Next Article in Journal
Association between Dietary and Supplemental Antioxidants Intake and Lung Cancer Risk: Evidence from a Cancer Screening Trial
Next Article in Special Issue
Microbiome Changes in Pregnancy Disorders
Previous Article in Journal
An Original Asteraceae Based Infused Drink Prevents Metabolic Syndrome in Fructose-Rat Model
Previous Article in Special Issue
Evidence of Increased Oxidative Stress in the Placental Tissue of Women Who Suffered an Episode of Psychosis during Pregnancy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Effect of Resveratrol on Pregnancy, Prenatal Complications and Pregnancy-Associated Structure Alterations

1
Departement de Biologie Animale, Université des Frères Mentouri Constantine 1, Constantine 25000, Algeria
2
Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
3
College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates
4
Department of Pharmacology and Toxicology, American University of Beirut, Beirut 11-0236, Lebanon
5
Department of Biology, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
6
Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha 2713, Qatar
7
Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar
*
Author to whom correspondence should be addressed.
Antioxidants 2023, 12(2), 341; https://doi.org/10.3390/antiox12020341
Submission received: 13 December 2022 / Revised: 24 January 2023 / Accepted: 28 January 2023 / Published: 31 January 2023

Abstract

:
Adverse pregnancy outcomes are considered significant health risks for pregnant women and their offspring during pregnancy and throughout their lifespan. These outcomes lead to a perturbated in-utero environment that impacts critical phases of the fetus’s life and correlates to an increased risk of chronic pathological conditions, such as diabetes, obesity, and cardiovascular diseases, in both the mother’s and adult offspring’s life. The dietary intake of naturally occurring antioxidants promotes health benefits and disease prevention. In this regard, maternal dietary intake of polyphenolic antioxidants is linked to a reduced risk of maternal obesity and cardio-metabolic disorders, positively affecting both the fetus and offspring. In this work, we will gather and critically appraise the current literature highlighting the effect/s of the naturally occurring polyphenol antioxidant resveratrol on oxidative stress, inflammation, and other molecular and physiological phenomena associated with pregnancy and pregnancy conditions, such as gestational diabetes, preeclampsia, and preterm labor. The resveratrol impact on prenatal complications and pregnancy-associated structures, such as the fetus and placenta, will also be discussed. Finally, we will draw conclusions from the current knowledge and provide future perspectives on potentially exploiting resveratrol as a therapeutic tool in pregnancy-associated conditions.

1. Introduction

Adverse pregnancy outcomes are considered major health risks for pregnant women and their offspring during pregnancy and throughout their lifespan, with several disorders recognized, including gestational diabetes, preterm birth, fetal-growth restriction, and hypertensive disorders of pregnancy such as gestational hypertension, pre-eclampsia, and related conditions [1,2]. These outcomes lead to a perturbated in-utero environment that impacts critical phases of fetus development. An increased risk of chronic non-communicable metabolic diseases, such as cardiovascular disease, diabetes, and obesity, is often related to detrimental structural changes of major functional systems during fetal life and persists through the adult offspring’s life [3].
Dietary intake of naturally occurring antioxidants has been linked to disease prevention and health benefits [4,5,6,7]. Indeed, natural products, such as curcumin, resveratrol, and epigallocatechin-3-gallate, have been shown to exhibit a plethora of biological functions that eventually provide prevention or protection against several pathological conditions, including cancer, diabetes, obesity, cardiovascular, and neurodegenerative [8,9,10,11,12]. Among the above-mentioned natural phytonutrients, resveratrol (3,5,4′-trihydroxy-trans-stilbene), an antioxidant polyphenolic compound found in various plants and foods including grapes, berries, peanuts, apples, cacao, and soybeans, has been proven to possess a wide spectrum of biological properties including antioxidant, anti-inflammatory, neuroprotective, cardioprotective, immunomodulatory, antiplatelet, and anticancer [13,14,15,16,17,18]. In this regard, a great body of literature evidence also suggested that maternal dietary intake of antioxidants polyphenols is linked to reduced risk of maternal obesity [19] and cardio-metabolic disorders [20], positively impacting both the fetus and offspring.
This literature review will gather and discuss the current experimental evidence highlighting the effect/s of resveratrol on inflammation, oxidative stress, and other molecular and physiological phenomena associated with pregnancy and pregnancy-associated complications, including preeclampsia, gestational diabetes, and preterm labor. The effect of resveratrol on prenatal issues and pregnancy-associated structures such as the placenta and fetus will also be discussed. Finally, conclusions on the current knowledge and future directions on potentially exploiting resveratrol as a therapeutic tool in pregnancy-associated conditions are provided to the readers.

2. Bioavailability, Toxicity, and Hormetic Effect of Resveratrol

Due to its numerous pharmacological activities, resveratrol’s effects on humans are widely studied. Nonetheless, results from the available human clinical trials are so far contradictory concerning the protective effects of resveratrol against diseases and their sequelae [3]. Similarly, resveratrol intake effects during human pregnancy and the interpretation of its supplementation’s effectiveness are poorly understood due to variations in species, differences in the characteristics of the enrolled patients, dosage, administration routes, duration of resveratrol, supplementation and in some cases, pregnancy models complication [3]. Although resveratrol’s high absorbance and rapid metabolization without pronounced toxicity characterized its beneficial effects in several pregnant women studies [21,22,23], few reports raised doubts about possible toxicity and adverse effects following its consumption. Hence, the adequate ranges of dosage capable of inducing remarkable long-term health benefits without raising toxicity issues remain controversial [3]. Indeed, resveratrol dosage ranges differently in vitro experimental models (micromolar range in cell culture media) and in vivo experimental models (nanomolar range in the blood); hence, it is difficult to precise the optimal concentrations for a maximum beneficial effect of resveratrol in human subjects. However, concerning this aspect, studies in vivo indicate that resveratrol can reach concentration levels similar to those found to exert biological effects in vitro. Indeed, resveratrol can accumulate at relatively high concentrations (up to 32 µM) in plasma [24,25], while concentrations of resveratrol around (~10–30 µM) have been detected in other tissues like the heart, liver, and kidney [26,27]. Such a resveratrol characteristic may be of particular relevance, given its ability to cross the placental barrier and directly interact with the fetus [28]. Another challenging issue that may affect resveratrol therapeutic effects making their interpretation difficult, is its poor oral bioavailability [8]. Resveratrol’s rapid elimination from the body complicates the maintenance of the bloodstream’s acceptable therapeutic levels [29]; moreover, resveratrol is metabolized to phenolic metabolites in the liver and intestinal epithelial cells [30,31,32], and these phase II metabolites can be transformed into the aglycone forming a systemic reservoir of biologically active molecules [8]. In this light, improving resveratrol oral bioavailability by establishing new formulations that are more bioavailable and less degradable with clear pharmacokinetic routes is a crucial future research goal [33].
In human clinical trials, resveratrol was proven to be generally tolerated, yet some adverse effects, including nephrotoxicity and gastrointestinal problems, were reported [34,35]. For instance, a resveratrol dose of 450 mg/day was shown to be safe for a 60 kg individual [36]. In contrast, doses of 1000 mg/day or higher were reported to inhibit cytochrome P450 isoenzymes such as CYP3A4, CYP2C9, and CYP2D6, while activating CYP1A2 leading to interference with the pharmacokinetics of concomitantly administered drugs [37]. Moreover, adverse effects linked to resveratrol intake have been reported on different parameters, including the metabolic status of type 2 diabetes patients and the endothelial health, inflammation, and cardiovascular markers of human patients [38]. Concerning this specific topic, a comprehensive analysis of resveratrol’s adverse effects has been reported elsewhere [8]. On pregnancy, a study conducted on a Japanese macaque experimental model indicated that pregnancy resveratrol intake might cause abnormalities in the fetus, an adverse effect clearly associated with high resveratrol supplementation. The findings of this study showed that resveratrol intake (0.2, 0.5, and 1.0 mg in 5 mL administered via intravenous infusion) managed to reduce the maternal weight, improve glucose tolerance, increase uterine artery volume blood flow, and decrease placental inflammation and liver triglyceride deposition. However, resveratrol intake parallelly affected the fetal pancreatic mass (enlarged by 42%), demonstrating that pregnancy resveratrol supplementation delivers beneficial effects on maternal, placental phenotype, and fetal liver but also an unexplained and concerning alteration of the fetal pancreatic development [39]. In this regard, Cottart et al. [21] highlighted that, despite the extensive amount of data concerning the benefits derived from resveratrol intake, there is scarce research aimed at assessing its harmful effects, and human clinical studies in this regard are limited [21].
Both in vitro and in vivo resveratrol’s biological effects are associated with its hermetic behavior, where low resveratrol doses induce beneficial effects while higher doses usually have a harmful effect [40]. Evidence suggests that such a phenomenon is linked to the biphasic resveratrol effect on the cellular redox state, antioxidant at low doses and pro-oxidant at high ones [15,40,41]. As such, many of the controversial results in the literature might be due to this hormetic aspect, besides other elements related to enrolled patients, doses, and duration of resveratrol supplementation. To summarize, face to the numerous human and animal studies that support the beneficial and protective properties of resveratrol, clinical studies that report on resveratrol’s harmful effects are few and controverted. Additionally, mechanisms underlying resveratrol’s molecular action need more investigation, walling for a more uniform design of clinical trials to properly investigate resveratrol’s therapeutic and preventive properties during pregnancy and related complications.

3. Resveratrol’s Mechanism of Action during Pregnancy Complications

Interestingly, maternal resveratrol intake has been reported to have potential beneficial effects in adverse human pregnancies [3] and has been intensively studied in rodent models as a potential therapeutic agent in several pregnancy-related disorders, including pre-eclampsia (PE) [42], gestational diabetes mellitus (GDM) [22], fetal-growth restriction (FGR) [43], insulin resistance, and dyslipidemia [23,44]. However, the molecular mechanisms underlying its efficiency are not entirely understood [45]. Studies suggested that resveratrol’s potential protective mechanisms in adverse pregnancy outcomes are related to its pleiotropic properties, including antioxidant, anti-inflammatory, anti-obesogenic, anti-atherosclerotic, and anti-diabetic, which evidently associate its consumption with the prevention of several non-communicable diseases [20,46]. Figure 1 schematizes the potential molecular mechanisms that may underpin the protective effects of resveratrol intake in mothers and their offspring in different models.
A systematic data analysis across different species and dosages indicated that resveratrol consumption could decrease inflammation and oxidative stress in placental and embryonic tissues [3]. Oxidative stress is a phenomenon caused by increased levels of reactive oxygen species (ROS) resulting in disturbance of the cellular redox balance, which causes the impairment of cellular functions and eventually initiates pathological pathways leading to disease development [47,48]. It is now evident that oxidative stress is significantly higher during normal pregnancy compared to non-pregnancy periods in women [49]. In fact, the gestational and prenatal changes of the maternal body, as well as the high oxygen and energy required for fetal development, favor ROS overproduction [50]. These elevated ROS levels appear to be implicated in pregnancy-related disorders such as PE, intrauterine growth restriction (IUGR), and fetal death [51,52]. Thus, evaluating the redox status during pregnancy in order to find adequate strategies to counteract it appears to be an issue of utmost importance.
In this regard, studies in murine models have shown that oxidative stress biomarkers and metabolic dysfunction caused by a low-protein diet in the mother, placenta and offspring could be improved by a resveratrol-rich diet in pregnant individuals [53]. Anti-atherogenic activities, low fetal oxidative stress, and reduced apoptosis were also detected in rodent streptozotocin-induced diabetic models treated with resveratrol [54,55]. Moreover, increased risk of infertility is often linked to advanced maternal age, where the major oxidative stress-associated causative factors encompass decreased oocyte quality, low fertilization rate, poor embryonic development, low pregnancy rate, and a high rate of chromosomal aberrations, telomerase shortening, and apoptosis [56,57]. In this regard, by delaying cells’ aging process and preventing age-related diseases through ameliorating mitochondrial function and reducing ROS generation, several studies have reported that resveratrol supplementation can improve in vitro oocyte maturation and embryonic developmental competence in different species [58,59,60,61]. A study realized in a pregnant-aged mice model demonstrated that resveratrol intake increases blastocyst development, ameliorates pregnancy and implantation rates, and parallelly decreases ROS production with a significant increase in mitochondrial membrane potential, suggesting that resveratrol supplementation may improve pregnancy oxidative stress-mediated outcomes in women with advanced maternal age [62]. In this regard, Liu et al. [63] have also suggested that resveratrol can induce oocyte maturation and subsequent embryonic development in aged mice [63]. Indeed, aging-associated reproductive pathologies are frequently associated with impaired DNA repair, metabolic disorders, genomic instability, telomeric shortening, apoptosis, and mitochondrial dysfunction [58,64,65], which are the molecular target of resveratrol action.
Epigenetic regulation represents another potential mechanism related to the maternal resveratrol intake effects and modulates methylation and acetylation processes affect gene expression [66]. Among the main epigenetic changes exerted by resveratrol in the zygotic pronuclei are the methylation and acetylation of histone H3 on lysine 9 (H3K9). Gestational resveratrol supplementation was demonstrated to induce breast cancer-1 promoter (BRCA-1) hypermethylation and to decrease BRCA-1 expression in the mammary tissue of rat offspring [20]. Another mechanism through which resveratrol’s maternal intake may improve pregnancy outcomes is by relieving oxidative stress-related mitophagy [67,68]. Mitophagy is defined as the selective removal of dysfunctional mitochondria by autophagy, a process aberrantly affected by ROS increase and physiologically restored by oxidative stress balance [69]. Consonantly, a study by Zha et al. [70] demonstrated that resveratrol supplementation in pregnant mice might promote mammary gland proliferation and antioxidant activity through mitophagy activation [70]. Pregnancy-related metabolic disorders, such as GDM, occur when ROS prevents insulin from facilitating cellular glucose uptake, subsequently leading to insulin resistance [71]. Hypertensive disorders of pregnancy comprising chronic, white, mask and gestational hypertension, PE, hemolysis, elevated liver enzymes, and low platelet count were also found to be tightly related to altered pro-inflammatory and oxidative stress signals and be responsible for the increased risk of chronic diseases such as obesity, type 2 diabetes, and cardiovascular diseases in adult life [20,72,73,74].
Resveratrol is also recognized as a Sirtuin 1 (SIRT1) activator. The Sirtuin proteins family has a beneficial impact on longevity mainly linked to their effects on metabolic control since they are substantially contributing to lipid and glucose regulation via the deacetylation of crucial metabolic signals relevant to the activated.
The Sirtuin proteins family has a beneficial impact on longevity primarily related to their effects on metabolic control as they substantially contribute to lipid and glucose regulation through the deacetylation of crucial metabolic signals related to the activated protein kinase-SIRT1-PPARG coactivator-1α axis [75,76]. In this context, a study realized on rats exposed to a maternal high-fat diet during pregnancy, resveratrol supplementation (10 mg/kg) managed to restore the impaired expression of SIRT1, phosphor-extracellular regulated kinases1/2, (p-ERK1/2) and phospho-peroxisome proliferator-activated receptor γ (PPARγ), adiponectin and Brain-Derived Neurotrophic Factor (BDNF), all molecules involved in insulin resistance and mild cognitive dysfunction [77]. These previous data underline resveratrol’s ability to improve many maternal-associated complications including PE, FGR, diabetes, insulin resistance (IR), obesity, and metabolic syndrome [77].

