induced Hypertension. Clin. Exp. Hypertens. 2016, 38, 666–672. [Google Scholar] [CrossRef] [PubMed]
  83. Ferguson, K.K.; Meeker, J.D.; McElrath, T.F.; Mukherjee, B.; Cantonwine, D.E. Repeated Measures of Inflammation and Oxidative Stress Biomarkers in Preeclamptic and Normotensive Pregnancies. Am. J. Obstet. Gynecol. 2017, 216, 527.e1–527.e9. [Google Scholar] [CrossRef]
  84. Karthiga, K.; Pal, G.K.; Dasari, P.; Nanda, N.; Velkumary, S.; Chinnakali, P. Attenuation of Baroreflex Sensitivity and Heart Rate Variability Is Linked to Reduced Levels of Nitric Oxide in Pregnant Women Having Risks of Developing Gestational Hypertension. Clin. Exp. Hypertens. 2021, 43, 356–362. [Google Scholar] [CrossRef] [PubMed]
  85. Talab, A.Y.; Hamza, H.A.; Mostafa, T.M. Antepartum and Postpartum Changes in Adipokines, Endothelial Dysfunction, Inflammatory Markers and Other Biochemical Parameters in Preeclamptic Women: A Prospective Observational Cohort Study. J. Appl. Biomed. 2021, 19, 62–72. [Google Scholar] [CrossRef]
  86. Molvarec, A.; Szarka, A.; Walentin, S.; Beko, G.; Karádi, I.; Prohászka, Z.; Rigó, J. Serum Heat Shock Protein 70 Levels in Relation to Circulating Cytokines, Chemokines, Adhesion Molecules and Angiogenic Factors in Women with Preeclampsia. Clin. Chim. Acta 2011, 412, 1957–1962. [Google Scholar] [CrossRef]
  87. Catarino, C.; Santos-Silva, A.; Belo, L.; Rocha-Pereira, P.; Rocha, S.; Patrício, B.; Quintanilha, A.; Rebelo, I. Inflammatory Disturbances in Preeclampsia: Relationship between Maternal and Umbilical Cord Blood. J. Pregnancy 2012, 2012, 684384. [Google Scholar] [CrossRef]
  88. Bernardi, F.C.B.; Felisberto, F.; Vuolo, F.; Petronilho, F.; Souza, D.R.; Luciano, T.F.; De Souza, C.T.; Ritter, C.; Dal-Pizzol, F. Oxidative Damage, Inflammation, and Toll-like Receptor 4 Pathway Are Increased in Preeclamptic Patients: A Case-Control Study. Oxid. Med. Cell. Longev. 2012, 2012, 636419. [Google Scholar] [CrossRef]
  89. Ouyang, Y.Q.; Li, S.J.; Zhang, Q.; Cai, H.B.; Chen, H.P. Interactions between Inflammatory and Oxidative Stress in Preeclampsia. Hypertens. Pregnancy 2009, 28, 56–62. [Google Scholar] [CrossRef]
  90. Molvarec, A.; Rigó, J.; Lázár, L.; Balogh, K.; Makó, V.; Cervenak, L.; Mézes, M.; Prohászka, Z. Increased Serum Heat-Shock Protein 70 Levels Reflect Systemic Inflammation, Oxidative Stress and Hepatocellular Injury in Preeclampsia. Cell Stress Chaperones 2009, 14, 151–159. [Google Scholar] [CrossRef]
  91. Silva, D.M.C.E.; Marreiro, D.D.N.; Moita Neto, J.M.; Brito, J.A.; Neta, E.A.D.S.; Matias, J.P.; Sampaio, F.A.; Nogueira, N.D.N. Oxidative Stress and Immunological Alteration in Women with Preeclampsia. Hypertens. Pregnancy 2013, 32, 304–311. [Google Scholar] [CrossRef] [PubMed]
  92. Ouyang, Y.Q.; Li, S.J.; Zhang, Q.; Xiang, W.P.; Shen, H.L.; Chen, H.P.; Chen, H.; Chen, H.Z. Plasma SFlt-1-to-PlGF Ratio Is Correlated with Inflammatory but Not with Oxidative Stress in Chinese Preeclamptic Women. Arch. Gynecol. Obstet. 2009, 280, 91–97. [Google Scholar] [CrossRef]
  93. Wang, C.N.; Chang, S.D.; Peng, H.H.; Lee, Y.S.; Chang, Y.L.; Cheng, P.J.; Chao, A.S.; Wang, T.H.; Wang, H.S. Change in Amniotic Fluid Levels of Multiple Anti-Angiogenic Proteins before Development of Preeclampsia and Intrauterine Growth Restriction. J. Clin. Endocrinol. Metab. 2010, 95, 1431–1441. [Google Scholar] [CrossRef] [PubMed]
  94. Ekun, O.A.; Ogidi, N.O.; Lawal, R.A.; Ogunmuyiwa, O.A.; Umewune, M.C.; Adefolaju, F.O.; Oshundun, M.F.; Oremosu, A.I. Interrelationship between Markers of Oxidative Stress, Inflammation and Hematological Parameters Among Preeclamptic Nigerian Women. Med. Sci. Monit. Basic Res. 2018, 24, 225–231. [Google Scholar] [CrossRef]
  95. Stenczer, B.; Rigó, J.; Prohászka, Z.; Derzsy, Z.; Lázár, L.; Makó, V.; Cervenak, L.; Balogh, K.; Mézes, M.; Karádi, I.; et al. Plasma Osteopontin Concentrations in Preeclampsia—Is There an Association with Endothelial Injury? Clin. Chem. Lab. Med. 2010, 48, 181–187. [Google Scholar] [CrossRef] [PubMed]
  96. Wang, R.; Chen, L.; Wang, X.; Liu, Y. Association between Serum Beta-Human Chorionic Gonadotropin and Inflammation, Oxidative Stress in Pregnancy-Induced Hypertension. Microvasc. Res. 2021, 135, 104130. [Google Scholar] [CrossRef]
  97. Barden, A.; Beilin, L.J.; Ritchie, J.; Croft, K.D.; Walters, B.N.; Michael, C.A. Plasma and Urinary 8-Iso-Prostane as an Indicator of Lipid Peroxidation in Pre-Eclampsia and Normal Pregnancy. Clin. Sci. 1996, 91, 711–718. [Google Scholar] [CrossRef]
  98. Ishihara, O.; Hayashi, M.; Osawa, H.; Kobayashi, K.; Takeda, S.; Vessby, B.; Basu, S. Isoprostanes, Prostaglandins and Tocopherols in Pre-Eclamsia, Normal Pregnancy and Non-Pregnancy. Free Radic. Res. 2004, 38, 913–918. [Google Scholar] [CrossRef]
  99. de Sousa Rocha, V.; Della Rosa, F.B.; Ruano, R.; Zugaib, M.; Colli, C. Association between Magnesium Status, Oxidative Stress and Inflammation in Preeclampsia: A Case-Control Study. Clin. Nutr. 2015, 34, 1166–1171. [Google Scholar] [CrossRef]
  100. Ferreira, R.C.; Fragoso, M.B.T.; dos Santos Tenório, M.C.; da Paz Martins, A.S.; Borbely, A.U.; Moura, F.A.; Goulart, M.O.F.; de Oliveira, A.C.M. Biomarkers of Placental Redox Imbalance in Pregnancies with Preeclampsia and Consequent Perinatal Outcomes. Arch. Biochem. Biophys. 2020, 691, 108464. [Google Scholar] [CrossRef]