4. Maternal Pregnancy-Related Disorders

4.1. Resveratrol Effect in Pre-Eclampsia and Related Disorders

Pregnancy is known to cause increased levels of oxidative stress, as an aspect of the systemic inflammatory response, resulting in high-circulated amounts of ROS, with the placenta representing the main source [78]. PE is a pregnancy-specific clinical syndrome characterized by a multifactorial physiopathology [79,80], that includes both new-onset of hypertension and new-onset proteinuria ≥300 mg/24 h after 20 weeks of gestation, most often near-term. However, when proteinuria registers normal levels, PE is considered thrombocytopenia-related hypertension, with altered liver function, renal insufficiency, pulmonary oedema, and new-onset cerebral or visual disturbances [81]. One condition that features oxidative stress during pregnancy is hypoxia, a common form of intrauterine stress defined as an abnormal exposure to low oxygen levels, resulting in an excessive generation of ROS [82]. Accordingly, increased placental hypoxia and uterine vascular resistance were revealed to be associated with PE and fetal/intrauterine growth restriction (FGR/IUGR) in several animal and human studies [83]. Gestational hypoxia induces ROS overproduction in uteroplacental cells’ mitochondria leading to oxidative stress [83]. In contrast, excessive ROS production causes uteroplacental dysfunction by altering cellular macromolecules, which underlies PE and FGR pathogenesis [83]. Resveratrol has been recently shown to exert a protective effect against prenatal hypoxia-induced programming of the metabolic syndrome when offspring-initiated treatment at weaning [84]. In a recent study, resveratrol intake (4 g/kg) managed to ameliorate adverse fetal outcomes in a rat model of severe prenatal hypoxia [28].
PE-associate oxidative stress originates from the placental impairment caused by hypoxia-reoxygenation imbalance, lower efficiency of both free radical scavengers and antioxidant enzymes, impaired angiogenesis, decreased nitric oxide (NO) bioavailability vascular endothelial dysfunction, cardiovascular complications, and exaggerated inflammatory response [85,86,87,88,89]. A poorly perfused fetoplacental unit releases free radicals and initiates oxidative stress impairment in placental cells [88,90,91]. Resveratrol was demonstrated to activate endothelial nitric oxide synthase (eNOS) and increase NO and nuclear factor-erythroid-derived 2-related factor-2 (Nrf2) production [92]. In response to resveratrol, Nrf2 binds to the antioxidant response element (ARE) promoter upregulating the expression of antioxidant proteins, including heme oxygenase-1 (HO-1) and glutathione reductase (GSR), thus counteracting oxidative stress and balancing the cellular redox state [93,94]. Indeed, in an in vitro PE model, resveratrol increased ARE activity and reduced ROS in endothelial cells exposed to plasma from PE patients compared to cells exposed to plasma from healthy pregnancies. Noteworthy, plasma from PE patients obtained 1 hr after the ingestion of polyphenol-rich whole red grapefruit juice (rich in resveratrol) significantly increased NO production and reduced antioxidant markers in the exposed cells compared with the serum before juice intake [94].
PE etiology is not fully understood, however, this condition is believed to be associated with impaired uterine artery blood flow [95]. In this context, recent studies have shown that resveratrol can induce uterine arteries relaxation during PE pathological events. Interestingly, resveratrol supplementation with a dose of 4 g/kg was demonstrated to ameliorate the PE-like phenotype by significantly increasing uterine artery blood flow velocity and fetal weight in pregnant C57BL/6J, eNOS⁻/⁻, and COMT⁻/⁻ mice murine models, suggesting resveratrol consumption as a potential therapeutic strategy for PE disorder [95].
It has been recently shown that circulating SIRT1 is reduced during PE [96]. In this regard, a group of researchers investigated the effects of resveratrol as a SIRT1 activator using the previously reported in vitro/clinical PE model compared to gestational hypertensive (GH) and healthy pregnant (HP) women. The authors showed that resveratrol-elicited SIRT1 activation alone did not exert any effect and may not be beneficial to women with PE, suggesting that pregnant women with PE may have different responsive mechanisms to this molecule [96]. Furthermore, aberrant trophoblast invasion is among the factors involved in PE occurrence and development since it contributes to the progression of multiple conditions and is characterized by impaired spiral artery remodeling [97], cell-matrix restructuring, and cytoskeleton dynamics [98], during which epithelial-mesenchymal transition (EMT) occurs leading to the breakdown of cell–cell adhesion, the loss of epithelial phenotypes, and cell depolarization [99,100]. During pregnancy, the acquisition of invasive phenotypes by the extra-villous trophoblast (EVT), was found to be involved in PE pathogenesis and was suggested to be linked to EMT [101]. In this regard, using both in vitro (HTR-8/SVeno cell culture) and in vivo (NG-nitro-l-arginine methyl ester mice model) PE models, Zou et al. [102] suggested that resveratrol (100 µmol/L resveratrol /20 mg/kg/day) might stimulate the invasive capability of human trophoblasts by promoting a Wnt/β-catenin pathway-mediated EMT transformation, increasing trophoblasts migration and the invasion in the cell model, and markedly ameliorating hypertension and proteinuria in mice [102]. Molecularly, resveratrol-mediated EMT activation occurred through the regulation of E-cadherin, β-catenin, N-cadherin, and vimentin expression, and altered the WNT-related gene expression, including WNT1, WNT3, and WNT5B [102].
Several studies suggested that the resveratrol-associated anti-hypertensive effect is mainly related to its capacity to attenuate blood pressure and hypertension symptoms. Interestingly, a clinical trial with 400 PE patients investigated the outcome of a nifedipine/resveratrol combined treatment against PE, revealing that resveratrol supplementation significantly reduced the time needed to control blood pressure with a better result in the group treated with both nifedipine (10 mg, up to 5 dosages) and resveratrol, which also had a significantly delayed time between the hypertensive crises intercourse (50 mg, up to 5 dosages) [23]. In contrast, resveratrol did not produce any apparent effect on rats in the study by Moraloglu et al. [42], where a Desoxycorticosterone acetate (DOCA) hypertension PE model was used to test the potential hypotensive effect of resveratrol on the blood flow. Interestingly, DOCA managed to increase blood pressure and placental and renal blood flow levels, whereas, no significant differences for the same parameters were rated in groups treated with resveratrol (20 mg/kg per day) [42].
Consistent with this, other studies indicated that resveratrol anti-hypertensive properties are also related to its ability to inhibit the release of endothelial dysfunction-associated anti-angiogenic factors and that their elevation is a main player in placental oxidative stress and inflammation during the maternal endothelial dysfunction in PE [103]. For instance, serum soluble fms-like tyrosine kinase-1 (sFlt-1), also known as a soluble receptor for vascular endothelial factors (VEGF), is a protein that binds and decreases the concentrations of circulating VEGF and placental growth factor (PlGF) [104,105]. While Endoglin (Eng) is a transmembrane glycoprotein and is considered another factor playing a role in PE etiology and an accessory receptor for the transforming growth factor-beta (TGF-β). Eng is highly expressed in syncytiotrophoblast and proliferating endothelial cells [106], it influences the TGF-β and eNOS signaling pathways resulting in a significant angiogenesis modulation [107]. In this regard, a study carried out in vitro by Hannan et al. [108] demonstrated that resveratrol (0–100 μM) decreases sFlt-1 and soluble Eng secretion from primary trophoblasts and human umbilical vein endothelial cells (HUVECs), a phenomenon that positively correlated with increased mRNA expression of the pro-inflammatory molecules NFκB, IL-6, and IL-1β in the trophoblast and decreased of IL-6, IL-1β, and TNF-α [108]. Additionally, resveratrol significantly increased the mRNA expression of several antioxidant enzymes including HO-1, NADPH Quinone Dehydrogenase (1NQO1), Glutamate-Cysteine Ligase Catalytic (GCLC) subunit and thioredoxin (TXN) in HUVECs, while reducing HO-1 protein levels in trophoblast cells. Expression of the cell adhesion molecule VCAM-1 and the adhesion of peripheral blood mononuclear cells were also increased by resveratrol supplementation [108]. Further, TNF-α-induced endothelial dysfunction in HUVECs was significantly ameliorated by resveratrol through i) the reduction in TNF-α-induced Endothelin-1 (a vasoconstrictor) expression and ii) the increase in endothelial eNOS phosphorylation [108]. Resveratrol (20 mg/kg/day) has also been reported to prevent cells from p53- and ROS-dependent apoptosis induced by IL-1β in an NG-Nitro-l-arginine methyl ester-induced PE rats model via the increase in superoxide dismutase (SOD) suggesting that resveratrol significantly opposes oxidative stress effects in vivo [109]. Overall, resveratrol can be suggested to alleviate PE symptoms and decrease its effects in pregnant individuals and their offspring in different models via multiple pathways mainly related to the attenuation of vascular injury, placental dysfunction, hypertension symptoms, anti-angiogenic factors, oxidative stress and inflammatory responses Table 1 Taken together, the reported data suggests resveratrol as a potential therapeutic tool in PE.

4.2. Resveratrol Effect in Gestational Diabetes and Related Metabolic Disorders

GDM is a type of diabetes characterized by an impaired glucose tolerance that occurs during pregnancy [110]. Although GDM is a reversible pathology and the impaired metabolism returns to normal after delivery, the risk of developing type 2 diabetes later is high [111]. GDM is extremely harmful to mothers and their offspring and may induce PE, premature rupture of membranes, and premature delivery [112]. The main features of GDM during pregnancy include increased glucose demand, increased insulin resistance, and relative insufficiency in insulin secretion [113]. Recent studies emphasized the protective effects of resveratrol in diabetes and its related-cardiovascular complications, involving the regulation of multiple signaling pathways, inhibition of oxidative stress and inflammation, enhancement of insulin sensitivity, induction of autophagy, regulation of lipid metabolism, promotion of GLUT4 expression, translocation, and activation of SIRT1/AMPK signaling axis [114]. Several studies have demonstrated that resveratrol lowers maternal blood sugar levels, ameliorates the maternal lipid profile [115], and prevents delays in embryo development in rat diabetic dams models [54]. In a recent study performed in streptozotocin GDM pregnant rat model, the ability of resveratrol to lower blood glucose and blood lipids was tested. Resveratrol supplementation demonstrated a dose-dependent effect (120 and 240 mg/kg), which positively correlates with a decrease in both insulin and blood glucose levels in comparison with the control group; remarkedly demonstrating a stronger effect than metformin hydrochloride in improving GDM outcomes [116]. Using the same previous model, it was reported that the resveratrol-zinc oxide complex encapsulated with chitosan (CS-ZnO-RS) (an encapsulated form of resveratrol to increase the stability and effectiveness of substances in gestational diabetes management), significantly decreased the blood glucose levels and maintained the lipid content similar to control group levels, while simultaneously reducing the level of pro-inflammatory factors (IL-6 and MCP-1) and significantly decreasing endoplasmic reticulum stress components (GRP78, p-IRE1α, p-eIF2α, and p-PERK), and further inhibiting α-glucosidase and α-amylase activity in a dose-dependent fashion [117].
Studies also demonstrated that GDM is often related to adverse metabolic health outcomes in offspring as a common pregnancy complication. In a GDM model of female Sprague Dawley rats fed with a high-fat and sucrose diet, Brawerman et al. [118] showed that maternal resveratrol supplementation (147 mg/kg/day) protects against GDM-induced glucose intolerance and offspring islet dysfunction by restoring glucose tolerance and normoglycemia and improving insulin secretion. At 15 weeks of age, hepatic steatosis, IR, glucose intolerance, and dysregulated gluconeogenesis were attenuated in offspring of resveratrol-treated dams [118]. Moreover, the dysregulation of several metabolic genes (e.g., lpl: lipoprotein lipase; ppara: peroxisome proliferator-activated receptor α; g6p: glucose-6-phosphatase) were also attenuated whilst glucose-stimulated insulin secretion was improved in the offspring islets of resveratrol-treated dams [118].
It is well known that insulin resistance (IR) caused by insufficient insulin secretion during pregnancy, may lead to metabolic disorders, mainly GDM and related pathologies [119]. Attention was recently given to the role of resveratrol in ameliorating IR in pregnancy-related metabolic disorders. Consistently, recent studies have shown that resveratrol may improve glucose uptake in adipocytes with IR, ameliorate IR in mice fed with a high-fat diet, and enhances glucose metabolism and insulin tolerance in GDM mice (100 mg/kg) [120,121]. MicroRNAs (miRNAs), a type of short-chain non-coding endogenous RNAs existing in eukaryotic organisms, are known to exert regulatory effects on several cell functions and play a different role in many diseases [122,123,124,125,126]. Recent studies confirmed the resveratrol’s capacity to regulate their expression [16], especially that of MiR-23a-3p, which was reported to be low-expressed in adipose tissues of obese and diabetic patients; in fact, its over-expression can reduce TNF-α-induced IR in adipocytes [127]. In a study realized on a high-fat diet GDM mice model, resveratrol supplementation (0.2%) managed to ameliorate glucose uptake and lipid metabolism, regulating the miR-23a-3p/NOV axis through the increase in Adiponectin, Leptin, Phosphoinositide 3-kinase (p-PI3K), and phosphorylated Akt (p-Akt) in adipocytes with IR and parallelly decreasing the nephroblastoma (NOV) overexpression [128]. In diabetic embryopathy-murine models, resveratrol (50 mg, up to 5 dosages) significantly ameliorates the embryonic outcome in terms of diminishing developmental abnormalities, a phenomenon likely associated with its antioxidative potential, anti-diabetic action, and anti-dyslipidemic nature [23].
Maternal obesity is also a metabolic complication that rates high maternal and fetal oxidative stress and inflammation. Hypothetically, offspring of obese mothers are at greater risk of developing obesity, diabetes, and cardiovascular disorders in adult life [129], with risk factors that include variations in both maternal glucose and lipid metabolism (predisposition to GDM in particular), abnormal pregnancy hormone concentrations [130,131,132], stillbirth, premature birth, and macrosomia [132,133].
A recent study conducted in obesity murine models revealed that maternal resveratrol supplementation (20 mg/kg/day) improves maternal metabolism and reduces the placental and liver oxidative stress of mothers and fetuses in a sex-dependent manner [134]. Compared to the untreated group, resveratrol supplementation in pregnant animals was able to lower adipocytes number, triglycerides serum concentrations, insulin resistance, liver fat accumulation, expression of genes related to insulin resistance, inflammatory processes, and lowered oxidative stress in mothers, placentas, and female fetal liver [134]. Resveratrol treatment (5 mg/kg/day) was shown to improve some of the altered metabolic symptoms in a programmed prenatal and postnatal high-fat exposure in the progeny of Sprague Dawley dams’ experimental model, including peripheral leptin resistance, and related dysbiosis of the gut [135,136]. Another study suggested that resveratrol’s protective effects during pregnancy and lactation are diet dependent, where resveratrol supplementation (2.0 to 2.5 mg/kg/d/dam) decreased body weight and adipose tissue content in offspring of dams on a high-fat diet but did not affect offspring from the low-fat diet-fed dams [137]. In addition, resveratrol supplementation (300 mg/kg) was demonstrated to increase high-density lipoprotein cholesterol and low-density lipoprotein cholesterol in the plasma and partially improved the fat metabolism in the adipose tissue in piglets’ experimental model [138]. Intrahepatic cholestasis of pregnancy (ICP), is a pregnancy-specific liver disease characterized by raised serum bile acids and adverse fetal outcomes partially related to SIRT1 dysregulation [139]. Liao et al. [139] studied the molecular and biochemical mechanism underpinning resveratrol’s regulation of the SIRT1- nuclear factor-κB (SIRT1-NF-κB) signaling pathway and bile acid biosynthesis in ICP. Resveratrol (30–120 mg/kg/day) was demonstrated to decrease bile acid levels in the ICP rat model and was suggested to protect syncytiotrophoblast against trichloroanisole (TCA)-induced inflammatory injury through the upregulation of SIRT1 and the downregulation of RelA/p65, a subunit of NF-κB recognized as an activator of SIRT1 [139]. In a related study, it was shown that resveratrol ameliorates ICP conditions by downregulating the overexpression of matrix metalloproteinases (MMPs) [140]. Indeed, in the ethinylestradiol-induced ICP rat model, resveratrol-diet (15 mg/kg/day) was able to inhibit the elevation of both MMP-2 and MMP-9, and exhibited better outcomes in restoring bile flow rate, serum enzymatic activities, and total bile acids (TBA) concentration compared to the ICP known drug ursodeoxycholic acid [140].
On the other hand, diabetes-associated lipid profile disruption may indirectly affect embryogenesis and organogenesis [141], and the impairment of the couple glucose levels/lipid status may simultaneously alter the fetal and neonatal growth; which may later impact offspring life and contribute to adult obesity. Using the same previous GDM model, Singh et al. [22] evaluated the effect of resveratrol on lipidic profile variations. Compared with the untreated GDM group, resveratrol supplementation demonstrated a dose-dependent effect (120 and 240 mg/kg), which significantly increased both HDL and adiponectin levels and inversely correlated with the levels of leptin, resistin, TNF-α, IL-6 levels, the body weight, total cholesterol (TC), triglycerides (TG), and low-density lipoprotein (LDL) [22]. Parallelly, resveratrol was shown to reduce mRNA levels of 3-hydroxy-3-methylglutaryl-CoA reductase (HMG-CoA reductase), the rate-limiting enzyme of cholesterol synthesis and the statin drugs target in high-fat fed hamster model [142]. Statin is an active drug that is used to lower human cholesterol and triglycerides but may be harmful to pregnant or nursing women [143]; therefore, in light of these findings, the authors suggested resveratrol as a potential therapy for an altered lipidic profile during pregnancy-associated metabolic disorders.
Overall, the above-mentioned studies evaluated the short-term effects of maternal resveratrol intake in diabetic and related-metabolic disorders experimental models Table 2. Resveratrol showed favorable results on metabolic homeostasis and redox state. Nevertheless, the mother and offspring’s longer-term outcomes remain poorly specified. Not to mention the variations of experimental models, range of doses, routes of administration and therapeutic periods that may contribute to inconclusive results regarding the potential metabolic benefits of maternal resveratrol supplementation during pregnancy and lactation. In this light, extended studies with well-established experimental models are needed.