  101. Mannaerts, D.; Faes, E.; Cos, P.; Briedé, J.J.; Gyselaers, W.; Cornette, J.; Gorbanev, Y.; Bogaerts, A.; Spaanderman, M.; Van Craenenbroeck, E.; et al. Oxidative Stress in Healthy Pregnancy and Preeclampsia Is Linked to Chronic Inflammation, Iron Status and Vascular Function. PLoS ONE 2018, 13, e0202919. [Google Scholar] [CrossRef]
  102. García-Gómez, E.; Bobadilla-Bravo, M.; Díaz-Díaz, E.; Vázquez-Martínez, E.R.; Nava-Salazar, S.; Torres-Ramos, Y.; García-Romero, C.S.; Camacho-Arroyo, I.; Cerbón, M. High Plasmatic Levels of Advanced Glycation End Products Are Associated with Metabolic Alterations and Insulin Resistance in Preeclamptic Women. Curr. Mol. Med. 2020, 20, 751–759. [Google Scholar] [CrossRef]
  103. Gandley, R.E.; Ph, D.; Rohland, J.; Zhou, Y.; Shibata, E.; Gail, F.; Rajakumar, A.; Ph, D.; Kagan, V.E.; Ph, D.; et al. Increased Myeloperoxidase in the Placenta and circulation of women with preeclampsia. Hypertension 2008, 52, 387–393. [Google Scholar] [CrossRef]
  104. Bainbridge, S.A.; Deng, J.S.; Roberts, J.M. Increased Xanthine Oxidase in the Skin of Preeclamptic Women. Reprod. Sci. 2009, 16, 468–478. [Google Scholar] [CrossRef] [PubMed]
  105. Rees, A.; Richards, O.; Chambers, M.; Jenkins, B.J.; Cronin, J.G.; Thornton, C.A. Immunometabolic Adaptation and Immune Plasticity in Pregnancy and the Bi-Directional Effects of Obesity. Clin. Exp. Immunol. 2022, 208, 132–146. [Google Scholar] [CrossRef] [PubMed]
  106. O’Neill, L.A.J.; Rigel, J.; Kishton, J.R. A guide to immunometabolism for immunologists. Nat. Rev. Immunol. 2017, 176, 139–148. [Google Scholar] [CrossRef]
  107. Loftus, R.M.; Finlay, D.K. Immunometabolism: Cellular Metabolism Turns Immune Regulator. J. Biol. Chem. 2016, 291, 1–10. [Google Scholar] [CrossRef]
  108. Joo, E.H.; Kim, Y.R.; Kim, N.; Jung, J.E.; Han, S.H.; Cho, H.Y. Effect of Endogenic and Exogenic Oxidative Stress Triggers on Adverse Pregnancy Outcomes: Preeclampsia, Fetal Growth Restriction, Gestational Diabetes Mellitus and Preterm Birth. Int. J. Mol. Sci. 2021, 22, 10122. [Google Scholar] [CrossRef]
  109. Kasture, V.; Sahay, A.; Joshi, S. Cell Death Mechanisms and Their Roles in Pregnancy Related Disorders, 1st ed.; Elsevier Inc.: Amsterdam, The Netherlands, 2021; Volume 126, ISBN 9780323853170. [Google Scholar]
  110. Langley-Evans, S.C.; Pearce, J.; Ellis, S. Overweight, Obesity and Excessive Weight Gain in Pregnancy as Risk Factors for Adverse Pregnancy Outcomes: A Narrative Review. J. Hum. Nutr. Diet. 2022, 35, 250–264. [Google Scholar] [CrossRef] [PubMed]
  111. Wawer, A.A.; Hodyl, N.A.; Fairweather-Tait, S.; Froessler, B. Are Pregnant Women Who Are Living with Overweight or Obesity at Greater Risk of Developing Iron Deficiency/Anaemia? Nutrients 2021, 13, 1572. [Google Scholar] [CrossRef]
  112. Álvarez, D.; Muñoz, Y.; Ortiz, M.; Maliqueo, M.; Chouinard-Watkins, R.; Valenzuela, R. Impact of Maternal Obesity on the Metabolism and Bioavailability of Polyunsaturated Fatty Acids during Pregnancy and Breastfeeding. Nutrients 2021, 13, 19. [Google Scholar] [CrossRef] [PubMed]
  113. Scifres, C.M.; Catov, J.M. The Impact of Maternal Obesity and Gestational Weight Gain on Early and Mid-Pregnancy Lipid Profiles. Physiol. Behav. 2017, 176, 139–148. [Google Scholar] [CrossRef]
  114. Maguire, R.L.; House, J.S.; Lloyd, D.T.; Skinner, H.G.; Allen, T.K.; Raffi, A.M.; Skaar, D.A.; Park, S.S.; McCullough, L.E.; Kollins, S.H.; et al. Associations between Maternal Obesity, Gestational Cytokine Levels, and Child Obesity in the NEST Cohort. Pediatr. Obes. 2021, 176, 139–148. [Google Scholar] [CrossRef] [PubMed]
  115. León-Reyes, G.; Guzmán-Grenfell, A.M.; Medina-Navarro, R.; Montoya-Estrada, A.; Moreno-Eutimio, M.A.; Fuentes-García, S.; Perichart Perera, O.; Muñoz-Manrique, C.; Espino y Sosa, S.; Hicks, G.J.J.; et al. Is Gestational Diabetes Mellitus in Obese Women Predicted by Oxidative Damage in Red Blood Cells? Gynecol. Endocrinol. 2018, 34, 995–1000. [Google Scholar] [CrossRef] [PubMed]
  116. Mostafavi, E.; Nargesi, A.A.; Asbagh, F.A.; Ghazizadeh, Z.; Heidari, B.; Mirmiranpoor, H.; Esteghamati, A.; Vigneron, C.; Nakhjavani, M. Abdominal Obesity and Gestational Diabetes: The Interactive Role of Magnesium. Magnes. Res. 2015, 28, 116–125. [Google Scholar] [CrossRef] [PubMed]
  117. Solis-Paredes, M.; Estrada-Gutierrez, G.; Perichart-Perera, O.; Montoya-Estrada, A.; Guzmán-Huerta, M.; Borboa-Olivares, H.; Bravo-Flores, E.; Cardona-Pérez, A.; Zaga-Clavellina, V.; Garcia-Latorre, E.; et al. Key Clinical Factors Predicting Adipokine and Oxidative Stress Marker Concentrations among Normal, Overweight and Obese Pregnant Women Using Artificial Neural Networks. Int. J. Mol. Sci. 2018, 19, 86. [Google Scholar] [CrossRef]
  118. Malti, N.; Merzouk, H.; Merzouk, S.A.; Loukidi, B.; Karaouzene, N.; Malti, A.; Narce, M. Oxidative Stress and Maternal Obesity: Feto-Placental Unit Interaction. Placenta 2014, 35, 411–416. [Google Scholar] [CrossRef]
  119. Hernández-Trejo, M.; Montoya-Estrada, A.; Torres-Ramos, Y.; Espejel-Núñez, A.; Guzmán-Grenfell, A.; Morales-Hernández, R.; Tolentino-Dolores, M.; Laresgoiti-Servitje, E. Oxidative Stress Biomarkers and Their Relationship with Cytokine Concentrations in Overweight/Obese Pregnant Women and Their Neonates. BMC Immunol. 2017, 18, 3. [Google Scholar] [CrossRef]
  120. Dao, M.C.; Sen, S.; Iyer, C.; Klebenov, D.; Meydani, S.N. Obesity during Pregnancy and Fetal Iron Status: Is Hepcidin the Link? J. Perinatol. 2013, 33, 177–181. [Google Scholar] [CrossRef]
  121. Zambon, M.; Mandò, C.; Lissoni, A.; Anelli, G.M.; Novielli, C.; Cardellicchio, M.; Leone, R.; Monari, M.N.; Massari, M.; Cetin, I.; et al. Inflammatory and Oxidative Responses in Pregnancies with Obesity and Periodontal Disease. Reprod. Sci. 2018, 25, 1474–1484. [Google Scholar] [CrossRef]
  122. Santos-Rosendo, C.; Bugatto, F.; González-Domínguez, A.; Lechuga-Sancho, A.M.; Mateos, R.M.; Visiedo, F. Placental Adaptive Changes to Protect Function and Decrease Oxidative Damage in Metabolically Healthy Maternal Obesity. Antioxidants 2020, 9, 794. [Google Scholar] [CrossRef] [PubMed]
  123. Sen, S.; Iyer, C.; Meydani, S.N. Obesity during Pregnancy Alters Maternal Oxidant Balance and Micronutrient Status. J. Perinatol. 2014, 34, 105–111. [Google Scholar] [CrossRef] [PubMed]
  124. Solis Paredes, J.M.; Perichart Perera, O.; Montoya Estrada, A.; Reyes Muñoz, E.; Espino, Y.; Sosa, S.; Ortega Castillo, V.; Medina Bastidas, D.; Tolentino Dolores, M.; Sanchez Martinez, M.; et al. Gestational Weight Gain Influences the Adipokine-Oxidative Stress Association during Pregnancy. Obes. Facts 2021, 14, 604–612. [Google Scholar] [CrossRef]
  125. Xie, W.; Wang, Y.; Xiao, S.; Qiu, L.; Yu, Y.; Zhang, Z. Association of Gestational Diabetes Mellitus with Overall and Type Specific Cardiovascular and Cerebrovascular Diseases: Systematic Review and Meta-Analysis. BMJ 2022, 378, e070244. [Google Scholar] [CrossRef]
  126. Sweeting, A.; Wong, J.; Murphy, H.R.; Ross, G.P. A Clinical Update on Gestational Diabetes Mellitus. Endocr. Rev. 2022, 43, 763–793. [Google Scholar] [CrossRef] [PubMed]
  127. Gaumnitz, T.; Jain, A.; Pertot, Y.; Huppert, M.; Jordan, I.; Ardana-Lamas, F.; Wörner, H.J. Streaking of 43-Attosecond Soft-X-Ray Pulses Generated by a Passively CEP-Stable Mid-Infrared Driver. Opt. Express 2017, 25, 27506. [Google Scholar] [CrossRef] [PubMed]
  128. Liu, X.; Sun, J.; Wen, X.; Duan, J.; Xue, D.; Pan, Y.; Sun, J.; Zhang, W.; Cheng, X.; Wang, C. Proteome Profiling of Gestational Diabetes Mellitus at 16–18 Weeks Revealed by LC-MS/MS. J. Clin. Lab. Anal. 2020, 34, e23424. [Google Scholar] [CrossRef] [PubMed]
  129. Shoelson, S.E.; Herrero, L.; Naaz, A. Obesity, Inflammation, and Insulin Resistance. Gastroenterology 2007, 132, 2169–2180. [Google Scholar] [CrossRef] [PubMed]
  130. Liu, S.; Li, X.; Wu, Y.; Duan, R.; Zhang, J.; Du, F.; Zhang, Q.; Li, Y.; Li, N. Effects of Vaspin on Pancreatic β Cell Secretion via PI3K/Akt and NF-ΚB Signaling Pathways. PLoS ONE 2017, 12, e0189722. [Google Scholar] [CrossRef]
  131. Li, C.; Cheng, H.; Adhikari, B.K.; Wang, S.; Yang, N.; Liu, W.; Sun, J.; Wang, Y. The Role of Apelin–APJ System in Diabetes and Obesity. Front. Endocrinol. 2022, 13, 1–11. [Google Scholar] [CrossRef]
  132. Sperling, M.; Grzelak, T.; Pelczyńska, M.; Bogdański, P.; Formanowicz, D.; Czyżewska, K. Association of Serum Omentin-1 Concentration with the Content of Adipose Tissue and Glucose Tolerance in Subjects with Central Obesity. Biomedicines 2023, 11, 331. [Google Scholar] [CrossRef]
  133. Briana, D.D.; Malamitsi-Puchner, A. Reviews: Adipocytokines in Normal and Complicated Pregnancies. Reprod. Sci. 2009, 16, 921–937. [Google Scholar] [CrossRef]
  134. Usluoğullari, B.; Usluogullari, C.A.; Balkan, F.; Orkmez, M. Role of Serum Levels of Irisin and Oxidative Stress Markers in Pregnant Women with and without Gestational Diabetes. Gynecol. Endocrinol. 2017, 33, 405–407. [Google Scholar] [CrossRef] [PubMed]
  135. Basu, J.; Datta, C.; Chowdhury, S.; Mandal, D.; Mondal, N.K.; Ghosh, A. Gestational Diabetes Mellitus in a Tertiary Care Hospital of Kolkata, India: Prevalence, Pathogenesis and Potential Disease Biomarkers. Exp. Clin. Endocrinol. Diabetes 2020, 128, 216–223. [Google Scholar] [CrossRef] [PubMed]
  136. Dincgez Cakmak, B.; Dundar, B.; Ketenci Gencer, F.; Yildiz, D.E.; Bayram, F.; Ozgen, G.; Aydin Boyama, B. Assessment of Relationship between Serum Vascular Adhesion Protein-1 (VAP-1) and Gestational Diabetes Mellitus. Biomarkers 2019, 24, 750–756. [Google Scholar] [CrossRef] [PubMed]
  137. Lappas, M.; Mittion, A.; Permezel, M. In Response to Oxidative Stress, the Expression of Inflammatory Cytokines and Antioxidant Enzymes Are Impaired in Placenta, but Not Adipose Tissue, of Women with Gestational Diabetes. J. Endocrinol. 2010, 204, 75–84. [Google Scholar] [CrossRef]
  138. Kurepa, D.; Pramanik, A.K.; Kakkilaya, V.; Caldito, G.; Groome, L.J.; Bocchini, J.A.; Jain, S.K. Elevated Acetoacetate and Monocyte Chemotactic Protein-1 Levels in Cord Blood of Infants of Diabetic Mothers. Neonatology 2012, 102, 163–168. [Google Scholar] [CrossRef]
  139. Martino, J.; Sebert, S.; Segura, M.T.; Garcia-Valdés, L.; Florido, J.; Padilla, M.C.; Marcos, A.; Rueda, R.; McArdle, H.J.; Budge, H.; et al. Maternal Body Weight and Gestational Diabetes Differentially Influence Placental and Pregnancy Outcomes. J. Clin. Endocrinol. Metab. 2016, 101, 59–68. [Google Scholar] [CrossRef]