5. Fetus-Related Abnormalities and Resveratrol

5.1. Prenatal and Resveratrol

Several studies supported resveratrol’s protective and therapeutic effects at the prenatal stage. In fact, offspring may show increased susceptibility to certain diseases due to prenatal or intrauterine conditions, which may be efficiently reversed by resveratrol. In this section, we present several examples of disease susceptibility due to intrauterine modifications and the potential role of resveratrol in reversing this susceptibility. These include susceptibility to metabolic, cardiovascular, and neurological disorders.
Non-alcoholic fatty liver disease (NAFLD) is a hepatic metabolic disorder [145] associated with IUGR [146]. A study showed that prenatal ethanol exposure (PEE) increases the susceptibility to NAFLD in female IUGR rat offspring by inducing intrauterine metabolic alterations [147]. These alterations enhance fetal hepatic lipogenesis and reduce lipid output in utero; moreover, the fetuses of PEE pregnant rats showed decreased weight and low levels of serum glucose and triglycerides, in addition to hepatocellular ultra-structure modifications [147]. Further analyses also showed attenuated SIRT1 expression and activity associated with the upregulation of SREBP1c and other downstream lipogenic genes, such as fatty acid synthase (FASN), acetyl-coenzyme A carboxylase α (ACCα), and stearyl-coenzyme A desaturase 1 (SCD1) [147]. Interestingly, the hepatic SIRT1-SREBP1c signaling pathway has been reported to play a crucial role in controlling hepatic lipid metabolism and thus promoting liver protection against NAFLD [148]. Accordingly, by targeting the hepatic SIRT1-SREBP1c pathway, the SIRT1 chemical activator resveratrol (50 mg/kg/day) was able to reduce the increased susceptibility to diet-induced NAFLD by reversing the intrauterine programming of hepatic lipogenesis after birth in PEE offspring rats [149]. Another study showed that a maternal high-fat (HF) diet affects the offspring of Sprague Dawley and may lead to metabolic dysregulation [136]. Results showed that offspring dams with maternal HF exposure have decreased acetate propionate and butyrate levels in plasma. In addition, the gut microbiota metagenome of these offspring was altered, thus altering their metabolic homeostasis. The study also suggested that this metabolic dysregulation in offspring was programmed intrauterine by the maternal HF diets and is related to their gut microbiota [136]. In the same study, resveratrol treatment (10 mg/kg/day) was able to reverse the effects induced by maternal HF diet exposure and ameliorate plasma propionate levels. In addition, resveratrol relieved metabolic syndrome dysregulation and related dysbiosis of gut microbiota [136].
Offspring susceptibility to hypertension may originate at the prenatal stage due to maternal conditions. A study showed that the HF diet exacerbated maternal L-NAME treatment-induced programmed hypertension of rat offspring [150]. This was associated with increased oxidative stress, attenuated AMP-activated protein kinase (AMPK)/ peroxisome proliferator-activated receptor γ co-activator 1α (PGC-1α) pathway and altered gut microbiota [150]. These alterations were initiated at the prenatal stage by induced hypertensive maternal condition, sustained till adulthood, and exacerbated by the HF diet. Resveratrol (50 mg/L in drinking water) reversed the increase in Firmicutes to Bacteroidetes ratio in the gut microbiota species while amplifying the abundance of phylum Verrucomicrobia and genus Akkermansia [150]. The overall data indicate resveratrol may attenuate hypertension of developmental origin by regulating the microbiota.
It is accepted that prenatal hypoxia increases offspring susceptibility to cardiovascular disease and metabolic disorders [151,152] such as altered cardiac morphology and dysfunction and metabolic complications in adulthood [153,154,155]. Furthermore, prenatal exposure to hypoxia was reported to set the stage for ischemia/reperfusion (I/R) injury [156,157] and metabolic syndrome in response to a secondary insult such as the HF diet [84]. Interestingly, prenatal hypoxia is able to compromise metabolic and cardiac function in a sex-specific manner [158]. Accordingly, prenatal hypoxia and the HF diet impaired metabolic function in the male but not in female offspring rat [158]. In addition, prenatal hypoxia compromised I/R injury recovery more profoundly in the male compared to female HF diet rat offspring [158]. These prenatally-programmed effects were alleviated by resveratrol administration. Indeed, resveratrol (4 g/kg diet) improved cardiac recovery and attenuated oxidative stress in both male and female rat offspring, thus, impeding their increased susceptibility to cardiovascular diseases [84]. In addition, resveratrol (D120020402 4 g kg−1; Research Diet) improved their metabolic function prenatally challenged by hypoxia and postnatally by HF diet [158]. Molecularly, resveratrol enhanced cardiovascular and metabolic function by mitigating oxidative stress and activating the AMP-activated protein kinase (AMPK)–acetyl CoA carboxylase (ACC) and AMPK/perioxysome proliferator-activated receptor-γ coactivator (PGC)-1α pathways, resulting in improved fatty acid oxidation, mitochondrial biogenesis, and metabolic and cardiac health [158].
Prenatal stress may predispose neurological complications in offspring. In a study realized on rat offspring, prenatal gestational stress increased lipid peroxidation and protein oxidation and decreased antioxidant activity and NO levels [159]. In addition, their dentate gyrus and CA3 hippocampal neurons were severely affected [159]. Resveratrol administration (10 mg/kg) alleviated prenatal stress-induced oxidative stress and the recovering neurons of the dentate gyrus [159]. Thus, resveratrol antioxidant potential is suggested to dispose of a neuroprotective role against prenatal stress-induced oxidative damage in neonatal rat brains [159]. In another study conducted by Sahu et al. [160], resveratrol prenatal-intake effect against cognitive deficit in rat offspring was investigated. Prenatal stress exposure managed to deteriorated spatial learning and memory and reduced Na(+), K(+)-ATPase activity in the rats’ offspring brains, while resveratrol administration (10 mg/kg) did not affect ATPase levels and it improved their spatial learning and memory, suggesting a neuroprotective efficacy of resveratrol against prenatal stress-induced cognitive impairment [160].
Autism spectrum disorder (ASD) is a neurological disorder characterized by decreased social communication and interaction [161]. While little is known about ASD causes, recent evidence supported its association with estrogen receptor β (ERβ) dysregulation [162,163]. In fact, maternal hormonal exposure to ERβ inhibitors such as natural progesterone and synthetic progestin impairs cognitive flexibility and contributes to ASD development [164,165]. A study showed that prenatal exposure of rat offspring to progestins leads to decreased ERβ expression in the amygdala [166]. This downregulation was concomitant with an autism-like behavior in the offspring. These prenatal progestin-induced biochemical and behavioral alterations were shown to be reversed by resveratrol administration (20 mg/kg) [166]. Resveratrol supplementation also reduced progestin-induced oxidative stress, mitochondrial dysfunction, and lipid dysregulation in the brain, through the activation of ERβ and its target genes leading to the amelioration of progestin-induced ASD-like behavior effects [166]. These results are promising for the protective effect of resveratrol against progestin intake, be it clinical such as oral contraceptives, or dietary such as in contaminated water and seafood, suggesting that resveratrol may limit the risk of ASD; however, more clinical studies are needed to determine the resveratrol doses and duration of treatment in pregnant women. Collectively, these data support the protective role of resveratrol against a prenatally-induced predisposition to cardiovascular, neuronal, and metabolic pathological conditions. This protective effect appears to be mainly associated with resveratrol antioxidative properties.

5.2. Placenta and Resveratrol

The placenta is a large organ that develops in the uterus during pregnancy. It provides oxygen and nutrients to the fetus and removes waste products from its blood. It plays a crucial role in fetal organ development and fetal growth [167]. In addition, the umbilical cord of the fetus arises from the placenta [168]. Assessing placenta oxidative status as oxidative stress indication during pregnancy is important due to two main reasons. First, the placenta is the interface between the mother and fetus [167]. Changes in oxygen levels in the placenta to support increased metabolic rate are associated with increased circulating ROS [169]. This redox imbalance in the placenta may have a direct effect on fetal development. The second reason is that the placenta exacerbates pregnancy inflammation by producing inflammatory factors [170,171,172,173]. Thus, it is an important site for the propagation of maternal inflammation.
Noteworthy resveratrol administration during pregnancy ameliorates placental oxidative status, which is achieved through a plethora of biological effects [167,174,175,176]. These resveratrol-initiated effects have been particularly shown to alleviate chemical, microbial, heavy metal, and high-fat diet (HFD)-induced inflammation in the placenta. Consistently with that, resveratrol suppressed cadmium (Cd)-induced expression of inflammatory cytokines and chemokines in the placenta of pregnant CD-1 mice and JEG-3 cells (0.2%) [177]. These include tumor necrosis factor-α (TNF-α), interferon-gamma (IFN-γ), monocyte chemoattractant protein 1 (MCP-1), macrophage inflammatory protein-2 (MIP-2), and chemokine (C–X–C motif) ligand 1 (KC) [177]. In contrast, a study conducted on unchallenged pregnant sows failed to report any influences of dietary resveratrol (300 mg/kg) on pro-inflammatory cytokine levels such as interleukin 1β (IL-1β), IL-6, IL-8 and tumor necrosis factor α (TNF-α) [167]. This discrepancy may be due to the dose variation and duration of resveratrol and stimulus administration (Cd versus no stimulus). It is also worth mentioning that in the latter study, resveratrol played an antioxidative rather than an anti-inflammatory role. In another study realized in the human placenta, resveratrol treatment (200 µM) significantly attenuated IL-1α, IL-1β, IL-6, and IL-8 mRNA expression and reduced the release of IL-6, IL-8, and MCP-1 [173]. Interestingly, these anti-inflammatory effects were observed when the human placenta was stimulated with TNF-α, IL-1β, or the synthetic viral dsRNA analogue polyinosinic: polycytidylic acid (poly (I:C)) [173]. Effectively, several studies support the resveratrol antioxidant effect at a placental level. For example, a study showed that resveratrol treatment (0.2 %) attenuated Cd-induced upregulation of endoplasmic reticulum (ER) stress markers in the placenta of pregnant mice [177]. A similar antioxidant effect of resveratrol (300 mg/kg) was reported in the placenta of pregnant sows [167] where resveratrol was able to increase the expression of Nrf2 and decreased the expression of Kelch-like ECH-associated protein-1 (Keap1) [167]. In addition, resveratrol increased the mRNA expression of antioxidant enzyme genes including catalase (CAT), glutathione peroxidase 1 (GPX1), SOD1 and HO-1. Furthermore, phase 2 detoxification genes, including glutamate-cysteine ligase modifier (GCLM), microsomal glutathione S-transferase 1(MGST1) and UDP glucuronosyltransferase family 1 member A1 (UGT1A1) were upregulated by resveratrol administration (300 mg/kg) [167].
The placenta favors the flow toward the fetus of epigenetic alterations induced by the mother’s conditions, such as GDM and obesity [178,179]. These epigenetic alterations may induce pregnancy complications, affect fetal and neonatal phenotype and induce offspring disease susceptibility [178,179]. In this regard, resveratrol elicits an anti-epigenetic activity by regulating epigenetic enzymes. For instance, resveratrol attenuated both DNA methyltransferase (DNMT) activity and DNMT3B expression in the placenta of Cd-challenged pregnant mice (0.2 %) [177]. In addition, resveratrol administration enhanced the activity and expression of the nuclear deacetylase, Sirtuin 1 (SIRT1), and inhibited Cd-induced PI3K/Akt signaling pathway [177]. A similar activating effect was elicited by resveratrol on the Sirt1 protein in the placenta of pregnant sows (300mg/kg) [167]. Furthermore, resveratrol (100 µmoL/L) has been shown able to prevent the up-regulation of the placental early growth response protein-1 (Egr-1), which is a protein that regulates DNA demethylation [180]. Although maternal nutritional epigenetics is an intriguing topic under intense investigation [181], further studies are necessary to better understand the role of resveratrol in modulating maternal epigenetics.
Studies on non-human primates have also shown that resveratrol reduced placental inflammation and improved glucose metabolism [39,144]. A study by Tran et al. [173] showed that resveratrol (200 µM) restored insulin signaling and glucose uptake aberrantly altered by the pro-inflammatory cytokines TNF-α and IL-1β, the bacterial lipopolysaccharides LPS and polyinosinic-polycytidylic acid (poly(I:C) in human placenta tissue [173]. Not only did resveratrol alleviate placental inflammation, but it also reduced insulin resistance in the human placenta tissue [173]. As previously stated, the release of endothelial dysfunction-associated anti-angiogenic factors such as sFlt-1 is one of the main features in placental oxidative stress and inflammation during PE [103,180]. In humans, resveratrol (100 µmoL/L) abolished cytokine-induced release of sFlt-1 from normal and from pre-eclamptic placental explants [180]. A similar inhibitory effect on sFlt-1 was observed in resveratrol-treated HUVEC or HTR-8/SVneo [180]. In these cells, resveratrol also increased the expression of HO-1, an enzyme found to be dysfunctional in preeclampsia [180]. These latest studies indicate that resveratrol plays anti-inflammatory, antioxidative, and growth factor-regulating roles at the level of the placenta by modulating oxidative/antioxidative enzymes expression, counteracting proinflammatory cytokines release and modulating the expression of growth factors concentration such as VEGF and PlGF through sFlt-1 [104,105].
Overall, the presented data indicate that dietary resveratrol supplementation during pregnancy improves the placental status, which is beneficial for reproductive performance and the offspring’s well-being. However, long-term clinical trials on pregnant women are warranted to reinforce resveratrol’s beneficial effects on the placenta and placenta-associated functions.

5.3. Preterm and Resveratrol

Preterm birth (PTB) (<37 weeks of gestation) is considered a leading cause of newborn death and a risk factor for short and long-term adverse health outcomes. Although the etiology of PTB remains unclear, it is thought that an inappropriate increase in net inflammatory load seems to be a pivotal key [182]. Genital tract infections by Gram-negative bacteria are a common complication in human pregnancy and have been shown to increase the risk of preterm delivery. Several studies demonstrated that bacterial Lipopolysaccharide (LPS) elicits a strong maternal inflammatory response that often results in preterm delivery and fetal death in the murine model endotoxin-induced preterm labor [183]. Such a phenomenon has been reported to occur via the induction of cytokine/chemokine production and infiltration of uterine, placental, and fetal tissues with leukocytes, leading to the release of prostaglandins, endocannabinoids, nitrogen, ROS, and MMPs [184]. Interestingly, prostaglandins, NO and TNF-α components were demonstrated to trigger preterm labor in both animal models [185,186,187] and humans [188]. The endocannabinoid system (ECS) is a cell-signaling system found in multiple organ systems and is integral to sustaining the microenvironment necessary for early pregnancy success and maintenance. A great body of literature suggests that in addition to early pregnancy events, the ECS plays a major role in regulating pregnancy maintenance and labor timing [189]. Through the ECS and PPAR-α systems modulation, resveratrol pre-treatment was found to prevent oxidative stress and inflammation by increasing the tissue levels of palmitoylethanolamide (PEA) and cannabinoid receptors type 1 CB1 and type 2 CB2 in sham animals. In the same model, resveratrol also induces a general increase in PPAR-α and COX-2 protein levels, thus attenuating inflammation and oxidative stress and leading to PTB prevention [190].
Despite the immensity of research into PTB pathophysiology, the resveratrol administration effect on this disorder remains unclear, although few studies suggested that resveratrol decreases PTB rate mainly via the suppression of inflammation and infection complications. Regarding the studies included in this review, only a few investigated the correlation between resveratrol consumption and PTB. In a study realized by Bariani et al. [183], in vivo treatment of 15-day pregnant BALB/c mice with resveratrol (3 mg/kg) prevented the LPS-induced PTB in 64% of the cases, whereas only 15% of mice in the LPS treated group had normal term birth. Resveratrol’s treatment resulted in a reduced NOS activity (p < 0.05) in the uterus of LPS-treated mice and parallelly reduced the expression of LPS-induced pro-inflammatory agents, such as iNOS (p < 0.05), COX-2 (p < 0.05), prostaglandin E2 (PGE2) (p < 0.05), and anandamide (AEA) (p < 0.05). Interestingly, resveratrol administration also prompted ameliorative changes in the LPS-altered uterine endocannabinoid profile [183]. In addition, spontaneous PTB is tightly linked with underlying intrauterine inflammation and infection that evokes an immune response involving the release of cytokines such as IL-1β, TNF-α, [191,192,193,194], cyclooxygenase (COX)-induced PGs [195], and matrix-degrading enzymes [196], which trigger uterine contractions, membrane rupture, and cervical ripening [197], leading to the infant preterm delivery. In Furuya et al.’s research [198], resveratrol administration (20 to 40 mg/kg) significantly decreased the PTB rate in an LPS-induced preterm mouse model and LPS-exposed peritoneal macrophages by suppressing the increased proinflammatory cytokines and consequent elevation of macrophages COX-2 [198]. Moreover, resveratrol uptake abolished TNF-α and interleukin IL-1β activity by downregulating their expression without any apparent effect on IL-6 levels. Parallelly, resveratrol treatment suppressed the elevation of TNF-α and IL-1β levels in LPS-exposed peritoneal macrophages and significantly eradicated the proinflammatory cytokine-mediated elevation of COX-2 in peritoneal macrophages [198].
It is well established that SIRT1 is mainly localized in the nucleus to exert its antiapoptotic effects during oxidative stress or toxic substance-induced damage, and its amount increases in the nucleus to protect the cell from apoptosis. In a tentative to identify the protective role of SIRT1 in PTB and the role of resveratrol to ameliorate its dysregulation, a study realized on PBMCs cells sampled from an established hyperoxia model of premature infants administered with different amounts of oxygen at birth, demonstrated a concentration-dependent increase in SIRT1 translocation rates, ROS and MDA levels. Interestingly, resveratrol (6 µmol/L) treatment managed to attenuate hyperoxia-associated outcomes resulting in opposite effects [199].
Overall, the effects of resveratrol in PTB complications appear to be based on its capacity to counteract infections, inflammation, and oxidative stress-related outcomes. Nevertheless, further investigations including clinical trials are needed to better elucidate the molecular underlying mechanisms of action related to the beneficial effect of resveratrol intake on PTB.

5.4. Fetus and Resveratrol

The “fetal basis of adult disease” hypothesis is now considered a keystone in the development of important diseases in adult/advanced age, such as cardiovascular and metabolic diseases. These events are associated with epigenetic dysregulations, and they are mainly supported by alterations in the fetus development during pregnancy and caused by non-optimal conditions in the uterus, such as GDM, maternal malnutrition, and functional placental deficit [200,201].
Resveratrol pregnancy intake appears to be able to reverse the negative effects on the fetus caused by in-utero adverse conditions and induce a positive impact on adult life health. Although studies have been carried out on different species and with different dosages and routes of administration, the overall data indicate that resveratrol is capable of providing positive effects on fetal development in different gestational phases [3].
Prenatal exposure to environmental stress can also lead to greater susceptibility to clinical disorders in adulthood. This is the case of fetal exposure in utero to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) leading to alterations in the differentiation of T cells in the thymus and to a great susceptibility to autoimmune diseases [202]. TCDD can trigger toxicity through activation of the aryl hydrocarbon receptor and severely affects maternal and fetal immune systems during pregnancy [203]. A study performed in mice by Singh et al. [204] evaluated whether resveratrol administration (100 mg/kg of body weight) inhibited TCDD-induced immunotoxicity during pregnancy in the mother and fetus. Resveratrol was observed to protect not only normal non-pregnant mice, but also pregnant mothers and their fetuses from TCDD-induced thymic atrophy, apoptosis, alterations in T cell receptor, molecule expression, and T cells differentiation stimulatory. Furthermore, Cytochrome P450 Family 1 Subfamily A Member 1 (CYP1A1) expression in the thymus of both mother and fetus is significantly reduced when resveratrol is used in vivo after exposure to TCDD. Taken together the results demonstrate that resveratrol consumption, during pregnancy, is able to protect the mother and the fetus from toxicity induced by environmental pollutants such as TCDD [204].
Mothers exposure to ethanol during pregnancy may also induce various disorders in their offspring, known as fetal alcohol spectrum disorders (FASD), which mainly affect the cerebellum from developing. A study by Kumar et al. [205] demonstrated that resveratrol exhibited neuroprotective and antioxidant effects in the cerebellum of a rat pups model exposed to ethanol by acting on redox regulatory proteins. In particular, resveratrol administration managed to inhibit apoptosis and increase cerebellar granule cell survival. On a molecular level, resveratrol was able to restore ethanol-induced Nrf2 variation levels and parallelly maintained physiological levels of the expression and the activity of its downstream gene targets, such as NADPH quinone oxidoreductase 1 and superoxide dismutase in the same experimental model [205].
Several benefits of resveratrol administration (1 g total trans-resveratrol was implanted subcutaneously into the animal) during pregnancy and fetal development were notable in the study by Darby et al. [206], where they showed its crucial role in increasing uterine artery blood flow in pregnant sheep, leading to increased fetal growth, optimal regulation of blood pressure and oxygenation. The authors also analyzed the direct resveratrol effects on the fetal heart in chronically catheterized pregnant sheep showing that resveratrol induced a significant increase in arterial blood oxygen saturation (SaO2), and fetal arterial oxygen partial pressure (PAO2). Parallelly, resveratrol supplementation did not affect SIRT1 mRNA levels or the activity of the AKT/mTOR or CAMKII signaling pathway in this model [206]. The authors concluded that maternal resveratrol supplementation could increase oxygenation and fetal growth in this animal model that mimics the fetal cardiac development conditions in humans and propose resveratrol as a possible candidate to restore fetal substrate supply in pregnancies with placental insufficiency [206]. Subsequently, the same authors reported the ability of resveratrol to increase body volume and fetal weight without affecting brain weight and volume and to optimize uterine artery blood flow, umbilical vein oxygen saturation, and fetal oxygen release. The authors concluded that increasing fetal weight might be beneficial to ameliorate fetal growth and oxygen supply impairment during pregnancy [207]. Employing a rat model, Bourque et al. [28] demonstrated that resveratrol could improve fetal outcomes associated with prenatal hypoxia, a common complication in pregnancy [28]. Indeed, resveratrol maternal intake during the gestational period (GD7 - GD21) was able to almost completely nullify fetal death in hypoxic pregnancies [28].
Another positive effect of resveratrol on fetal outcomes is its ability to increase fetal hemoglobin levels (HbF) [208]. Among the hemoglobinopathies, beta-thalassemia is characterized by a reduced synthesis of the beta-globin chain, with an unbalanced production of the globin chain, ineffective erythropoiesis, and anemia. This imbalance can be reduced by inducing an increase in gamma-globin gene expression, which combines with excess alpha-globin chains and produces HbF. Other events related to this disease lead to secondary tissue damage due to excess ROS production. [208]. In a study performed in human K562 cell line and erythroid precursors derived from normal donors and patients with beta-thalassemia, Fibach et al. [209] demonstrated that resveratrol (0–100 µM) exhibited antioxidant activity and stimulated the expression of gamma-globin genes leading to HbF accumulation [209]. A later study by Theodorou et al. [210] reported that resveratrol and one of its semi-synthetic derivatives act as (0–100 µM) therapeutic molecules carrying out an antioxidant effect and an inductive effect on HbF synthesis in primary erythroid progenitor cells from healthy donors. It is worth noting that the association of resveratrol and the drug decitabine caused a significant increase in hemoglobin induction activity above the level induced by the stilbene alone [210].
Moreover, a recent work by Bosquesi et al. [211] reported that both resveratrol and its synthetic derivatives (100 µmol/Kg) possess a potential positive effect in the treatment of sickle cell anemia (SCD) symptoms via the induction of gamma-globin. In CD34+ stem cells, resveratrol and one of its semi-synthetic compounds were able to induce twice the production of gamma-globin chains (γG + γA), compared to the vehicle. This semi-synthetic compound emerges as a new molecule in the gamma-globin inducers group. Aside from this property, anti-inflammatory and analgesic activities were also proven for these compounds suggesting them as an alternative in the treatment of SCD symptoms [211].
Resveratrol owns multiple positive health effects on different metabolic parameters, but it also possesses many different negative activities on the cellular life of various tissues/organs in different animal species [212,213,214,215]. In this context, the study [216] evaluated resveratrol’s effects on human fetal adrenocortical steroidogenesis during the 9–12th gestational weeks in vitro using human primary culture of fetal adrenocortical cells (HFAC) from glands of aborted fetuses. The authors showed that resveratrol (10 µM/24 h) significantly suppressed the synthesis of dehydroepiandrosterone, androstenedione, and 11- deoxycortisol in cells stimulated with adrenocorticotropic hormone (ACTH), an event associated with the inhibition of the activities and expression of cytochromes 17α-hydroxylase/17,20 lyase (CYP17) and 21-hydroxylase (CYP21). Based on the data obtained in this model, the authors advise against resveratrol intake by women who are in the early stages of pregnancy.

6. Conclusions and Future Directions

Resveratrol has been reported to possess numerous benefits against complicated pregnancies, with its activity elicited by regulating different physiological and cellular functions and modulating multiple intracellular signaling pathways implicated in pregnancy and pregnancy-associated conditions. Resveratrol demonstrated potent antioxidant activities by blocking DNA damage, regulating antioxidant enzymes, and modulating redox-regulated intracellular signals. Resveratrol also showed anti-inflammatory properties by inhibiting pro-inflammatory signaling pathways and the secretions of pro-inflammatory cytokines and growth factors. Resveratrol has the capability to cross the placenta, improve its oxidative status and directly affect the fetus during the gestational period producing beneficial effects for the reproductive performance and the offspring’s well-being. Resveratrol may offer potential therapeutic benefits for PE due to its antioxidative stress/anti-inflammatory properties that prevent cells from undergoing apoptosis and mediates a protective effect by decreasing blood pressure and attenuating vascular endothelial injury. Interestingly, resveratrol mediates a positive response in blood glucose regulation and attenuates GDM-related complications.
Despite the multiple reported benefits, the use of resveratrol in pregnancy needs to be better investigated to dissolve the high-dose-related toxicity issues highlighted by some papers and the intricate information derived from the available literature analysis. Indeed, although clinical trials indicate resveratrol safety for human consumption, results from the same studies are inconclusive regarding resveratrol’s protective effects against diseases and their complications. In particular, due to the vast difference in experimental models employed, duration of treatments, dosages, routes of administration, and formulations, the final interpretation of resveratrol effects on pregnancy and associated complications remains to be extrapolated.
Particular attention should be directed to performing systematic long-term clinical trials on pregnant women using determined resveratrol dosages to possibly reinforce and ultimately demonstrate resveratrol’s protective and therapeutic role. Specifically, the optimal resveratrol dosages to be employed in humans remains to be clarified. Indeed, ranges of biologically active resveratrol concentrations in vitro and in vivo experimental models differ enormously; hence, based on the currently available data, it is difficult to extrapolate the resveratrol concentration/s that provide the maximum beneficial effect in human subjects without provoking toxicity. This aspect is even more critical considering the resveratrol hormetic properties that tightly associate its final biological outcomes to the employed concentrations. Not to mention the interaction of resveratrol with the cellular and body redox state, which also influences its final effect in being beneficial or harmful. Furthermore, due to its poor bioavailability, as performed for other natural compounds [217,218,219], new resveratrol formulations that provide better molecule absorption and pharmacodynamics need to be developed and commercialized. In this light, many of the controversial resveratrol results in the literature might be due to the aforementioned aspects, which warrant more profound and systematic investigations to reach a final word on resveratrol therapeutic potentials.
Overall, the data presented in our review indicates that resveratrol pregnancy consumption has numerous benefits on metabolic health for pregnant women and their offspring therapeutic approach in pregnancy-related pathological conditions such as elevated inflammation, preeclampsia, and GDM (Figure 2). Therefore, although some adverse effects have been highlighted at high dosages, clinical trials proved resveratrol safe for human consumption, making its recommendation by physicians to pregnant women conceivable. If that ever happens, the final validation of resveratrol therapeutic employment in pregnancy by evidence-based research would have massive consequences both on the mother and the fetus, allowing for early intervention and ensuring healthy mothers and children.

Author Contributions

Conceptualization, R.G. and G.P.; writing—original draft preparation, I.R., A.M.P., R.G., G.F., M.F., R.I., A.H.E. and H.Z.; writing—review and editing, G.P.; visualization, I.R., R.G. and G.P.; supervision, G.P.; project administration, G.P.; funding acquisition, G.P. All authors have read and agreed to the published version of the manuscript.