  140. Renugasundari, M.; Pal, G.K.; Chaturvedula, L.; Nanda, N.; Harichandrakumar, K.T.; Durgadevi, T. Inflammation and Decreased Cardiovagal Modulation Are Linked to Stress and Depression at 36th Week of Pregnancy in Gestational Diabetes Mellitus. Sci. Rep. 2023, 13, 10348. [Google Scholar] [CrossRef]
  141. Yakut, K.; Öcal, D.F.; Öztürk, F.H.; Öztürk, M.; Oğuz, Y.; Sınacı, S.; Çağlar, T. Is GDF-15 Level Associated with Gestational Diabetes Mellitus and Adverse Perinatal Outcomes? Taiwan J. Obstet. Gynecol. 2021, 60, 221–224. [Google Scholar] [CrossRef]
  142. Mandò, C.; Anelli, G.M.; Novielli, C.; Panina-Bordignon, P.; Massari, M.; Mazzocco, M.I.; Cetin, I. Impact of Obesity and Hyperglycemia on Placental Mitochondria. Oxid. Med. Cell. Longev. 2018, 2018, 2378189. [Google Scholar] [CrossRef]
  143. Clausen, T.D.; Mathiesen, E.R.; Hansen, T.; Pedersen, O.; Jensen, D.M.; Lauenborg, J.; Schmidt, L.; Damm, P. Overweight and the Metabolic Syndrome in Adult Offspring of Women with Diet-Treated Gestational Diabetes Mellitus or Type 1 Diabetes. J. Clin. Endocrinol. Metab. 2009, 94, 2464–2470. [Google Scholar] [CrossRef] [PubMed]
  144. Lu, J.; Zhang, S.; Li, W.; Leng, J.; Wang, L.; Liu, H.; Li, W.; Zhang, C.; Qi, L.; Tuomilehto, J.; et al. Maternal Gestational Diabetes Is Associated with Offspring’s Hypertension. Am. J. Hypertens. 2019, 32, 335–342. [Google Scholar] [CrossRef]
  145. Bayman, M.G.; Inal, Z.O.; Hayiroglu, F.; Ozturk, E.N.Y.; Gezginc, K. Foetal Umbilical Cord Brain-Derived Neurotrophic Factor (BDNF) Levels in Pregnancy with Gestational Diabetes Mellitus. J. Obstet. Gynaecol. 2022, 42, 1097–1102. [Google Scholar] [CrossRef] [PubMed]
  146. Taschereau-Charron, A.; Bilodeau, J.F.; Larose, J.; Greffard, K.; Berthiaume, L.; Audibert, F.; Fraser, W.D.; Julien, P.; Rudkowska, I. F 2-Isoprostanes and Fatty Acids Profile in Early Pregnancy Complicated by Pre-Existing Diabetes. Prostaglandins Leukot. Essent. Fat. Acids 2018, 135, 115–120. [Google Scholar] [CrossRef]
  147. Piuri, G.; Basello, K.; Rossi, G.; Soldavini, C.M.; Duiella, S.; Privitera, G.; Spadafranca, A.; Costanzi, A.; Tognon, E.; Cappelletti, M.; et al. Methylglyoxal, Glycated Albumin, PAF, and TNF-α: Possible Inflammatory and Metabolic Biomarkers for Management of Gestational Diabetes. Nutrients 2020, 12, 479. [Google Scholar] [CrossRef]
  148. Phoswa, W.N.; Khaliq, O.P. The Role of Oxidative Stress in Hypertensive Disorders of Pregnancy (Preeclampsia, Gestational Hypertension) and Metabolic Disorder of Pregnancy (Gestational Diabetes Mellitus). Oxid. Med. Cell. Longev. 2021, 2021, 5581570. [Google Scholar] [CrossRef]
  149. Zych, B.; Górka, A.; Myszka, A.; Błoniarz, D.; Siekierzyńska, A.; Błaż, W. Status of Oxidative Stress during Low-Risk Labour: Preliminary Data. Int. J. Environ. Res. Public Health 2023, 20, 157. [Google Scholar] [CrossRef]
  150. Li, J.; Xu, Y.; Sun, Z.; Cai, Y.; Wang, B.; Zhang, M.; Ban, Y.; Hou, X.; Hao, Y.; Ouyang, Q.; et al. Differential Lipids in Pregnant Women with Subclinical Hypothyroidism and Their Correlation to the Pregnancy Outcomes. Sci. Rep. 2021, 11, 19689. [Google Scholar] [CrossRef] [PubMed]
  151. Zhou, Y.; Peng, H.; Xu, H.; Li, J.; Golovko, M.; Cheng, H.; Lynch, E.C.; Liu, L.; McCauley, N.; Kennedy, L.; et al. Maternal Diet Intervention before Pregnancy Primes Offspring Lipid Metabolism in Liver. Lab. Investig. 2020, 100, 553–569. [Google Scholar] [CrossRef]
  152. Galvan-Martinez, D.H.; Bosquez-Mendoza, V.M.; Ruiz-Noa, Y.; del Rocio Ibarra-Reynoso, L.; Barbosa-Sabanero, G.; Lazo-De-La-Vega-Monroy, M.L. Nutritional, Pharmacological, and Environmental Programming of NAFLD in Early Life. Am. J. Physiol.—Gastrointest. Liver Physiol. 2023, 324, G99–G114. [Google Scholar] [CrossRef]
  153. Hagström, H.; Simon, T.G.; Roelstraete, B.; Stephansson, O.; Söderling, J.; Ludvigsson, J.F. Maternal Obesity Increases the Risk and Severity of NAFLD in Offspring. J. Hepatol. 2021, 75, 1042–1048. [Google Scholar] [CrossRef]
  154. Mikolajczak, A.; Sallam, N.A.; Singh, R.D.; Scheidl, T.B.; Walsh, E.J.; Larion, S.; Huang, C.; Thompson, J.A. Accelerated Developmental Adipogenesis Programs Adipose Tissue Dysfunction and Cardiometabolic Risk in Offspring Born to Dams with Metabolic Dysfunction. Am. J. Physiol.—Endocrinol. Metab. 2021, 321, E581–E591. [Google Scholar] [CrossRef] [PubMed]
  155. Scheidl, T.B.; Wager, J.L.; Baker, L.G.; Brightwell, A.L.; Melan, K.M.; Larion, S.; Sarr, O.; Regnault, T.; Urbanski, S.J.; Thompson, J. High Maternal Adiposity During Pregnancy Programs an Imbalance in the Lipidome and Predisposes to Diet-Induced Hepatosteatosis in the Offspring. Biosci. Rep. 2023, 43, BSR20231060. [Google Scholar] [CrossRef] [PubMed]
  156. Reyes, R.B.; Carrocera, L.A.F. Programación Metabólica Fetal. Perinatol. Y Reprod. Humana 2015, 29, 99–105. [Google Scholar] [CrossRef]
  157. Charlton, F.; Tooher, J.; Rye, K.A.; Hennessy, A. Cardiovascular Risk, Lipids and Pregnancy: Preeclampsia and the Risk of Later Life Cardiovascular Disease. Hear. Lung Circ. 2014, 23, 203–212. [Google Scholar] [CrossRef] [PubMed]