Funding

This work has been made possible thanks to grants from Progetto Fondazione di Sardegna -bando 2022–2023 and FAR2020-Pintus.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jowell, A.R.; Sarma, A.A.; Gulati, M.; Michos, E.D.; Vaught, A.J.; Natarajan, P.; Powe, C.E.; Honigberg, M.C. Interventions to Mitigate Risk of Cardiovascular Disease After Adverse Pregnancy Outcomes: A Review. JAMA Cardiol. 2022, 7, 346–355. [Google Scholar] [CrossRef]
  2. Parikh, N.I.; Gonzalez, J.M.; Anderson, C.A.M.; Judd, S.E.; Rexrode, K.M.; Hlatky, M.A.; Gunderson, E.P.; Stuart, J.J.; Vaidya, D.; On behalf of the American Heart Association Council on Epidemiology and Prevention; et al. Adverse Pregnancy Outcomes and Cardiovascular Disease Risk: Unique Opportunities for Cardiovascular Disease Prevention in Women: A Scientific Statement From the American Heart Association. Circulation 2021, 143, e902–e916. [Google Scholar] [CrossRef]
  3. Darby, J.R.T.; Dollah, M.H.B.M.; Regnault, T.R.H.; Williams, M.T.; Morrison, J.L. Systematic review: Impact of resveratrol exposure during pregnancy on maternal and fetal outcomes in animal models of human pregnancy complications-Are we ready for the clinic? Pharmacol. Res. 2019, 144, 264–278. [Google Scholar] [CrossRef]
  4. Phu, H.T.; Thuan, D.T.; Nguyen, T.H.; Posadino, A.M.; Eid, A.H.; Pintus, G. Herbal medicine for slowing aging and aging-associated conditions: Efficacy, mechanisms and safety. Curr. Vasc. Pharmacol. 2020, 18, 369–393. [Google Scholar] [CrossRef]
  5. Hossain, R.; Quispe, C.; Khan, R.A.; Saikat, A.S.M.; Ray, P.; Ongalbek, D.; Yeskaliyeva, B.; Jain, D.; Smeriglio, A.; Trombetta, D. Propolis: An update on its chemistry and pharmacological applications. Chin. Med. 2022, 17, 100. [Google Scholar]
  6. Shaito, A.; Thuan, D.T.B.; Phu, H.T.; Nguyen, T.H.D.; Hasan, H.; Halabi, S.; Abdelhady, S.; Nasrallah, G.K.; Eid, A.H.; Pintus, G. Herbal medicine for cardiovascular diseases: Efficacy, mechanisms, and safety. Front. Pharmacol. 2020, 11, 422. [Google Scholar] [CrossRef] [Green Version]
  7. Maaliki, D.; Shaito, A.A.; Pintus, G.; El-Yazbi, A.; Eid, A.H. Flavonoids in hypertension: A brief review of the underlying mechanisms. Curr. Opin. Pharmacol. 2019, 45, 57–65. [Google Scholar] [CrossRef]
  8. Shaito, A.; Posadino, A.M.; Younes, N.; Hasan, H.; Halabi, S.; Alhababi, D.; Al-Mohannadi, A.; Abdel-Rahman, W.M.; Eid, A.H.; Nasrallah, G.K. Potential adverse effects of resveratrol: A literature review. Int. J. Mol. Sci. 2020, 21, 2084. [Google Scholar] [CrossRef] [Green Version]
  9. Giordo, R.; Cossu, A.; Porcu, M.C.; Cappuccinelli, R.; Biosa, G.; Sharifi-Rad, J.; Pretti, L.; Nasrallah, G.K.; Pintus, G.; Posadino, A.M. Cytoprotective, antioxidant, and anti-migratory activity of Pistacia lentiscus L. supercritical carbon dioxide extract on primary human endothelial cells. Nat. Prod. Res. 2022, 1–7. [Google Scholar] [CrossRef]
  10. Ramli, I.; Posadino, A.M.; Zerizer, S.; Spissu, Y.; Barberis, A.; Djeghim, H.; Azara, E.; Bensouici, C.; Kabouche, Z.; Rebbas, K. Low concentrations of Ambrosia maritima L. phenolic extract protect endothelial cells from oxidative cell death induced by H2O2 and sera from Crohn’s disease patients. J. Ethnopharmacol. 2023, 300, 115722. [Google Scholar] [CrossRef]
  11. Posadino, A.M.; Cossu, A.; Giordo, R.; Piscopo, A.; Abdel-Rahman, W.M.; Piga, A.; Pintus, G. Antioxidant properties of olive mill wastewater polyphenolic extracts on human endothelial and vascular smooth muscle cells. Foods 2021, 10, 800. [Google Scholar] [CrossRef] [PubMed]
  12. Posadino, A.M.; Phu, H.T.; Cossu, A.; Giordo, R.; Fois, M.; Thuan, D.T.B.; Piga, A.; Sotgia, S.; Zinellu, A.; Carru, C. Oxidative stress-induced Akt downregulation mediates green tea toxicity towards prostate cancer cells. Toxicol. Vitr. 2017, 42, 255–262. [Google Scholar] [CrossRef] [PubMed]
  13. Giordo, R.; Zinellu, A.; Eid, A.H.; Pintus, G. Therapeutic potential of resveratrol in COVID-19-associated hemostatic disorders. Molecules 2021, 26, 856. [Google Scholar] [CrossRef] [PubMed]
  14. Koushki, M.; Amiri-Dashatan, N.; Ahmadi, N.; Abbaszadeh, H.A.; Rezaei-Tavirani, M. Resveratrol: A miraculous natural compound for diseases treatment. Food Sci. Nutr. 2018, 6, 2473–2490. [Google Scholar] [CrossRef] [Green Version]
  15. Posadino, A.M.; Giordo, R.; Cossu, A.; Nasrallah, G.K.; Shaito, A.; Abou-Saleh, H.; Eid, A.H.; Pintus, G. Flavin oxidase-induced ROS generation modulates PKC biphasic effect of resveratrol on endothelial cell survival. Biomolecules 2019, 9, 209. [Google Scholar] [CrossRef] [Green Version]
  16. Giordo, R.; Wehbe, Z.; Posadino, A.M.; Erre, G.L.; Eid, A.H.; Mangoni, A.A.; Pintus, G. Disease-associated regulation of non-coding RNAs by resveratrol: Molecular insights and therapeutic applications. Front. Cell Dev. Biol. 2022, 10, 894305. [Google Scholar] [CrossRef]
  17. Giordo, R.; Nasrallah, G.K.; Posadino, A.M.; Galimi, F.; Capobianco, G.; Eid, A.H.; Pintus, G. Resveratrol-elicited pkc inhibition counteracts nox-mediated endothelial to mesenchymal transition in human retinal endothelial cells exposed to high glucose. Antioxidants 2021, 10, 224. [Google Scholar] [CrossRef]
  18. Shaito, A.; Al-Mansoob, M.; Ahmad, S.M.; Haider, M.Z.; Eid, A.H.; Posadino, A.M.; Pintus, G.; Giordo, R. Resveratrol-mediated regulation of mitochondria biogenesis-associated pathways in neurodegenerative diseases: Molecular insights and potential therapeutic applications. Curr. Neuropharmacol. 2022. [Google Scholar] [CrossRef]
  19. Zou, T.; Chen, D.; Yang, Q.; Wang, B.; Zhu, M.-J.; Nathanielsz, P.W.; Du, M. Resveratrol supplementation of high-fat diet-fed pregnant mice promotes brown and beige adipocyte development and prevents obesity in male offspring. J. Physiol. 2017, 595, 1547–1562. [Google Scholar] [CrossRef] [Green Version]
  20. Lacerda, D.C.; Costa, P.C.T.; de Oliveira, Y.; de Brito Alves, J.L. The effect of resveratrol in cardio-metabolic disorders during pregnancy and offspring outcomes: A review. J. Dev. Orig. Health Dis. 2022, 14, 3–14. [Google Scholar] [CrossRef]
  21. Cottart, C.-H.; Nivet-Antoine, V.; Laguillier-Morizot, C.; Beaudeux, J.-L. Resveratrol bioavailability and toxicity in humans. Mol. Nutr. Food Res. 2010, 54, 7–16. [Google Scholar] [CrossRef]
  22. Singh, C.K.; Kumar, A.; LaVoie, H.A.; DiPette, D.J.; Singh, U.S. Diabetic complications in pregnancy: Is resveratrol a solution? Exp. Biol. Med. 2013, 238, 482–490. [Google Scholar] [CrossRef] [Green Version]
  23. Ding, J.; Kang, Y.; Fan, Y.; Chen, Q. Efficacy of resveratrol to supplement oral nifedipine treatment in pregnancy-induced preeclampsia. Endocr. Connect. 2017, 6, 595–600. [Google Scholar] [CrossRef] [Green Version]
  24. Böttner, M.; Christoffel, J.; Jarry, H.; Wuttke, W. Effects of long-term treatment with resveratrol and subcutaneous and oral estradiol administration on pituitary function in rats. J. Endocrinol. 2006, 189, 77–88. [Google Scholar] [CrossRef] [Green Version]
  25. Sale, S.; Verschoyle, R.D.; Boocock, D.; Jones, D.; Wilsher, N.; Ruparelia, K.C.; Potter, G.; Farmer, P.B.; Steward, W.P.; Gescher, A.J. Pharmacokinetics in mice and growth-inhibitory properties of the putative cancer chemopreventive agent resveratrol and the synthetic analogue trans 3, 4, 5, 4′-tetramethoxystilbene. Br. J. Cancer 2004, 90, 736–744. [Google Scholar] [CrossRef] [PubMed]
  26. Bertelli, A.; Giovannini, L.; Stradi, R.; Bertelli, A.; Tillement, J. Plasma, urine and tissue levels of trans-and cis-resveratrol (3, 4′, 5-trihydroxystilbene) after short-term or prolonged administration of red wine to rats. Int. J. Tissue React. 1996, 18, 67–71. [Google Scholar]
  27. Vitrac, X.; Desmouliere, A.; Brouillaud, B.; Krisa, S.; Deffieux, G.; Barthe, N.; Rosenbaum, J.; Mérillon, J.-M. Distribution of [14C]-trans-resveratrol, a cancer chemopreventive polyphenol, in mouse tissues after oral administration. Life Sci. 2003, 72, 2219–2233. [Google Scholar] [CrossRef]
  28. Bourque, S.L.; Dolinsky, V.W.; Dyck, J.R.B.; Davidge, S.T. Maternal resveratrol treatment during pregnancy improves adverse fetal outcomes in a rat model of severe hypoxia. Placenta 2012, 33, 449–452. [Google Scholar] [CrossRef] [PubMed]
  29. Park, E.-J.; Pezzuto, J.M. The pharmacology of resveratrol in animals and humans. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2015, 1852, 1071–1113. [Google Scholar] [CrossRef] [Green Version]
  30. Wenzel, E.; Somoza, V. Metabolism and bioavailability of trans-resveratrol. Mol. Nutr. Food Res. 2005, 49, 472–481. [Google Scholar] [CrossRef] [PubMed]
  31. Walle, T. Bioavailability of resveratrol. Ann. N. Y. Acad. Sci. 2011, 1215, 9–15. [Google Scholar] [CrossRef] [PubMed]
  32. Wenzel, E.; Soldo, T.; Erbersdobler, H.; Somoza, V. Bioactivity and metabolism of trans-resveratrol orally administered to Wistar rats. Mol. Nutr. Food Res. 2005, 49, 482–494. [Google Scholar] [CrossRef] [PubMed]
  33. Kemper, C.; Behnam, D.; Brothers, S.; Wahlestedt, C.; Volmar, C.-H.; Bennett, D.; Hayward, M. Safety and pharmacokinetics of a highly bioavailable resveratrol preparation (JOTROL TM). AAPS Open 2022, 8, 11. [Google Scholar] [CrossRef] [PubMed]
  34. Howells, L.M.; Berry, D.P.; Elliott, P.J.; Jacobson, E.W.; Hoffmann, E.; Hegarty, B.; Brown, K.; Steward, W.; Gescher, A.J. Phase I randomized, double-blind pilot study of micronized resveratrol (SRT501) in patients with hepatic metastases—Safety, pharmacokinetics, and pharmacodynamics. Cancer Prev. Res. 2011, 4, 1419–1425. [Google Scholar] [CrossRef] [Green Version]
  35. Poulsen, M.M.; Vestergaard, P.F.; Clasen, B.F.; Radko, Y.; Christensen, L.P.; Stødkilde-Jørgensen, H.; Møller, N.; Jessen, N.; Pedersen, S.B.; Jørgensen, J.O.L. High-dose resveratrol supplementation in obese men: An investigator-initiated, randomized, placebo-controlled clinical trial of substrate metabolism, insulin sensitivity, and body composition. Diabetes 2013, 62, 1186–1195. [Google Scholar] [CrossRef] [Green Version]
  36. Moon, R.T.; Kohn, A.D.; Ferrari, G.V.D.; Kaykas, A. WNT and β-catenin signalling: Diseases and therapies. Nat. Rev. Genet. 2004, 5, 691–701. [Google Scholar] [CrossRef]
  37. Detampel, P.; Beck, M.; Krähenbühl, S.; Huwyler, J. Drug interaction potential of resveratrol. Drug Metab. Rev. 2012, 44, 253–265. [Google Scholar] [CrossRef]
  38. Ramírez-Garza, S.L.; Laveriano-Santos, E.P.; Marhuenda-Muñoz, M.; Storniolo, C.E.; Tresserra-Rimbau, A.; Vallverdú-Queralt, A.; Lamuela-Raventós, R.M. Health effects of resveratrol: Results from human intervention trials. Nutrients 2018, 10, 1892. [Google Scholar] [CrossRef] [Green Version]
  39. Roberts, V.H.; Pound, L.D.; Thorn, S.R.; Gillingham, M.B.; Thornburg, K.L.; Friedman, J.E.; Frias, A.E.; Grove, K.L. Beneficial and cautionary outcomes of resveratrol supplementation in pregnant nonhuman primates. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2014, 28, 2466–2477. [Google Scholar] [CrossRef] [Green Version]
  40. Calabrese, E.J.; Mattson, M.P.; Calabrese, V. Resveratrol commonly displays hormesis: Occurrence and biomedical significance. Hum. Exp. Toxicol. 2010, 29, 980–1015. [Google Scholar] [CrossRef] [PubMed]
  41. Rocha, K.; Souza, G.; Ebaid, G.X.; Seiva, F.; Cataneo, A.; Novelli, E. Resveratrol toxicity: Effects on risk factors for atherosclerosis and hepatic oxidative stress in standard and high-fat diets. Food Chem. Toxicol. 2009, 47, 1362–1367. [Google Scholar] [CrossRef] [PubMed]
  42. Moraloglu, O.; Engin-Ustun, Y.; Tonguç, E.; Var, T.; Tapisiz, Ö.L.; Ergün, H.; Guvenc, T.; Gacar, A. The effect of resveratrol on blood pressure in a rat model of preeclampsia. J. Matern.-Fetal Neonatal Med. 2012, 25, 845–848. [Google Scholar] [CrossRef] [PubMed]
  43. Shah, A.; Quon, A.; Morton, J.S.; Davidge, S.T. Postnatal resveratrol supplementation improves cardiovascular function in male and female intrauterine growth restricted offspring. Physiol. Rep. 2017, 5, e13109. [Google Scholar] [CrossRef] [PubMed]
  44. Malvasi, A.; Kosmas, I.; Mynbaev, O.A.; Sparic, R.; Gustapane, S.; Guido, M.; Tinelli, A. Can trans resveratrol plus d-chiro-inositol and myo-inositol improve maternal metabolic profile in overweight pregnant patients? Clin. Ter. 2017, 168, e240–e247. [Google Scholar] [CrossRef] [PubMed]
  45. Costa-Silva, J.H.; Simões-Alves, A.C.; Fernandes, M.P. Developmental Origins of Cardiometabolic Diseases: Role of the Maternal Diet. Front. Physiol. 2016, 7, 504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. dos Santos, A.S.; de Albuquerque, T.M.R.; de Brito Alves, J.L.; de Souza, E.L. Effects of Quercetin and Resveratrol on in vitro Properties Related to the Functionality of Potentially Probiotic Lactobacillus Strains. Front. Microbiol. 2019, 10, 2229. [Google Scholar] [CrossRef] [Green Version]
  47. Badran, A.; Nasser, S.A.; Mesmar, J.; El-Yazbi, A.F.; Bitto, A.; Fardoun, M.M.; Baydoun, E.; Eid, A.H. Reactive Oxygen Species: Modulators of Phenotypic Switch of Vascular Smooth Muscle Cells. Int. J. Mol. Sci. 2020, 21, 8764. [Google Scholar] [CrossRef] [PubMed]
  48. Shaito, A.; Aramouni, K.; Assaf, R.; Parenti, A.; Orekhov, A.; El Yazbi, A.; Pintus, G.; Eid, A.H. Oxidative Stress-Induced Endothelial Dysfunction in Cardiovascular Diseases. Front. Biosci.-Landmark 2022, 27, 105. [Google Scholar] [CrossRef] [PubMed]
  49. Mihu, D.; Sabau, L.; Costin, N.; Ciortea, R.; Malutan, A.; Mihu, C.M. Implications of maternal systemic oxidative stress in normal pregnancy and in pregnancy complicated by preeclampsia. J. Matern.-Fetal Neonatal Med. 2012, 25, 944–951. [Google Scholar] [CrossRef]
  50. Tan, C.; Wei, H.; Sun, H.; Ao, J.; Long, G.; Jiang, S.; Peng, J. Effects of Dietary Supplementation of Oregano Essential Oil to Sows on Oxidative Stress Status, Lactation Feed Intake of Sows, and Piglet Performance. BioMed Res. Int. 2015, 2015, 525218. [Google Scholar] [CrossRef] [Green Version]
  51. Pereira, A.C.; Martel, F. Oxidative stress in pregnancy and fertility pathologies. Cell Biol. Toxicol. 2014, 30, 301–312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Sanchez-Aranguren, L.C.; Prada, C.E.; Riano-Medina, C.E.; Lopez, M. Endothelial dysfunction and preeclampsia: Role of oxidative stress. Front. Physiol. 2014, 5, 372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Vega, C.C.; Reyes-Castro, L.A.; Rodríguez-González, G.L.; Bautista, C.J.; Vázquez-Martínez, M.; Larrea, F.; Chamorro-Cevallos, G.A.; Nathanielsz, P.W.; Zambrano, E. Resveratrol partially prevents oxidative stress and metabolic dysfunction in pregnant rats fed a low protein diet and their offspring. J. Physiol. 2016, 594, 1483–1499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Singh, C.K.; Kumar, A.; Hitchcock, D.B.; Fan, D.; Goodwin, R.; LaVoie, H.A.; Nagarkatti, P.; DiPette, D.J.; Singh, U.S. Resveratrol prevents embryonic oxidative stress and apoptosis associated with diabetic embryopathy and improves glucose and lipid profile of diabetic dam. Mol. Nutr. Food Res. 2011, 55, 1186–1196. [Google Scholar] [CrossRef] [Green Version]
  55. Trejo-González, N.L.; Chirino-Galindo, G.; Palomar-Morales, M. Capacidad antiteratogénica del resveratrol en diabetes inducida por estreptozotocina en ratas. Rev. Peru. Med. Exp. Salud Pública 2015, 32, 457–463. [Google Scholar] [CrossRef] [Green Version]
  56. Wang, L.; Tang, J.; Wang, L.; Tan, F.; Song, H.; Zhou, J.; Li, F. Oxidative stress in oocyte aging and female reproduction. J. Cell. Physiol. 2021, 236, 7966–7983. [Google Scholar] [CrossRef] [PubMed]
  57. Homer, H.A. The role of oocyte quality in explaining “unexplained” infertility. Semin. Reprod. Med. 2020, 38, 021–028. [Google Scholar] [CrossRef] [PubMed]
  58. Wang, T.; Zhang, M.; Jiang, Z.; Seli, E. Mitochondrial dysfunction and ovarian aging. Am. J. Reprod. Immunol. 2017, 77, e12651. [Google Scholar] [CrossRef] [Green Version]
  59. Liang, Q.-X.; Lin, Y.-H.; Zhang, C.-H.; Sun, H.-M.; Zhou, L.; Schatten, H.; Sun, Q.-Y.; Qian, W.-P. Resveratrol increases resistance of mouse oocytes to postovulatory aging in vivo. Aging 2018, 10, 1586–1596. [Google Scholar] [CrossRef] [PubMed]
  60. Takeo, S.; Sato, D.; Kimura, K.; Monji, Y.; Kuwayama, T.; Kawahara-Miki, R.; Iwata, H. Resveratrol Improves the Mitochondrial Function and Fertilization Outcome of Bovine Oocytes. J. Reprod. Dev. 2014, 60, 92–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Liu, M.-J.; Sun, A.-G.; Zhao, S.-G.; Liu, H.; Ma, S.-Y.; Li, M.; Huai, Y.-X.; Zhao, H.; Liu, H.-B. Resveratrol improves in vitro maturation of oocytes in aged mice and humans. Fertil. Steril. 2018, 109, 900–907. [Google Scholar] [CrossRef] [PubMed]
  62. Yoon, J.; Juhn, K.-M.; Jung, E.-H.; Park, H.-J.; Yoon, S.-H.; Ko, Y.; Hur, C.-Y.; Lim, J.-H. Effects of resveratrol, granulocyte-macrophage colony-stimulating factor or dichloroacetic acid in the culture media on embryonic development and pregnancy rates in aged mice. Aging 2020, 12, 2659–2669. [Google Scholar] [CrossRef] [PubMed]
  63. Liu, M.; Yin, Y.; Ye, X.; Zeng, M.; Zhao, Q.; Keefe, D.L.; Liu, L. Resveratrol protects against age-associated infertility in mice. Hum. Reprod. 2013, 28, 707–717. [Google Scholar] [CrossRef] [Green Version]
  64. Igarashi, H.; Takahashi, T.; Abe, H.; Nakano, H.; Nakajima, O.; Nagase, S. Poor embryo development in post-ovulatory in vivo-aged mouse oocytes is associated with mitochondrial dysfunction, but mitochondrial transfer from somatic cells is not sufficient for rejuvenation. Hum. Reprod. 2016, 31, 2331–2338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Li, H.; Chian, R.-C. Mitochondrial Dysfunction and Age-related Oocyte Quality. Reprod. Dev. Med. 2017, 1, 45–54. [Google Scholar] [CrossRef]
  66. Zheng, S.; Feng, Q.; Cheng, J.; Zheng, J. Maternal resveratrol consumption and its programming effects on metabolic health in offspring mechanisms and potential implications. Biosci. Rep. 2018, 38, BSR20171741. [Google Scholar] [CrossRef] [Green Version]
  67. Park, E.-Y.; Park, J.-B. High glucose-induced oxidative stress promotes autophagy through mitochondrial damage in rat notochordal cells. Int. Orthop. 2013, 37, 2507–2514. [Google Scholar] [CrossRef] [Green Version]
  68. Sebori, R.; Kuno, A.; Hosoda, R.; Hayashi, T.; Horio, Y. Resveratrol Decreases Oxidative Stress by Restoring Mitophagy and Improves the Pathophysiology of Dystrophin-Deficient mdx Mice. Oxidative Med. Cell. Longev. 2018, 2018, 9179270. [Google Scholar] [CrossRef] [Green Version]
  69. Ding, W.-X.; Yin, X.-M. Mitophagy: Mechanisms, pathophysiological roles, and analysis. Biol. Chem. 2012, 393, 547–564. [Google Scholar] [CrossRef] [Green Version]
  70. Zha, C.; Xiao, H.; Song, B.; Zheng, C.; Yang, X.; Wang, W.; Wang, L. Resveratrol promotes mammary cell proliferation and antioxidation capacity during pregnancy and lactation in mice. J. Appl. Microbiol. 2021, 130, 450–463. [Google Scholar] [CrossRef]
  71. 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). Oxidative Med. Cell. Longev. 2021, 2021, 5581570. [Google Scholar] [CrossRef] [PubMed]
  72. Rodrigo, R.; González, J.; Paoletto, F. The role of oxidative stress in the pathophysiology of hypertension. Hypertens. Res. 2011, 34, 431–440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Magee, L.A.; Kenny, L.; Ananth Karumanchi, S.; McCarthy, F.; Saito, S.; Hall, D.R.; Warren, C.E.; Adoyi, G.; Mohammed, S.I. TEMPORARY REMOVAL: The hypertensive disorders of pregnancy: ISSHP classification, diagnosis and management recommendations for international practice 2018. Pregnancy Hypertens. 2018, 72, 24–43. [Google Scholar] [CrossRef]
  74. Agarwal, P.; Morriseau, T.S.; Kereliuk, S.M.; Doucette, C.A.; Wicklow, B.A.; Dolinsky, V.W. Maternal obesity, diabetes during pregnancy and epigenetic mechanisms that influence the developmental origins of cardiometabolic disease in the offspring. Crit. Rev. Clin. Lab. Sci. 2018, 55, 71–101. [Google Scholar] [CrossRef] [PubMed]
  75. Yu, J.; Auwerx, J. The Role of Sirtuins in the Control of Metabolic Homeostasis. Ann. N. Y. Acad. Sci. 2009, 1173, E10–E19. [Google Scholar] [CrossRef]
  76. Yu, J.; Auwerx, J. Protein deacetylation by SIRT1: An emerging key post-translational modification in metabolic regulation. Pharmacol. Res. 2010, 62, 35–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Li, S.-W.; Yu, H.-R.; Sheen, J.-M.; Tiao, M.-M.; Tain, Y.-L.; Lin, I.C.; Lin, Y.-J.; Chang, K.-A.; Tsai, C.-C.; Huang, L.-T. A maternal high-fat diet during pregnancy and lactation, in addition to a postnatal high-fat diet, leads to metabolic syndrome with spatial learning and memory deficits: Beneficial effects of resveratrol. Oncotarget 2017, 8, 111998. [Google Scholar] [CrossRef] [Green Version]
  78. Burton, G.J.; Jauniaux, E. Oxidative stress. Best Pract. Res. Clin. Obstet. Gynaecol. 2011, 25, 287–299. [Google Scholar] [CrossRef] [Green Version]
  79. Lambert, G.; Brichant, J.F.; Hartstein, G.; Bonhomme, V.; Dewandre, P.Y. Preeclampsia: An update. Acta Anaesthesiol. Belg. 2014, 65, 137–149. [Google Scholar]
  80. Sibai, B.; Dekker, G.; Kupferminc, M. Pre-eclampsia. Lancet 2005, 365, 785–799. [Google Scholar] [CrossRef]
  81. Pishko, A.M.; Levine, L.D.; Cines, D.B. Thrombocytopenia in pregnancy: Diagnosis and approach to management. Blood Rev. 2020, 40, 100638. [Google Scholar] [CrossRef] [PubMed]
  82. Silvestro, S.; Calcaterra, V.; Pelizzo, G.; Bramanti, P.; Mazzon, E. Prenatal Hypoxia and Placental Oxidative Stress: Insights from Animal Models to Clinical Evidences. Antioxidants 2020, 9, 414. [Google Scholar] [CrossRef] [PubMed]
  83. Hu, X.-Q.; Zhang, L. Hypoxia and Mitochondrial Dysfunction in Pregnancy Complications. Antioxidants 2021, 10, 405. [Google Scholar] [CrossRef] [PubMed]
  84. Dolinsky, V.W.; Rueda-Clausen, C.F.; Morton, J.S.; Davidge, S.T.; Dyck, J.R. Continued postnatal administration of resveratrol prevents diet-induced metabolic syndrome in rat offspring born growth restricted. Diabetes 2011, 60, 2274–2284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Baumwell, S.; Karumanchi, S.A. Pre-Eclampsia: Clinical Manifestations and Molecular Mechanisms. Nephron Clin. Pract. 2007, 106, c72–c81. [Google Scholar] [CrossRef] [PubMed]
  86. Rosser, M.L.; Katz, N.T. Preeclampsia: An Obstetrician’s Perspective. Adv. Chronic Kidney Dis. 2013, 20, 287–296. [Google Scholar] [CrossRef] [PubMed]
  87. Eiland, E.; Nzerue, C.; Faulkner, M. Preeclampsia 2012. J. Pregnancy 2012, 2012, 586578. [Google Scholar] [CrossRef] [Green Version]
  88. Hansson, S.R.; Nääv, Å.; Erlandsson, L. Oxidative stress in preeclampsia and the role of free fetal hemoglobin. Front. Physiol. 2015, 5, 516. [Google Scholar] [CrossRef] [Green Version]
  89. Sagrillo-Fagundes, L.; Laurent, L.; Bienvenue-Pariseault, J.; Vaillancourt, C. In Vitro Induction of Hypoxia/Reoxygenation on Placental Cells: A Suitable Model for Understanding Placental Diseases. In Preeclampsia: Methods and Protocols; Murthi, P., Vaillancourt, C., Eds.; Springer: New York, NY, USA, 2018; pp. 277–283. [Google Scholar]
  90. Aydın, S.; Benian, A.; Madazli, R.; Uludaǧ, S.; Uzun, H.; Kaya, S. Plasma malondialdehyde, superoxide dismutase, sE-selectin, fibronectin, endothelin-1 and nitric oxide levels in women with preeclampsia. Eur. J. Obstet. Gynecol. Reprod. Biol. 2004, 113, 21–25. [Google Scholar] [CrossRef]
  91. Gupta, S.; Agarwal, A.; Sharma, R.K. The Role of Placental Oxidative Stress and Lipid Peroxidation in Preeclampsia. Obstet. Gynecol. Surv. 2005, 60, 807–816. [Google Scholar] [CrossRef] [Green Version]
  92. Parsamanesh, N.; Asghari, A.; Sardari, S.; Tasbandi, A.; Jamialahmadi, T.; Xu, S.; Sahebkar, A. Resveratrol and endothelial function: A literature review. Pharmacol. Res. 2021, 170, 105725. [Google Scholar] [CrossRef] [PubMed]
  93. Mann, G.E.; Niehueser-Saran, J.; Watson, A.; Gao, L.; Ishii, T.; de Winter, P.; Siow, R.C. Nrf2/ARE regulated antioxidant gene expression in endothelial and smooth muscle cells in oxidative stress: Implications for atherosclerosis and preeclampsia. Acta Physiol. Sin.-Chin. Ed. 2007, 59, 117. [Google Scholar]
  94. Caldeira-Dias, M.; Viana-Mattioli, S.; de Souza Rangel Machado, J.; Carlström, M.; de Carvalho Cavalli, R.; Sandrim, V.C. Resveratrol and grape juice: Effects on redox status and nitric oxide production of endothelial cells in in vitro preeclampsia model. Pregnancy Hypertens. 2021, 23, 205–210. [Google Scholar] [CrossRef]
  95. Poudel, R.; Stanley, J.L.; Rueda-Clausen, C.F.; Andersson, I.J.; Sibley, C.P.; Davidge, S.T.; Baker, P.N. Effects of Resveratrol in Pregnancy Using Murine Models with Reduced Blood Supply to the Uterus. PLoS ONE 2013, 8, e64401. [Google Scholar] [CrossRef] [Green Version]
  96. Viana-Mattioli, S.; Cinegaglia, N.; Bertozzi-Matheus, M.; Bueno-Pereira, T.O.; Caldeira-Dias, M.; Cavalli, R.C.; Sandrim, V.C. SIRT1-dependent effects of resveratrol and grape juice in an in vitro model of preeclampsia. Biomed. Pharmacother. 2020, 131, 110659. [Google Scholar] [CrossRef] [PubMed]
  97. Khong, T.Y.; De Wolf, F.; Robertson, W.B.; Brosens, I. Inadequate maternal vascular response to placentation in pregnancies complicated by pre-eclampsia and by small-for-gestational age infants. BJOG Int. J. Obstet. Gynaecol. 1986, 93, 1049–1059. [Google Scholar] [CrossRef]
  98. Labarrere, C.A.; Althabe, O.H. Inadequate maternal vascular response to placentation in pregnancies complicated by preeclampsia and by small-for-gestational-age infants. BJOG Int. J. Obstet. Gynaecol. 1987, 94, 1113–1115. [Google Scholar] [CrossRef] [PubMed]
  99. Friedl, P.; Wolf, K. Tumour-cell invasion and migration: Diversity and escape mechanisms. Nat. Rev. Cancer 2003, 3, 362–374. [Google Scholar] [CrossRef]
  100. Mao, X.-W.; Xiao, J.-Q.; Li, Z.-Y.; Zheng, Y.-C.; Zhang, N. Effects of microRNA-135a on the epithelial–mesenchymal transition, migration and invasion of bladder cancer cells by targeting GSK3β through the Wnt/β-catenin signaling pathway. Exp. Mol. Med. 2018, 50, e429. [Google Scholar] [CrossRef] [Green Version]
  101. Kokkinos, M.I.; Murthi, P.; Wafai, R.; Thompson, E.W.; Newgreen, D.F. Cadherins in the human placenta—Epithelial–mesenchymal transition (EMT) and placental development. Placenta 2010, 31, 747–755. [Google Scholar] [CrossRef]
  102. Zou, Y.; Li, S.; Wu, D.; Xu, Y.; Wang, S.; Jiang, Y.; Liu, F.; Jiang, Z.; Qu, H.; Yu, X.; et al. Resveratrol promotes trophoblast invasion in pre-eclampsia by inducing epithelial-mesenchymal transition. J. Cell. Mol. Med. 2019, 23, 2702–2710. [Google Scholar] [CrossRef] [Green Version]
  103. Liu, Z.; Wang, C.; Pei, J.; Li, M.; Gu, W. SIRT1: A Novel Protective Molecule in Pre-eclampsia. Int. J. Med. Sci. 2022, 19, 993–1002. [Google Scholar] [CrossRef]
  104. Romero, R.; Chaiworapongsa, T. Preeclampsia: A link between trophoblast dysregulation and an antiangiogenic state. J. Clin. Investig. 2013, 123, 2775–2777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. 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]
  106. Venkatesha, S.; Toporsian, M.; Lam, C.; Hanai, J.-i.; Mammoto, T.; Kim, Y.M.; Bdolah, Y.; Lim, K.-H.; Yuan, H.-T.; Libermann, T.A.; et al. Soluble endoglin contributes to the pathogenesis of preeclampsia. Nat. Med. 2006, 12, 642–649. [Google Scholar] [CrossRef] [PubMed]
  107. Romero, R.; Nien, J.K.; Espinoza, J.; Todem, D.; Fu, W.; Chung, H.; Kusanovic, J.P.; Gotsch, F.; Erez, O.; Mazaki-Tovi, S.; et al. A longitudinal study of angiogenic (placental growth factor) and anti-angiogenic (soluble endoglin and soluble vascular endothelial growth factor receptor-1) factors in normal pregnancy and patients destined to develop preeclampsia and deliver a small for gestational age neonate. J. Matern.-Fetal Neonatal Med. 2008, 21, 9–23. [Google Scholar] [CrossRef] [PubMed]
  108. Hannan, N.J.; Brownfoot, F.C.; Cannon, P.; Deo, M.; Beard, S.; Nguyen, T.V.; Palmer, K.R.; Tong, S.; Kaitu’u-Lino, T.u.J. Resveratrol inhibits release of soluble fms-like tyrosine kinase (sFlt-1) and soluble endoglin and improves vascular dysfunction—Implications as a preeclampsia treatment. Sci. Rep. 2017, 7, 1819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Zou, Y.; Zuo, Q.; Huang, S.; Yu, X.; Jiang, Z.; Zou, S.; Fan, M.; Sun, L. Resveratrol Inhibits Trophoblast Apoptosis through Oxidative Stress in Preeclampsia-Model Rats. Molecules 2014, 19, 20570–20579. [Google Scholar] [CrossRef] [Green Version]
  110. Jiao, X.C.; Xiao, M.; Gao, Z.X.; Xie, J.; Liu, Y.; Yin, M.J.; Wu, Y.; Tao, R.X.; Zhu, P. Effects of comorbid gestational diabetes mellitus and depression on glucose metabolism during pregnancy and neonatal morphological outcome. Zhonghua Yu Fang Yi Xue Za Zhi 2020, 54, 968–973. [Google Scholar] [CrossRef]
  111. Kim, J.; Piao, Y.; Pak, Y.K.; Chung, D.; Han, Y.M.; Hong, J.S.; Jun, E.J.; Shim, J.-Y.; Choi, J.; Kim, C.J. Umbilical Cord Mesenchymal Stromal Cells Affected by Gestational Diabetes Mellitus Display Premature Aging and Mitochondrial Dysfunction. Stem Cells Dev. 2014, 24, 575–586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Choi, D.-J.; Yoon, C.-H.; Lee, H.; Ahn, S.-Y.; Oh, K.J.; Park, H.-Y.; Lee, H.Y.; Cho, M.C.; Chung, I.-M.; Shin, M.-S.; et al. The Association of Family History of Premature Cardiovascular Disease or Diabetes Mellitus on the Occurrence of Gestational Hypertensive Disease and Diabetes. PLoS ONE 2016, 11, e0167528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Wang, X.-M.; Gao, Y.; Eriksson, J.G.; Chen, W.; Chong, Y.S.; Tan, K.H.; Zhang, C.; Zhou, L.; Li, L.-J. Metabolic signatures in the conversion from gestational diabetes mellitus to postpartum abnormal glucose metabolism: A pilot study in Asian women. Sci. Rep. 2021, 11, 16435. [Google Scholar] [CrossRef] [PubMed]
  114. Su, M.; Zhao, W.; Xu, S.; Weng, J. Resveratrol in Treating Diabetes and Its Cardiovascular Complications: A Review of Its Mechanisms of Action. Antioxidants 2022, 11, 1085. [Google Scholar] [CrossRef]
  115. Do, G.-M.; Jung, U.J.; Park, H.-J.; Kwon, E.-Y.; Jeon, S.-M.; McGregor, R.A.; Choi, M.-S. Resveratrol ameliorates diabetes-related metabolic changes via activation of AMP-activated protein kinase and its downstream targets in db/db mice. Mol. Nutr. Food Res. 2012, 56, 1282–1291. [Google Scholar] [CrossRef]
  116. Zhang, G.; Wang, X.; Ren, B.; Zhao, Q.; Zhang, F. The Effect of Resveratrol on Blood Glucose and Blood Lipids in Rats with Gestational Diabetes Mellitus. Evid.-Based Complement. Altern. Med. 2021, 2021, 2956795. [Google Scholar] [CrossRef] [PubMed]
  117. Du, S.; Lv, Y.; Li, N.; Huang, X.; Liu, X.; Li, H.; Wang, C.; Jia, Y.-F. Biological investigations on therapeutic effect of chitosan encapsulated nano resveratrol against gestational diabetes mellitus rats induced by streptozotocin. Drug Deliv. 2020, 27, 953–963. [Google Scholar] [CrossRef]
  118. Brawerman, G.M.; Kereliuk, S.M.; Brar, N.; Cole, L.K.; Seshadri, N.; Pereira, T.J.; Xiang, B.; Hunt, K.L.; Fonseca, M.A.; Hatch, G.M.; et al. Maternal resveratrol administration protects against gestational diabetes-induced glucose intolerance and islet dysfunction in the rat offspring. J. Physiol. 2019, 597, 4175–4192. [Google Scholar] [CrossRef]
  119. Chen, S.; Zhao, Z.; Ke, L.; Li, Z.; Li, W.; Zhang, Z.; Zhou, Y.; Feng, X.; Zhu, W. Resveratrol improves glucose uptake in insulin-resistant adipocytes via Sirt1. J. Nutr. Biochem. 2018, 55, 209–218. [Google Scholar] [CrossRef]
  120. Yi, L.; Chen, C.Y.; Jin, X.; Mi, M.T.; Yu, B.; Chang, H.; Ling, W.H.; Zhang, T. Structural requirements of anthocyanins in relation to inhibition of endothelial injury induced by oxidized low-density lipoprotein and correlation with radical scavenging activity. Febs. Lett. 2010, 584, 583–590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Shu, L.; Zhao, H.A.-O.; Huang, W.; Hou, G.; Song, G.; Ma, H. Resveratrol Upregulates mmu-miR-363-3p via the PI3K-Akt Pathway to Improve Insulin Resistance Induced by a High-Fat Diet in Mice. Diabetes Metab. Syndr. Obes. Targets Ther. 2020, 13, 391–403. [Google Scholar] [CrossRef] [Green Version]