  158. Adank, M.C.; Johansen, A.K.; Benschop, L.; Van Streun, S.P.; Smak Gregoor, A.M.; Øyri, L.K.L.; Mulder, M.T.; Steegers, E.A.P.; Holven, K.B.; Roeters van Lennep, J.E. Maternal Lipid Levels in Early Pregnancy as a Predictor of Childhood Lipid Levels: A Prospective Cohort Study. BMC Pregnancy Childbirth 2022, 22, 588. [Google Scholar] [CrossRef] [PubMed]
  159. Bausserman, L.L.; Herbert, P.N.; McAdam, K.P. Heterogeneity of Human Serum Amyloid A Proteins. J. Exp. Med. 1980, 52, 641–656. [Google Scholar] [CrossRef]
  160. Davidson, W.S.; Shah, A.S.; Sexmith, H.; Gordon, S.M. The HDL Proteome Watch: Compilation of Studies Leads to New Insights on HDL Function. Biochim. Biophys. Acta—Mol. Cell Biol. Lipids 2022, 1867, 159072. [Google Scholar] [CrossRef]
  161. Shah, A.S.; Tan, L.; Long, J.L.; Davidson, W.S. Proteomic Diversity of High Density Lipoproteins: Our Emerging Understanding of Its Importance in Lipid Transport and Beyond. J. Lipid Res. 2013, 54, 2575–2585. [Google Scholar] [CrossRef]
  162. Ouimet, M.; Barrett, T.J.; Fisher, E.A. HDL and Reverse Cholesterol Transport: Basic Mechanisms and Their Roles in Vascular Health and Disease. Circ. Res. 2019, 124, 1505–1518. [Google Scholar] [CrossRef]
  163. Cuchel, M.; Rader, D.J. Macrophage Reverse Cholesterol Transport: Key to the Regression of Atherosclerosis? Circulation 2006, 113, 2548–2555. [Google Scholar] [CrossRef]
  164. Chen, C.; Khismatullin, D.B. Oxidized Low-Density Lipoprotein Contributes to Atherogenesis via Co-Activation of Macrophages and Mast Cells. PLoS ONE 2015, 10, e0123088. [Google Scholar] [CrossRef]
  165. Rysz, J.; Gluba-Brzózka, A.; Rysz-Górzyńska, M.; Franczyk, B. The Role and Function of HDL in Patients with Chronic Kidney Disease and the Risk of Cardiovascular Disease. Int. J. Mol. Sci. 2020, 21, 601. [Google Scholar] [CrossRef] [PubMed]
  166. Buendía-Abad, M.; García-Palencia, P.; de Pablos, L.M.; Alunda, J.M.; Osuna, A.; Martín-Hernández, R.; Higes, M. First Description of Lotmaria Passim and Crithidia Mellificae Haptomonad Stages in the Honeybee Hindgut. Int. J. Parasitol. 2022, 52, 65–75. [Google Scholar] [CrossRef] [PubMed]
  167. Stadler, J.T.; Scharnagl, H.; Wadsack, C.; Marsche, G. Preeclampsia Affects Lipid Metabolism and HDL Function in Mothers and Their Offspring. Antioxidants 2023, 12, 795. [Google Scholar] [CrossRef] [PubMed]
  168. Eren, E. High Density Lipoprotein and It’s Dysfunction. Open Biochem. J. 2012, 6, 78–93. [Google Scholar] [CrossRef]
  169. Gordon, T.; Castelli, W.P.; Hjortland, M.C.; Kannel, W.B.; Dawber, T.R. High Density Lipoprotein as a Protective Factor against Coronary Heart Disease. The Framingham Study. Am. J. Med. 1977, 62, 707–714. [Google Scholar] [CrossRef]
  170. Patanapirunhakit, P.; Karlsson, H.; Mulder, M.; Ljunggren, S.; Graham, D.; Freeman, D. Sphingolipids in HDL—Potential Markers for Adaptation to Pregnancy? Biochim. Biophys. Acta—Mol. Cell Biol. Lipids 2021, 1866, 158955. [Google Scholar] [CrossRef]
  171. Luna-Luna, M.; Niesor, E.; Pérez-Méndez, Ó. HDL as Bidirectional Lipid Vectors: Time for New Paradigms. Biomedicines 2022, 10, 1180. [Google Scholar] [CrossRef]
  172. Chen, R.; Chen, Q.; Zheng, J.; Zeng, Z.; Chen, M.; Li, L.; Zhang, S. Serum Amyloid Protein A in Inflammatory Bowel Disease: From Bench to Bedside. Cell Death Discov. 2023, 9, 154. [Google Scholar] [CrossRef] [PubMed]
  173. Streese, L.; Habisch, H.; Deiseroth, A.; Carrard, J.; Infanger, D.; Schmidt-Trucksäss, A.; Madl, T.; Hanssen, H. Lipoprotein Subclasses Independently Contribute to Subclinical Variance of Microvascular and Macrovascular Health. Molecules 2022, 27, 4760. [Google Scholar] [CrossRef]
  174. Woudberg, N.J.; Goedecke, J.H.; Blackhurst, D.; Frias, M.; James, R.; Opie, L.H.; Lecour, S. Association between Ethnicity and Obesity with High-Density Lipoprotein (HDL) Function and Subclass Distribution. Lipids Health Dis. 2016, 15, 92. [Google Scholar] [CrossRef]
  175. Muñoz-Vega, M.; Massó, F.; Páez, A.; Vargas-Alarcón, G.; Coral-Vázquez, R.; Mas-Oliva, J.; Carreón-Torres, E.; Pérez-Méndez, Ó. Hdl-Mediated Lipid Influx to Endothelial Cells Contributes to Regulating Intercellular Adhesion Molecule (Icam)-1 Expression and Enos Phosphorylation. Int. J. Mol. Sci. 2018, 19, 3394. [Google Scholar] [CrossRef] [PubMed]
  176. Brites, F.; Martin, M.; Guillas, I.; Kontush, A. Antioxidative Activity of High-Density Lipoprotein (HDL): Mechanistic Insights into Potential Clinical Benefit. BBA Clin. 2017, 8, 66–77. [Google Scholar] [CrossRef]
  177. Jayaraman, S.; Haupt, C.; Gursky, O. Thermal Transitions in Serum Amyloid A in Solution and on the Lipid: Implications for Structure and Stability of Acute-Phase HDL. J. Lipid Res. 2015, 56, 1531–1542. [Google Scholar] [CrossRef]
  178. Göçmen, A.Y.; Şahin, E.; Semiz, E.; Gümüslü, S. Is Elevated Serum Ceruloplasmin Level Associated with Increased Risk of Coronary Artery Disease? Can. J. Cardiol. 2008, 24, 209–212. [Google Scholar] [CrossRef] [PubMed]
  179. Khovidhunkit, W.; Kim, M.S.; Memon, R.A.; Shigenaga, J.K.; Moser, A.H.; Feingold, K.R.; Grunfeld, C. Effects of Infection and Inflammation on Lipid and Lipoprotein Metabolism: Mechanisms and Consequences to the Host. J. Lipid Res. 2004, 45, 1169–1196. [Google Scholar] [CrossRef]
  180. Rosenson, R.S.; Brewer, H.B., Jr.; Ansell, B.J.; Barter, P.; Chapman, M.J.; Heinecke, J.W.; Kontush, A.; Tall, A.R.; Webb, N.R. Dysfunctional HDL and Atherosclerotic Cardiovascular Disease. Nat. Rev. Cardiol. 2016, 13, 48–60. [Google Scholar] [CrossRef]