  122. Ferrante, M.; Conti, O.G. Environment and Neurodegenerative Diseases: An Update on miRNA Role. MicroRNA 2017, 6, 157–165. [Google Scholar] [CrossRef] [PubMed]
  123. Mishra, S.; Yadav, T.; Rani, V. Exploring miRNA based approaches in cancer diagnostics and therapeutics. Crit. Rev. Oncol./Hematol. 2016, 98, 12–23. [Google Scholar] [CrossRef] [PubMed]
  124. Tiwari, A.; Mukherjee, B.; Dixit, M. MicroRNA Key to Angiogenesis Regulation: MiRNA Biology and Therapy. Curr. Cancer Drug Targets 2018, 18, 266–277. [Google Scholar] [CrossRef] [PubMed]
  125. Giordo, R.; Ahmed, Y.M.; Allam, H.; Abusnana, S.; Pappalardo, L.; Nasrallah, G.K.; Mangoni, A.A.; Pintus, G. EndMT regulation by small RNAs in diabetes-associated fibrotic conditions: Potential link with oxidative stress. Front. Cell Dev. Biol. 2021, 9, 683594. [Google Scholar] [CrossRef]
  126. Wehbe, N.; Nasser, S.A.; Pintus, G.; Badran, A.; Eid, A.H.; Baydoun, E. MicroRNAs in cardiac hypertrophy. Int. J. Mol. Sci. 2019, 20, 4714. [Google Scholar] [CrossRef] [Green Version]
  127. Lozano-Bartolomé, J.; Llauradó, G.; Portero-Otin, M.; Altuna-Coy, A.; Rojo-Martínez, G.; Vendrell, J.; Jorba, R.; Rodríguez-Gallego, E.; Chacón, M.R. Altered Expression of miR-181a-5p and miR-23a-3p Is Associated With Obesity and TNFα-Induced Insulin Resistance. J. Clin. Endocrinol. Metab. 2018, 103, 1447–1458. [Google Scholar] [CrossRef] [Green Version]
  128. Zheng, T.; Chen, H. Resveratrol ameliorates the glucose uptake and lipid metabolism in gestational diabetes mellitus mice and insulin-resistant adipocytes via miR-23a-3p/NOV axis. Mol. Immunol. 2021, 137, 163–173. [Google Scholar] [CrossRef] [PubMed]
  129. Downs, D.S. Obesity in Special Populations: Pregnancy. Prim. Care Clin. Off. Pract. 2016, 43, 109–120. [Google Scholar] [CrossRef] [Green Version]
  130. Reichetzeder, C. Overweight and obesity in pregnancy: Their impact on epigenetics. Eur. J. Clin. Nutr. 2021, 75, 1710–1722. [Google Scholar] [CrossRef]
  131. Snelgrove-Clarke, E.; Macdonald, D.; Helwig, M.; Alsius, A. Women’s experiences of living with obesity during pregnancy, birthing, and postpartum: A qualitative systematic review protocol. JBI Evid. Synth. 2021, 19, 3183–3189. [Google Scholar] [CrossRef]
  132. 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]
  133. Mission, J.F.; Marshall, N.E.; Caughey, A.B. Pregnancy Risks Associated with Obesity. Obstet. Gynecol. Clin. North Am. 2015, 42, 335–353. [Google Scholar] [CrossRef] [PubMed]
  134. Rodríguez-González, G.L.; Vargas-Hernández, L.; Reyes-Castro, L.A.; Ibáñez, C.A.; Bautista, C.J.; Lomas-Soria, C.; Itani, N.; Estrada-Gutierrez, G.; Espejel-Nuñez, A.; Flores-Pliego, A.; et al. Resveratrol Supplementation in Obese Pregnant Rats Improves Maternal Metabolism and Prevents Increased Placental Oxidative Stress. Antioxidants 2022, 11, 1871. [Google Scholar] [CrossRef] [PubMed]
  135. Yu, H.-R.; Sheen, J.-M.; Tiao, M.-M.; Tain, Y.-L.; Chen, C.-C.; Lin, I.C.; Lai, Y.-J.; Tsai, C.-C.; Lin, Y.-J.; Tsai, C.-C.; et al. Resveratrol Treatment Ameliorates Leptin Resistance and Adiposity Programed by the Combined Effect of Maternal and Post-Weaning High-Fat Diet. Mol. Nutr. Food Res. 2019, 63, 1801385. [Google Scholar] [CrossRef]
  136. Huang, Y.-C.; Huang, L.-T.; Sheen, J.-M.; Hou, C.-Y.; Yeh, Y.-T.; Chiang, C.-P.; Lin, I.C.; Tiao, M.-M.; Tsai, C.-C.; Lin, Y.-J.; et al. Resveratrol treatment improves the altered metabolism and related dysbiosis of gut programed by prenatal high-fat diet and postnatal high-fat diet exposure. J. Nutr. Biochem. 2020, 75, 108260. [Google Scholar] [CrossRef]
  137. Ros, P.; Díaz, F.; Freire-Regatillo, A.; Argente-Arizón, P.; Barrios, V.; Argente, J.; Chowen, J.A. Resveratrol Intake During Pregnancy and Lactation Modulates the Early Metabolic Effects of Maternal Nutrition Differently in Male and Female Offspring. Endocrinology 2017, 159, 810–825. [Google Scholar] [CrossRef] [Green Version]
  138. Sun, S.; Meng, Q.; Luo, Z.; Shi, B.; Bi, C.; Shan, A. Effects of dietary resveratrol supplementation during gestation and lactation of sows on milk composition of sows and fat metabolism of sucking piglets. J. Anim. Physiol. Anim. Nutr. 2019, 103, 813–821. [Google Scholar] [CrossRef] [PubMed]
  139. Liao, E.; Li, Z.; Shao, Y. Resveratrol regulates the silent information regulator 1–nuclear factor-κB signaling pathway in intrahepatic cholestasis of pregnancy. Hepatol. Res. 2018, 48, 1031–1044. [Google Scholar] [CrossRef]
  140. Chen, Z.; Hu, L.; Lu, M.; Shen, Z. Resveratrol reduces matrix metalloproteinases and alleviates intrahepatic cholestasis of pregnancy in rats. Can. J. Physiol. Pharmacol. 2015, 94, 402–407. [Google Scholar] [CrossRef]
  141. Herrera, E.; Ortega-Senovilla, H. Disturbances in lipid metabolism in diabetic pregnancy—Are these the cause of the problem? Best Pract. Res. Clin. Endocrinol. Metab. 2010, 24, 515–525. [Google Scholar] [CrossRef]
  142. Cho, I.J.; Ahn, J.Y.; Kim, S.; Choi, M.S.; Ha, T.Y. Resveratrol attenuates the expression of HMG-CoA reductase mRNA in hamsters. Biochem. Biophys. Res. Commun. 2008, 367, 190–194. [Google Scholar] [CrossRef] [PubMed]
  143. Ofori, B.; Rey, E.; Bérard, A. Risk of congenital anomalies in pregnant users of statin drugs. Br. J. Clin. Pharmacol. 2007, 64, 496–509. [Google Scholar] [CrossRef] [PubMed]
  144. Yao, L.; Wan, J.; Li, H.; Ding, J.; Wang, Y.; Wang, X.; Li, M. Resveratrol relieves gestational diabetes mellitus in mice through activating AMPK. Reprod. Biol. Endocrinol. 2015, 13, 118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Bellentani, S. The epidemiology of non-alcoholic fatty liver disease. Liver Int. Off. J. Int. Assoc. Study Liver 2017, 37 (Suppl. S1), 81–84. [Google Scholar] [CrossRef] [Green Version]
  146. Seferovic, M.D.; Goodspeed, D.M.; Chu, D.M.; Krannich, L.A.; Gonzalez-Rodriguez, P.J.; Cox, J.E.; Aagaard, K.M. Heritable IUGR and adult metabolic syndrome are reversible and associated with alterations in the metabolome following dietary supplementation of 1-carbon intermediates. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2015, 29, 2640–2652. [Google Scholar] [CrossRef] [Green Version]
  147. Shen, L.; Liu, Z.; Gong, J.; Zhang, L.; Wang, L.; Magdalou, J.; Chen, L.; Wang, H. Prenatal ethanol exposure programs an increased susceptibility of non-alcoholic fatty liver disease in female adult offspring rats. Toxicol. Appl. Pharmacol. 2014, 274, 263–273. [Google Scholar] [CrossRef]
  148. Ding, R.B.; Bao, J.; Deng, C.X. Emerging roles of SIRT1 in fatty liver diseases. Int. J. Biol. Sci. 2017, 13, 852–867. [Google Scholar] [CrossRef]
  149. Shen, L.; Xie, L.; Chen, L.; Liu, L.; Shi, X.; Wang, X.; Chen, D.; Wang, H.; Quan, S.; Wang, Z. Resveratrol reverses the programmed high-susceptibility to non-alcoholic fatty liver disease by targeting the hepatic SIRT1-SREBP1c pathway in prenatal ethanol-exposed rat offspring. J. Toxicol. Sci. 2021, 46, 413–423. [Google Scholar] [CrossRef]
  150. Chen, H.E.; Lin, Y.J.; Lin, I.C.; Yu, H.R.; Sheen, J.M.; Tsai, C.C.; Huang, L.T.; Tain, Y.L. Resveratrol prevents combined prenatal N(G)-nitro-L-arginine-methyl ester (L-NAME) treatment plus postnatal high-fat diet induced programmed hypertension in adult rat offspring: Interplay between nutrient-sensing signals, oxidative stress and gut microbiota. J. Nutr. Biochem. 2019, 70, 28–37. [Google Scholar] [CrossRef]
  151. Giussani, D.A.; Davidge, S.T. Developmental programming of cardiovascular disease by prenatal hypoxia. J. Dev. Orig. Health Dis 2013, 4, 328–337. [Google Scholar] [CrossRef]
  152. Demicheva, E.; Crispi, F. Long-term follow-up of intrauterine growth restriction: Cardiovascular disorders. Fetal Diagn. Ther. 2014, 36, 143–153. [Google Scholar] [CrossRef]
  153. Barker, D.J. Fetal origins of coronary heart disease. BMJ 1995, 311, 171–174. [Google Scholar] [CrossRef]
  154. Bjarnegard, N.; Morsing, E.; Cinthio, M.; Lanne, T.; Brodszki, J. Cardiovascular function in adulthood following intrauterine growth restriction with abnormal fetal blood flow. Ultrasound Obstet. Gynecol. 2013, 41, 177–184. [Google Scholar] [CrossRef]
  155. Jaquet, D.; Gaboriau, A.; Czernichow, P.; Levy-Marchal, C. Insulin resistance early in adulthood in subjects born with intrauterine growth retardation. J. Clin. Endocrinol. Metab. 2000, 85, 1401–1406. [Google Scholar] [CrossRef]
  156. Rueda-Clausen, C.F.; Morton, J.S.; Davidge, S.T. Effects of hypoxia-induced intrauterine growth restriction on cardiopulmonary structure and function during adulthood. Cardiovasc. Res. 2009, 81, 713–722. [Google Scholar] [CrossRef] [Green Version]
  157. Xue, Q.; Zhang, L. Prenatal hypoxia causes a sex-dependent increase in heart susceptibility to ischemia and reperfusion injury in adult male offspring: Role of protein kinase C epsilon. J. Pharmacol. Exp. Ther. 2009, 330, 624–632. [Google Scholar] [CrossRef] [Green Version]
  158. Shah, A.; Reyes, L.M.; Morton, J.S.; Fung, D.; Schneider, J.; Davidge, S.T. Effect of resveratrol on metabolic and cardiovascular function in male and female adult offspring exposed to prenatal hypoxia and a high-fat diet. J. Physiol. 2016, 594, 1465–1482. [Google Scholar] [CrossRef] [Green Version]
  159. Madhyastha, S.; Sahu, S.S.; Rao, G. Resveratrol for prenatal-stress-induced oxidative damage in growing brain and its consequences on survival of neurons. J. Basic Clin. Physiol. Pharmacol. 2014, 25, 63–72. [Google Scholar] [CrossRef]
  160. Sahu, S.S.; Madhyastha, S.; Rao, G.M. Neuroprotective effect of resveratrol against prenatal stress induced cognitive impairment and possible involvement of Na+, K+-ATPase activity. Pharmacol. Biochem. Behav. 2013, 103, 520–525. [Google Scholar] [CrossRef]
  161. Abrahams, B.S.; Geschwind, D.H. Advances in autism genetics: On the threshold of a new neurobiology. Nat. Rev. Genet. 2008, 9, 341–355. [Google Scholar] [CrossRef] [Green Version]
  162. Strifert, K. An epigenetic basis for autism spectrum disorder risk and oral contraceptive use. Med. Hypotheses 2015, 85, 1006–1011. [Google Scholar] [CrossRef]
  163. Zou, Y.; Lu, Q.; Zheng, D.; Chu, Z.; Liu, Z.; Chen, H.; Ruan, Q.; Ge, X.; Zhang, Z.; Wang, X.; et al. Prenatal levonorgestrel exposure induces autism-like behavior in offspring through ERbeta suppression in the amygdala. Mol. Autism 2017, 8, 46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Willing, J.; Wagner, C.K. Exposure to the Synthetic Progestin, 17alpha-Hydroxyprogesterone Caproate During Development Impairs Cognitive Flexibility in Adulthood. Endocrinology 2016, 157, 77–82. [Google Scholar] [CrossRef]
  165. Aguirre, C.; Jayaraman, A.; Pike, C.; Baudry, M. Progesterone inhibits estrogen-mediated neuroprotection against excitotoxicity by down-regulating estrogen receptor-beta. J. Neurochem. 2010, 115, 1277–1287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Xie, W.; Ge, X.; Li, L.; Yao, A.; Wang, X.; Li, M.; Gong, X.; Chu, Z.; Lu, Z.; Huang, X.; et al. Resveratrol ameliorates prenatal progestin exposure-induced autism-like behavior through ERbeta activation. Mol. Autism 2018, 9, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Meng, Q.; Guo, T.; Li, G.; Sun, S.; He, S.; Cheng, B.; Shi, B.; Shan, A. Dietary resveratrol improves antioxidant status of sows and piglets and regulates antioxidant gene expression in placenta by Keap1-Nrf2 pathway and Sirt1. J. Anim. Sci. Biotechnol. 2018, 9, 34. [Google Scholar] [CrossRef]
  168. Burton, G.J.; Jauniaux, E. What is the placenta? Am. J. Obstet. Gynecol. 2015, 213, S6.e1–S6.e4. [Google Scholar] [CrossRef] [Green Version]
  169. Wu, F.; Tian, F.J.; Lin, Y.; Xu, W.M. Oxidative Stress: Placenta Function and Dysfunction. Am. J. Reprod. Immunol. 2016, 76, 258–271. [Google Scholar] [CrossRef]
  170. Lappas, M. Activation of inflammasomes in adipose tissue of women with gestational diabetes. Mol. Cell. Endocrinol. 2014, 382, 74–83. [Google Scholar] [CrossRef]
  171. Liong, S.; Lappas, M. Activation of AMPK improves inflammation and insulin resistance in adipose tissue and skeletal muscle from pregnant women. J. Physiol. Biochem. 2015, 71, 703–717. [Google Scholar] [CrossRef]
  172. Lappas, M.; Yee, K.; Permezel, M.; Rice, G.E. Release and regulation of leptin, resistin and adiponectin from human placenta, fetal membranes, and maternal adipose tissue and skeletal muscle from normal and gestational diabetes mellitus-complicated pregnancies. J. Endocrinol. 2005, 186, 457–465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Tran, H.T.; Liong, S.; Lim, R.; Barker, G.; Lappas, M. Resveratrol ameliorates the chemical and microbial induction of inflammation and insulin resistance in human placenta, adipose tissue and skeletal muscle. PLoS ONE 2017, 12, e0173373. [Google Scholar] [CrossRef] [PubMed]
  174. Baur, J.A.; Sinclair, D.A. Therapeutic potential of resveratrol: The in vivo evidence. Nat. Rev. Drug Discov. 2006, 5, 493–506. [Google Scholar] [CrossRef]
  175. Bonnefont-Rousselot, D. Resveratrol and Cardiovascular Diseases. Nutrients 2016, 8, 250. [Google Scholar] [CrossRef]
  176. Lykkesfeldt, J.; Svendsen, O. Oxidants and antioxidants in disease: Oxidative stress in farm animals. Vet. J. 2007, 173, 502–511. [Google Scholar] [CrossRef]
  177. Wang, W.; Liu, G.; Jiang, X.; Wu, G. Resveratrol ameliorates toxic effects of cadmium on placental development in mouse placenta and human trophoblast cells. Birth Defects Res. 2021, 113, 1470–1483. [Google Scholar] [CrossRef]
  178. Lizarraga, D.; Garcia-Gasca, A. The Placenta as a Target of Epigenetic Alterations in Women with Gestational Diabetes Mellitus and Potential Implications for the Offspring. Epigenomes 2021, 5, 13. [Google Scholar] [CrossRef]
  179. Şanlı, E.; Kabaran, S. Maternal obesity, maternal overnutrition and fetal programming: Effects of epigenetic mechanisms on the development of metabolic disorders. Curr. Genom. 2019, 20, 419–427. [Google Scholar] [CrossRef]
  180. Cudmore, M.J.; Ramma, W.; Cai, M.; Fujisawa, T.; Ahmad, S.; Al-Ani, B.; Ahmed, A. Resveratrol inhibits the release of soluble fms-like tyrosine kinase (sFlt-1) from human placenta. Am. J. Obstet. Gynecol. 2012, 206, 253.e10–253.e15. [Google Scholar] [CrossRef]
  181. Joshi, R.O.; Chellappan, S.; Kukshal, P. Exploring the role of maternal nutritional epigenetics in congenital heart disease. Curr. Dev. Nutr. 2020, 4, nzaa166. [Google Scholar] [CrossRef]
  182. Samuel, T.M.; Sakwinska, O.; Makinen, K.; Burdge, G.C.; Godfrey, K.M.; Silva-Zolezzi, I. Preterm Birth: A Narrative Review of the Current Evidence on Nutritional and Bioactive Solutions for Risk Reduction. Nutrients 2019, 11, 1811. [Google Scholar] [CrossRef] [Green Version]
  183. Bariani, M.V.; Correa, F.; Leishman, E.; Domínguez Rubio, A.P.; Arias, A.; Stern, A.; Bradshaw, H.B.; Franchi, A.M. Resveratrol protects from lipopolysaccharide-induced inflammation in the uterus and prevents experimental preterm birth. Mol. Hum. Reprod. 2017, 23, 571–581. [Google Scholar] [CrossRef] [Green Version]
  184. Romero, R.; Espinoza, J.; Gonçalves, L.F.; Kusanovic, J.P.; Friel, L.A.; Nien, J.K. Inflammation in preterm and term labour and delivery. Semin. Fetal Neonatal Med. 2006, 11, 317–326. [Google Scholar] [CrossRef]
  185. Cella, M.; Farina, M.G.; Dominguez Rubio, A.P.; Di Girolamo, G.; Ribeiro, M.L.; Franchi, A.M. Dual effect of nitric oxide on uterine prostaglandin synthesis in a murine model of preterm labour. Br. J. Pharmacol. 2010, 161, 844–855. [Google Scholar] [CrossRef] [Green Version]
  186. Burdet, J.; Sacerdoti, F.; Cella, M.; Franchi, A.M.; Ibarra, C. Role of TNF-α in the mechanisms responsible for preterm delivery induced by Stx2 in rats. Br. J. Pharmacol. 2013, 168, 946–953. [Google Scholar] [CrossRef] [Green Version]