  181. Zafaranieh, S.; Stadler, J.T.; Pammer, A.; Marsche, G.; van Poppel, M.N.M.; Desoye, G. The Association of Physical Activity and Sedentary Behavior with Maternal and Cord Blood Anti-Oxidative Capacity and HDL Functionality: Findings of DALI Study. Antioxidants 2023, 12, 827. [Google Scholar] [CrossRef]
  182. Schliefsteiner, C.; Hirschmugl, B.; Kopp, S.; Curcic, S.; Bernhart, E.M.; Marsche, G.; Lang, U.; Desoye, G.; Wadsack, C. Maternal Gestational Diabetes Mellitus Increases Placental and Foetal Lipoprotein-Associated Phospholipase A2 Which Might Exert Protective Functions against Oxidative Stress. Sci. Rep. 2017, 7, 12628. [Google Scholar] [CrossRef]
  183. León-Reyes, G.; Espino, Y.; Sosa, S.; Medina-Navarro, R.; Guzmán-Grenfell, A.M.; Medina-Urrutia, A.X.; Fuentes-García, S.; Hicks, G.J.J.; Torres-Ramos, Y.D. Oxidative Modifications of Foetal LDL-c and HDL-c Lipoproteins in Preeclampsia. Lipids Health Dis. 2018, 17, 110. [Google Scholar] [CrossRef] [PubMed]
  184. Jayalekshmi, V.S.; Jaikumar, V.S.; Mehra, P.; Thulaseedharan, T.; Vinod, V.M.; Ramachandran, S. Differential Expression of Lipid Metabolic Genes in Hypercholesterolemic Rabbit Placenta Predisposes the Offspring to Develop Atherosclerosis in Early Adulthood. Life Sci. 2023, 327, 121823. [Google Scholar] [CrossRef]
  185. Rodríguez-Hernández, H.; Simental-Mendía, L.E. The Triglycerides and Glucose Index Is Highly Associated with Non-Alcoholic Fatty Liver Disease in Overweight and Obese Women. Ir. J. Med. Sci. 2023, 2023, 11845. [Google Scholar] [CrossRef] [PubMed]
  186. Mocciaro, G.; Allison, M.; Jenkins, B.; Azzu, V.; Huang-Doran, I.; Herrera-Marcos, L.V.; Hall, Z.; Murgia, A.; Susan, D.; Frontini, M.; et al. Non-Alcoholic Fatty Liver Disease Is Characterised by a Reduced Polyunsaturated Fatty Acid Transport via Free Fatty Acids and High-Density Lipoproteins (HDL). Mol. Metab. 2023, 73, 101728. [Google Scholar] [CrossRef]
  187. Pelczyńska, M.; Moszak, M.; Wesołek, A.; Bogdański, P. The Preventive Mechanisms of Bioactive Food Compounds against Obesity-Induced Inflammation. Antioxidants 2023, 12, 1232. [Google Scholar] [CrossRef]
  188. Pathikkal, A.; Puthusseri, B.; Divya, P.; Rudrappa, S.; Chauhan, V.S. Folate Derivatives, 5-Methyltetrahydrofolate and 10-Formyltetrahydrofolate, Protect BEAS-2B Cells from High Glucose–Induced Oxidative Stress and Inflammation. Vitr. Cell. Dev. Biol.—Anim. 2022, 58, 419–428. [Google Scholar] [CrossRef]
  189. Zhou, K.; Raffoul, J.J. Potential Anticancer Properties of Grape Antioxidants. J. Oncol. 2012, 2012, 803294. [Google Scholar] [CrossRef]
  190. Delmastro-Greenwood, M.; Freeman, B.A.; Wendell, S.G. Redox-Dependent Anti-Inflammatory Signaling Actions of Unsaturated Fatty Acids. Annu. Rev. Physiol. 2014, 76, 79–105. [Google Scholar] [CrossRef] [PubMed]
  191. Gong, S.; Ji, X.; Su, J.; Wang, Y.; Yan, X.; Wang, G.; Xiao, B.; Dong, H.; Xiang, X.; Liu, S. Yeast Fermentate Prebiotic Ameliorates Allergic Asthma, Associating with Inhibiting Inflammation and Reducing Oxidative Stress Level through Suppressing Autophagy. Mediat. Inflamm. 2021, 2021, 4080935. [Google Scholar] [CrossRef]
  192. Dai, Y.; Quan, J.; Xiong, L.; Luo, Y.; Yi, B. Probiotics Improve Renal Function, Glucose, Lipids, Inflammation and Oxidative Stress in Diabetic Kidney Disease: A Systematic Review and Meta-Analysis. Ren. Fail. 2022, 44, 862–880. [Google Scholar] [CrossRef] [PubMed]
  193. Kelesidis, T.; Jackson, N.; Mccomsey, G.A.; Wang, X.; Elashoff, D.; Dube, M.P.; Brown, T.T.; Yang, O.O.; Stein, J.H.; Currier, J.S. Oxidized Lipoproteins Are Associated with Markers of Inflammation and Immune Activation in HIV-1 Infection. AIDS 2016, 30, 2625–2633. [Google Scholar] [CrossRef]
  194. Vergès, B. Dyslipidemia in Type 1 Diabetes: A Masked Danger. Trends Endocrinol. Metab. 2020, 31, 422–434. [Google Scholar] [CrossRef]
  195. Ganjali, S.; Dallinga-Thie, G.M.; Simental-Mendía, L.E.; Banach, M.; Pirro, M.; Sahebkar, A. HDL Functionality in Type 1 Diabetes. Atherosclerosis 2017, 267, 99–109. [Google Scholar] [CrossRef]
  196. Stadler, J.T.; Marsche, G. Dietary Strategies to Improve Cardiovascular Health: Focus on Increasing High-Density Lipoprotein Functionality. Front. Nutr. 2021, 8, 1–16. [Google Scholar] [CrossRef]
  197. Cardner, M.; Yalcinkaya, M.; Goetze, S.; Luca, E.; Balaz, M.; Hunjadi, M.; Hartung, J.; Shemet, A.; Kränkel, N.; Radosavljevic, S.; et al. Structure-Function Relationships of HDL in Diabetes and Coronary Heart Disease. JCI Insight 2020, 5, 1–18. [Google Scholar] [CrossRef] [PubMed]
  198. Luna-Castillo, K.P.; Lin, S.; Muñoz-Valle, J.F.; Vizmanos, B.; López-Quintero, A.; Márquez-Sandoval, F. Functional Food and Bioactive Compounds on the Modulation of the Functionality of Hdl-c: A Narrative Review. Nutrients 2021, 13, 1165. [Google Scholar] [CrossRef]
  199. Vaisar, T.; Kanter, J.E.; Wimberger, J.; Irwin, A.D.; Gauthier, J.; Wolfson, E.; Bahnam, V.; Wu, I.H.; Shah, H.; Keenan, H.A.; et al. High Concentration of Medium-Sized HDL Particles and Enrichment in HDL Paraoxonase 1 Associate with Protection from Vascular Complications in People with Long-Standing Type 1 Diabetes. Diabetes Care 2020, 43, 178–186. [Google Scholar] [CrossRef] [PubMed]
  200. Gu, X.; Huang, Y.; Levison, B.S.; Gerstenecker, G.; DiDonato, A.J.; Hazen, L.B.; Lee, J.; Gogonea, V.; DiDonato, J.A.; Hazen, S.L. Identification of Critical Paraoxonase 1 Residues Involved in High Density Lipoprotein Interaction. J. Biol. Chem. 2016, 291, 1890–1904. [Google Scholar] [CrossRef] [PubMed]
  201. Dorantes-Morales, A.; Estrada-Luna, D.; Bautista-Pérez, R.; Betanzos-Cabrera, G.; Luna-Luna, M.; Flores-Castillo, C.; Vargas-Alarcón, G.; Fragoso, J.M.; Pérez-Méndez, Ó.; Carreón-Torres, E.; et al. Microencapsulated Pomegranate Modifies the Composition and Function of High-Density. Molecules 2020, 25, 3297. [Google Scholar] [CrossRef]
  202. Hernáez, Á.; Castañer, O.; Elosua, R.; Pintó, X.; Estruch, R.; Salas-Salvadó, J.; Corella, D.; Arós, F.; Serra-Majem, L.; Fiol, M.; et al. Mediterranean Diet Improves High-Density Lipoprotein Function in High-Cardiovascular-Risk Individuals. Circulation 2017, 135, 633–643. [Google Scholar] [CrossRef] [PubMed]
  203. Hernáez, Á.; Fernández-Castillejo, S.; Farràs, M.; Catalán, Ú.; Subirana, I.; Montes, R.; Solà, R.; Muñoz-Aguayo, D.; Gelabert-Gorgues, A.; Díaz-Gil, Ó.; et al. Olive Oil Polyphenols Enhance High-Density Lipoprotein Function in Humans: A Randomized Controlled Trial. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 2115–2119. [Google Scholar] [CrossRef] [PubMed]
  204. Sola, R.; Valls, R.M.; Puzo, J.; Calabuig, J.R.; Brea, A.; Pedret, A.; Morinã, D.; Villar, J.; Millán, J.; Anguera, A. Effects of Poly-Bioactive Compounds on Lipid Profile and Body Weight in a Moderately Hypercholesterolemic Population with Low Cardiovascular Disease Risk: A Multicenter Randomized Trial. PLoS ONE 2014, 9, e101978. [Google Scholar] [CrossRef]
  205. Bub, A.; Malpuech-Brugère, C.; Orfila, C.; Amat, J.; Arianna, A.; Blot, A.; Di Nunzio, M.; Holmes, M.; Kertész, Z.; Marshall, L.; et al. A Dietary Intervention of Bioactive Enriched Foods Aimed at Adults at Risk of Metabolic Syndrome: Protocol and Results from PATHWAY-27 Pilot Study. Nutrients 2019, 11, 1814. [Google Scholar] [CrossRef]
  206. Muralidharan, J.; Papandreou, C.; Soria-Florido, M.T.; Sala-Vila, A.; Blanchart, G.; Estruch, R.; Martínez-González, M.A.; Corella, D.; Ros, E.; Ruiz-Canela, M.; et al. Cross-Sectional Associations between HDL Structure or Function, Cell Membrane Fatty Acid Composition, and Inflammation in Elderly Adults. J. Nutr. 2022, 152, 789–795. [Google Scholar] [CrossRef] [PubMed]
  207. Estrada-Luna, D.; Carreón-Torres, E.; Bautista-Perez, R.; Betanzos-Cabrera, G.; Dorantes-Morales, A.; Luna-Luna, M.; Vargas-Barrón, J.; Mejía, A.M.; Fragoso, J.M.; Carvajal-Aguilera, K.; et al. Microencapsulated Pomegranate Reverts High-Density Lipoprotein (HDL)-Induced Endothelial Dysfunction and Reduces Postprandial Triglyceridemia in Women with Acute Coronary Syndrome. Nutrients 2019, 11, 1710. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Inflammation and OS processes during and after pregnancy.
Figure 1. Inflammation and OS processes during and after pregnancy.
Antioxidants 12 01894 g001
Figure 2. General structure for lipoproteins.
Figure 2. General structure for lipoproteins.
Antioxidants 12 01894 g002
Figure 3. Functional and dysfunctional HDL during pregnancy.
Figure 3. Functional and dysfunctional HDL during pregnancy.
Antioxidants 12 01894 g003
Table 1. Pregnancy phases and inflammation.
Table 1. Pregnancy phases and inflammation.
TrimesterCytokinesClinical Outcome in the MotherClinical Outcome in the Fetus/InfantRef.
First Trimester
(Proinflammatory)
IL-6Infection and chorioamnionitis increased prevalenceFetal inflammatory response disturbances
Reactive airway disease
Ectopic lipid accumulation in liver
[61,62,63]
IL-8Preeclampsia developmentInsulin resistance
Bronchopulmonary dysplasia
[51,61,63,64]
TNF-αIncreases obesity and GDM riskAdipose tissue alterations[61,65]
MCP-1Increased risk of miscarriage and GDM riskUndetermined[61]
RANTES
G-SCF
Increased risk of miscarriageUndetermined[61]
Second TrimesterIL-4Decreases the risk of preterm birth, preeclampsia
Increased risk of systemic lupus erythematosus (SLE)
Undetermined[61]
IL-10Decreases the risk of preterm birth and preeclampsia
Increases obesity and GDM risk
Decreased expression of the inflammatory cytokine gene in the uterus and placenta[61,66]
IL-13Development of fetal inflammatory response syndromeUndetermined[61]
Third Trimester
(Proinflammatory)
IL-6Increased risk of miscarriage, obesity, GDM, and preeclampsia risk
Development of reactive airway disease (RAD)
Development of reactive airway disease (RAD)[51,61,62,63,65,66,67]
IL-8Increased prevalence of infection, preeclampsia, and chorioamnionitis
Development of fetal inflammatory response syndrome
Undetermined[61,66]
MCP-1Increased risk of miscarriage, autoimmune diseases, and GDM Undetermined[61]
RANTES-G-SCFIncreased risk of miscarriageUndetermined[61]
TNF-αPreeclampsiaWheezing and lower respiratory tract infections.
Intrauterine growth restriction
[61,63,67]
Table 3. Markers of OS and inflammation in pregnant women with obesity.
Table 3. Markers of OS and inflammation in pregnant women with obesity.
Study GroupsOxidative and Inflammatory MarkersOS ChangesInflammation ChangesEffects in Mothers and NewbornsRef.
PW Ctrl = 28
PWOw = 26
PWOb = 26
MDA, LPH in plasma
CP in RBCs
Plasmatic IL-6 and TNF-α
↑ MDA, LOOH and CP in PWOb and PWOw vs. Ctrl in the 3rd trimester vs. the 2nd trimester↑ TNF-α in PWOb vs. PW at 12–13 months of gestation
↑ TNF-α and IL-6 in women who developed GDM
+ Correlation of LOOH, MDA and CP with gestational age in all groups and higher levels of these markers in PW who developed GDM vs. PWOb in 2nd and 3rd trimesters[115]
Ctrl = 15
PWOb = 15
Serum iron and transferrin saturation (Tsat), CRP, IL-6, GSH, and GSSGRatio of serum GSSG/GSH was higher in the PWOb compared to Ctrl↑ TNF-α in PWOb vs. Ctrl.
No changes in IL-6 and iron
+ Correlation between maternal BMI and CRP.
- Correlation between BMI and iron status in cord blood.
Serum iron and Tsat were found to be significantly lower in cord blood from PWOb compared to Ctrl
[120]
Ctrl = 40
PWOb = 40
Serum MDA and hsCRP at 28 weeks of gestation↑ MDA and glucose after 3 h OGTT were higher in PWOb
No changes at baseline.
Higher levels of hsCRP at baseline and after 3 h of OGTT in PWOb vs. CtrlsAfter the OGTT at 28 weeks of gestation, 16 patients in the obese group and 1 in the control group had GDM.[116]
Weight gain
Insufficient, IWG = 28
Normal, NWG = 20
Excessive, EWG = 26
Serum LOOH, MDA, CP, 8-oxodG, adiponectin, leptin, and resistinNo significant differences in LOOH, MDA, and CP concentrations between groups. A trend to lower 8-oxodG in PWOb↓ Adiponectin in EWG
+ Correlation between Adiponectin and 8-oxodG concentration in NWG. In EWG, leptin was associated with LOOH, MDA, and CP.
ND[124]
Ctrl = 25
PWOw = 21
PWOb = 22
Adiponectin, leptin, and resistin in serum. MDA, CP, and 8-oxodG in plasma↑ MDA and CP in PWOb vs. Ctrl↓ Adiponectin and resistin in PWObBMI estimating adiponectin and CP concentrations (37%). Gestational age predicts resistin and MDA (34%).[117]
Ctrl = 15
PWOb = 15
CRP, GSSH, GSH, TNF-α and IL-6 levels in serum and cord↓ GSSG/GSH ratio in PWOb vs. Ctrl↑ CRP and IL-6 in PWOb vs. Ctrl.
↑ CRP in cord of PWOb vs. Ctrl
Maternal BMI correlated with CRP, GSSG/GSH ratio, and IL-6.
Infants born from PWOb mothers did not have higher measures of OS.
[120]
Ctrl = 50
PWOb = 40
Nitric oxide (NO), CAT, SOD, GSH, CP and MDA in plasma and placenta↓ SOD and CAT and GSSG levels in PWOb and their newborns vs. Ctrl
↑ MDA, CP, Nitrite and Red-NTB in plasma PWOb and their newborns vs. Ctrl
↑ CAT, SOD, GSH, MDA and CP in placenta of PWOb vs. Ctrl
+ Correlation between maternal and fetal levels of TG and plasma and placental MDA.
Maternal nitrite and reduced NBT levels were significantly correlated with those of newborns of PWOb
[118]
Ctrl = 27
PWOb = 18
PW with obesity and GDM (ObGDM) = 17
TAC and CRP in saliva
CRP in plasma
↑ TAC in saliva in ObGDM vs. PW.
+ Correlation between TAC and CRP in saliva or plasma
↑ CRP in saliva and plasma in ObGDM vs. PW.
↑ CRP in plasma in PWOb vs. PW
TAC and CRP correlates positively with BMI and glycemia.
A significant interaction between maternal BMI and periodontitis for s-TAC levels.
[121]
Additional abbreviations. CP, carbonylated proteins; EWG, excessive weight gain; LOOH, lipohydroperoxides; PWOb, pregnant women with obesity; PWOw, pregnant women with overweight; red NBT, reduced nitroblue tetrazolium.
Table 5. HDL main functions.
Table 5. HDL main functions.
HDL FunctionProteins and Lipids Associated with HDL FunctionRef.
Reverse cholesterol transportHDL promotes cholesterol efflux from various cell types.ABCAI, ABCG1, SR-BI, cubilin, ApoE receptor[168,169]
Removing excess cholesterol from lipid-laden macrophages is a crucial process in HDL-mediated vascular protection.
OxidantHDL has antioxidant properties whereby it can remove and inactivate lipid peroxides from LDL and cells.PON 1, Apo AI, PAF-AH, LCAT, Apo M, S1P, phospholipids[159]
InflammationControlling the activation of monocytes, preventing macrophage migration, and inhibiting the oxidation of LDL by blocking the 12-lipoxygenase that produces lipid hydroperoxides and leads to the oxidation of the LDLVCAM, ICAM, TNF-α, SAA, ceramides[169]
Vascular functionModulation of endothelial nitric oxide synthase (eNOS) expression, leading to increased nitric oxide (NO) production and vasodilationABCA1, SR-BI, S1PR, S1P, Apo M[170,171,172]
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

Jiménez-Osorio, A.S.; Carreón-Torres, E.; Correa-Solís, E.; Ángel-García, J.; Arias-Rico, J.; Jiménez-Garza, O.; Morales-Castillejos, L.; Díaz-Zuleta, H.A.; Baltazar-Tellez, R.M.; Sánchez-Padilla, M.L.; et al. Inflammation and Oxidative Stress Induced by Obesity, Gestational Diabetes, and Preeclampsia in Pregnancy: Role of High-Density Lipoproteins as Vectors for Bioactive Compounds. Antioxidants 2023, 12, 1894. https://doi.org/10.3390/antiox12101894

AMA Style

Jiménez-Osorio AS, Carreón-Torres E, Correa-Solís E, Ángel-García J, Arias-Rico J, Jiménez-Garza O, Morales-Castillejos L, Díaz-Zuleta HA, Baltazar-Tellez RM, Sánchez-Padilla ML, et al. Inflammation and Oxidative Stress Induced by Obesity, Gestational Diabetes, and Preeclampsia in Pregnancy: Role of High-Density Lipoproteins as Vectors for Bioactive Compounds. Antioxidants. 2023; 12(10):1894. https://doi.org/10.3390/antiox12101894

Chicago/Turabian Style

Jiménez-Osorio, Angélica Saraí, Elizabeth Carreón-Torres, Emmanuel Correa-Solís, Julieta Ángel-García, José Arias-Rico, Octavio Jiménez-Garza, Lizbeth Morales-Castillejos, Hugo Alexander Díaz-Zuleta, Rosa María Baltazar-Tellez, María Luisa Sánchez-Padilla, and et al. 2023. "Inflammation and Oxidative Stress Induced by Obesity, Gestational Diabetes, and Preeclampsia in Pregnancy: Role of High-Density Lipoproteins as Vectors for Bioactive Compounds" Antioxidants 12, no. 10: 1894. https://doi.org/10.3390/antiox12101894

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