  187. Domínguez Rubio, A.P.; Sordelli, M.S.; Salazar, A.I.; Aisemberg, J.; Bariani, M.V.; Cella, M.; Rosenstein, R.E.; Franchi, A.M. Melatonin prevents experimental preterm labor and increases offspring survival. J. Pineal Res. 2014, 56, 154–162. [Google Scholar] [CrossRef]
  188. Törnblom, S.A.; Maul, H.; Klimaviciute, A.; Garfield, R.E.; Byström, B.; Malmström, A.; Ekman-Ordeberg, G. mRNA expression and localization of bNOS, eNOS and iNOS in human cervix at preterm and term labour. Reprod. Biol. Endocrinol. 2005, 3, 33. [Google Scholar] [CrossRef] [Green Version]
  189. Kozakiewicz, M.L.; Grotegut, C.A.; Howlett, A.C. Endocannabinoid System in Pregnancy Maintenance and Labor: A Mini-Review. Front. Endocrinol. 2021, 12, 699951. [Google Scholar] [CrossRef]
  190. Carta, G.; Poddighe, L.; Serra, M.P.; Boi, M.; Melis, T.; Lisai, S.; Murru, E.; Muredda, L.; Collu, M.; Banni, S.; et al. Preventive Effects of Resveratrol on Endocannabinoid System and Synaptic Protein Modifications in Rat Cerebral Cortex Challenged by Bilateral Common Carotid Artery Occlusion and Reperfusion. Int. J. Mol. Sci. 2018, 19, 426. [Google Scholar] [CrossRef] [Green Version]
  191. Denison, R. Pregnancy & perinatal transmission update. WORLD 1998, 89, 6–7. [Google Scholar]
  192. Osman, I.; Young, A.; Ledingham, M.A.; Thomson, A.J.; Jordan, F.; Greer, I.A.; Norman, J.E. Leukocyte density and pro-inflammatory cytokine expression in human fetal membranes, decidua, cervix and myometrium before and during labour at term. Mol. Hum. Reprod. 2003, 9, 41–45. [Google Scholar] [CrossRef] [Green Version]
  193. Sennström, M.B.; Ekman, G.; Westergren-Thorsson, G.; Malmström, A.; Byström, B.; Endrésen, U.; Mlambo, N.; Norman, M.; Ståbi, B.; Brauner, A. Human cervical ripening, an inflammatory process mediated by cytokines. Mol. Hum. Reprod. 2000, 6, 375–381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Young, A.; Thomson, A.J.; Ledingham, M.; Jordan, F.; Greer, I.A.; Norman, J.E. Immunolocalization of Proinflammatory Cytokines in Myometrium, Cervix, and Fetal Membranes During Human Parturition at Term1. Biol. Reprod. 2002, 66, 445–449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Olson, D.M. The role of prostaglandins in the initiation of parturition. Best Pract. Res. Clin. Obstet. Gynaecol. 2003, 17, 717–730. [Google Scholar] [CrossRef] [PubMed]
  196. Xu, P.; Alfaidy, N.; Challis, J.R.G. Expression of Matrix Metalloproteinase (MMP)-2 and MMP-9 in Human Placenta and Fetal Membranes in Relation to Preterm and Term Labor. J. Clin. Endocrinol. Metab. 2002, 87, 1353–1361. [Google Scholar] [CrossRef] [PubMed]
  197. Goldenberg, R.L.; Culhane, J.F.; Iams, J.D.; Romero, R. Epidemiology and causes of preterm birth. Lancet 2008, 371, 75–84. [Google Scholar] [CrossRef]
  198. Furuya, H.; Taguchi, A.; Kawana, K.; Yamashita, A.; Inoue, E.; Yoshida, M.; Nakamura, H.; Fujimoto, A.; Inoue, T.; Sato, M.; et al. Resveratrol Protects Against Pathological Preterm Birth by Suppression of Macrophage-Mediated Inflammation. Reprod. Sci. 2015, 22, 1561–1568. [Google Scholar] [CrossRef]
  199. Yang, X.; Dong, W.-B.; Lei, X.-P.; Li, Q.-P.; Zhang, L.-Y.; Zhang, L.-P. Resveratrol suppresses hyperoxia-induced nucleocytoplasmic shuttling of SIRT1 and ROS production in PBMC from preterm infants in vitro. J. Matern.-Fetal Neonatal Med. 2018, 31, 1142–1150. [Google Scholar] [CrossRef]
  200. Skogen, J.C.; Øverland, S. The fetal origins of adult disease: A narrative review of the epidemiological literature. JRSM Short Rep. 2012, 3, 59. [Google Scholar] [CrossRef] [Green Version]
  201. Heindel, J.J. The fetal basis of adult disease: Role of environmental exposures—Introduction. Birth Defects Res. Part A Clin. Mol. Teratol. 2005, 73, 131–132. [Google Scholar] [CrossRef]
  202. Camacho, I.A.; Nagarkatti, M.; Nagarkatti, P.S. Effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on maternal immune response during pregnancy. Arch. Toxicol. 2004, 78, 290–300. [Google Scholar] [CrossRef] [PubMed]
  203. Marlowe, J.L.; Puga, A. Aryl hydrocarbon receptor, cell cycle regulation, toxicity, and tumorigenesis. J. Cell. Biochem. 2005, 96, 1174–1184. [Google Scholar] [CrossRef] [PubMed]
  204. Singh, N.P.; Singh, U.S.; Nagarkatti, M.; Nagarkatti, P.S. Resveratrol (3,5,4′-trihydroxystilbene) protects pregnant mother and fetus from the immunotoxic effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin. Mol. Nutr. Food Res. 2011, 55, 209–219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Kumar, A.; Singh, C.K.; LaVoie, H.A.; DiPette, D.J.; Singh, U.S. Resveratrol Restores Nrf2 Level and Prevents Ethanol-Induced Toxic Effects in the Cerebellum of a Rodent Model of Fetal Alcohol Spectrum Disorders. Mol. Pharmacol. 2011, 80, 446–457. [Google Scholar] [CrossRef] [Green Version]
  206. Darby, J.R.T.; Saini, B.S.; Soo, J.Y.; Lock, M.C.; Holman, S.L.; Bradshaw, E.L.; McInnes, S.J.P.; Voelcker, N.H.; Macgowan, C.K.; Seed, M.; et al. Subcutaneous maternal resveratrol treatment increases uterine artery blood flow in the pregnant ewe and increases fetal but not cardiac growth. J. Physiol. 2019, 597, 5063–5077. [Google Scholar] [CrossRef]
  207. Aujla, T.; Darby, J.R.T.; Saini, B.S.; Lock, M.C.; Holman, S.L.; Bradshaw, E.L.; Perumal, S.R.; McInnes, S.J.P.; Voelcker, N.H.; Wiese, M.D.; et al. Impact of resveratrol-mediated increase in uterine artery blood flow on fetal haemodynamics, blood pressure and oxygenation in sheep. Exp. Physiol. 2021, 106, 1166–1180. [Google Scholar] [CrossRef]
  208. Mukherjee, M.; Rahaman, M.; Ray, S.K.; Shukla, P.C.; Dolai, T.K.; Chakravorty, N. Revisiting fetal hemoglobin inducers in beta-hemoglobinopathies: A review of natural products, conventional and combinatorial therapies. Mol. Biol. Rep. 2022, 49, 2359–2373. [Google Scholar] [CrossRef]
  209. Fibach, E.; Prus, E.; Bianchi, N.; Zuccato, C.; Breveglieri, G.; Salvatori, F.; Finotti, A.; Lipucci di Paola, M.; Brognara, E.; Lampronti, I.; et al. Resveratrol: Antioxidant activity and induction of fetal hemoglobin in erythroid cells from normal donors and beta-thalassemia patients. Int. J. Mol. Med. 2012, 29, 974–982. [Google Scholar] [CrossRef] [Green Version]
  210. Theodorou, A.; Phylactides, M.; Forti, L.; Cramarossa, M.R.; Spyrou, P.; Gambari, R.; Thein, S.L.; Kleanthous, M. The investigation of resveratrol and analogs as potential inducers of fetal hemoglobin. Blood Cell Mol. Dis. 2016, 58, 6–12. [Google Scholar] [CrossRef] [Green Version]
  211. Bosquesi, P.L.; Melchior, A.C.B.; Pavan, A.R.; Lanaro, C.; de Souza, C.M.; Rusinova, R.; Chelucci, R.C.; Barbieri, K.P.; Fernandes, G.F.D.; Carlos, I.Z.; et al. Synthesis and evaluation of resveratrol derivatives as fetal hemoglobin inducers. Bioorg. Chem. 2020, 100, 103948. [Google Scholar] [CrossRef]
  212. Giordo, R.; Cossu, A.; Pasciu, V.; Hoa, P.T.; Posadino, A.M.; Pintus, G. Different redox response elicited by naturally occurring antioxidants in human endothelial cells. Open Biochem. J. 2013, 7, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Pasciu, V.; Posadino, A.M.; Cossu, A.; Sanna, B.; Tadolini, B.; Gaspa, L.; Marchisio, A.; Dessole, S.; Capobianco, G.; Pintus, G. Akt downregulation by flavin oxidase—Induced ROS generation mediates dose-dependent endothelial cell damage elicited by natural antioxidants. Toxicol. Sci. 2010, 114, 101–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Posadino, A.M.; Cossu, A.; Giordo, R.; Zinellu, A.; Sotgia, S.; Vardeu, A.; Hoa, P.T.; Carru, C.; Pintus, G. Resveratrol alters human endothelial cells redox state and causes mitochondrial-dependent cell death. Food Chem. Toxicol. 2015, 78, 10–16. [Google Scholar] [CrossRef] [PubMed]
  215. Salehi, B.; Mishra, A.P.; Nigam, M.; Sener, B.; Kilic, M.; Sharifi-Rad, M.; Fokou, P.V.T.; Martins, N.; Sharifi-Rad, J. Resveratrol: A double-edged sword in health benefits. Biomedicines 2018, 6, 91. [Google Scholar] [CrossRef] [Green Version]
  216. Savchuk, I.; Morvan, M.L.; Søeborg, T.; Antignac, J.P.; Gemzell-Danielsson, K.; Le Bizec, B.; Söder, O.; Svechnikov, K. Resveratrol inhibits steroidogenesis in human fetal adrenocortical cells at the end of first trimester. Mol. Nutr. Food Res. 2017, 61, 1600522. [Google Scholar] [CrossRef]
  217. Quispe, C.; Cruz-Martins, N.; Manca, M.L.; Manconi, M.; Sytar, O.; Hudz, N.; Shanaida, M.; Kumar, M.; Taheri, Y.; Martorell, M. Nanoderived Therapeutic Formulations with Curcumin in Inflammation-Related Diseases. Oxidative Med. Cell. Longev. 2021, 2021, 3149223. [Google Scholar] [CrossRef]
  218. Sanna, V.; Roggio, A.M.; Posadino, A.M.; Cossu, A.; Marceddu, S.; Mariani, A.; Alzari, V.; Uzzau, S.; Pintus, G.; Sechi, M. Novel docetaxel-loaded nanoparticles based on poly (lactide-co-caprolactone) and poly (lactide-co-glycolide-co-caprolactone) for prostate cancer treatment: Formulation, characterization, and cytotoxicity studies. Nanoscale Res. Lett. 2011, 6, 260. [Google Scholar] [CrossRef] [Green Version]
  219. Sanna, V.; Pintus, G.; Roggio, A.M.; Punzoni, S.; Posadino, A.M.; Arca, A.; Marceddu, S.; Bandiera, P.; Uzzau, S.; Sechi, M. Targeted biocompatible nanoparticles for the delivery of (−)-epigallocatechin 3-gallate to prostate cancer cells. J. Med. Chem. 2011, 54, 1321–1332. [Google Scholar] [CrossRef]
Figure 1. Potential molecular mechanisms underpinning the effects of resveratrol intake in mothers and their offspring in different models.
Figure 1. Potential molecular mechanisms underpinning the effects of resveratrol intake in mothers and their offspring in different models.
Antioxidants 12 00341 g001
Figure 2. Maternal resveratrol supplementation decreases the risk of adverse pregnancy outcomes by ameliorating the in-utero conditions. Schematic representation of the resveratrol-impacted crosstalk between abnormal in-utero conditions and adverse pregnancy risk.
Figure 2. Maternal resveratrol supplementation decreases the risk of adverse pregnancy outcomes by ameliorating the in-utero conditions. Schematic representation of the resveratrol-impacted crosstalk between abnormal in-utero conditions and adverse pregnancy risk.
Antioxidants 12 00341 g002
Table 1. Studies using resveratrol in the treatment of models of pre-eclampsia in pregnancy.
Table 1. Studies using resveratrol in the treatment of models of pre-eclampsia in pregnancy.
ReferenceType of the StudyPE Experimental ModelDoseMechanism of ActionOutcomes of Supplementation in the Experimental Model
[94]In vitro/Clinical trial-ECs
-Human
200 mL of polyphenol rich-grape fruit/1 µM of trans-resveratrol NO, HO-1, miRNA expression, GSH, ARE, ROS levels↑ ARE, NO, HO-1, GSH, ARE
↓ ROS, oxidative stress
No studied effect in offspring
[96]In vitro/Clinical trial-ECs
-Human
200 mL of polyphenol rich-grape fruit/1µM of trans-resveratrolSIRT1 expressionNo significant effect
No studied effect in offspring
[102]In vitro/In vivo-HTR-8/SVeno cell culture/
-NG-nitro-l-arginine methyl ester mice model (L-NAME)
100 µmol/L resveratrol
20 mg/kg/day
Expression of genes regulating migration, invasion, angiogenesis and EMT-related factors in trophoblasts
Hypertension and proteinuria measurement.
Endothelial dysfunction/ injury
↑ E-cadherin, β-catenin, N-cadherin, and vimentin expression,
↓ Alteration of WNT-related gene expression, including WNT1, WNT3 and WNT5B.
↓ Hypertension and proteinuria
No studied effect in offspring
[108]In vitroprimary trophoblasts and HUVECs0–100 μM-Angiogenesis activation
(sFlt-1) (sEng) secretion
Anti-inflammatory effect
-NFκB
, IL-6 and IL-1β
-Antioxidant effect
HO-1, NQO1, GCLC, (TXN)
Endothelial dysfunction, VCAM Endothelin-1 eNOS
↓ sFlt-1, sEng
↓ IL-6, IL-1β and TNF-α.‘
↑, NQO1, GCLC TXN
↓ HO-1 protein in trophoblast.
↑ VCAM-1
↓ Endotelin-1, ↑eNOS
No studied effect in offspring
[109]In vivoNG-nitro-l-arginine methyl ester mice model (L-NAME)20 mg/kg/day-Antioxidant effect, Apoptosis
SOD, MDA
↑ SOD, MDA
No studied effect in offspring
Ecs: Endothelial cells, NO: Nitric oxide, HO-1: Heme oxygenase-1, miRNA: microRNA, GSH: Gluthatione, ARE: antioxidant response element, ROS: Reactive oxygen species, SIRT1: sirtuin1, WNT: WNT genes, sFlt-1: soluble fms-like tyrosine kinase-1, sEng: soluble endoglin, NFκB: nuclear factor-kappa B, IL-6: Interleukin-6, IL-1β: Interleukin-1β, TXN: thioredoxin, NADPH, NQO1: NADPH Quinone Dehydrogenase 1, TNF-α: Tumor necrosis factor-alpha, GCLC: Glutamate-Cysteine Ligase Catalytic Subunit, VCAM-1: Vascular cell adhesion molecule-1, eNOS: endothelial nitric oxide synthase, SOD: Superoxide dismutase, MDA: malondialdehyde.
Table 2. Studies using resveratrol in the treatment of models of gestational diabetes miletus (GDM) in pregnancy.
Table 2. Studies using resveratrol in the treatment of models of gestational diabetes miletus (GDM) in pregnancy.
ReferenceType of the StudyGDM Experimental ModelDoseMechanism of ActionOutcomes of Supplementation in the Experimental Model
[116]In vivoStreptozotocin GDM pregnant rats model120 and 240 mg/kgAmelioration of blood glucose and blood lipids levels↑ Insulin levels,
↓ blood glucose levels
Amelioration of lipidic profile
No studied effect in offspring
[117]In vivoStreptozotocin GDM pregnant rats model500 μg/mL (CS-ZnO-RS)Anti-diabetic effect
Anti-inflammatory effect
↓ blood glucose levels
lipid content reduced the level of
↓ IL-6 and MCP-1, GRP78, p-IRE1α, p-eIF2α, and p-PERK
Inhibition of α-glucosidase and α-amylase
No studied effect in offspring
[22]In vivoStreptozotocin GDM pregnant rats model120 and 240 mg/kgAmelioration of the lipids metabolic profile ↑ HDL-C and adiponectin
↓ leptin, resistin, TNF-α, IL-6 levels, the body weight, TC, TG, and LDL-C
No studied effect in offspring
[142]In vivohigh-fat fed hamster model0.025%Amelioration of the lipids metabolic profile-HMG-CoA reductase expression
No studied effect in offspring
[128]In vivohigh-fat diet GDM mice model
IR adipocyte model was established by dexamethasone-inducing
0.2%Amelioration of glucose and lipids metabolic profile
Amelioration of IR in adipocytes
↓ The bodyweight, serum glucose
↑ serum insulin
Upregulations of miR-23a-3p/NOV axis
↑ Adiponectin, Leptin, p-PI3K, and p-Akt
No studied effect in offspring
[118]In vivofemale Sprague-Dawley rat model, fed with a high-fat and sucrose diet 147 mg/kg/dayProtection against gestational diabetes-induced glucose intolerance and islet dysfunction-Restored glucose tolerance, normoglycaemia and improved insulin secretion in offspring
-Attenuation of hepatic steatosis, insulin resistance, glucose intolerance and dysregulated gluconeogenesis in offspring
-Downregulation of lpl, ppara, g6p genes
[144]In vivoC57BL/KsJ-Lep (db/+) (db/+) genetic GDM pregnant mouse model10 mg/kg/dayAmelioration of glucose metabolic profile via SIRT1/AMPK pathway
Amelioration of IR
↑ Glucose metabolism, insulin tolerance and reproductive outcome of the pregnant db/+ females
AMPK activation
↓ glucose-6-phosphatase in both pregnant db/+ females and their offspring
GDM: Gestational diabetes miletus, CS-ZnO-RS: resveratrol-zinc oxide complex encapsulated with chitosan, IL-6: Interleukin-6, MCP-1: Monocyte Chemoattractant Protein-1, GRP78, p-IRE1α, p-eIF2α, and p-PERK: endoplasmic reticulum stress components, TNF-α: Tumor necrosis factor-alpha, TC: Total cholesterol, TG: Triglycerides, LDL-C: low-density lipoprotein-C, IR: Insulin resistance, miR-23a-3p/NOV axis: microRNA-23a-3p, p-Akt: phosphorylated-AKT, p-PI3K: Phosphoinositide 3-kinase, lpl: lipoprotein lipase gene, ppara: Peroxisome proliferator-activated receptor alpha, g6p: glucose-6 phosphatase. AMPK: AMP-activated protein kinase.
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

Ramli, I.; Posadino, A.M.; Giordo, R.; Fenu, G.; Fardoun, M.; Iratni, R.; Eid, A.H.; Zayed, H.; Pintus, G. Effect of Resveratrol on Pregnancy, Prenatal Complications and Pregnancy-Associated Structure Alterations. Antioxidants 2023, 12, 341. https://doi.org/10.3390/antiox12020341

AMA Style

Ramli I, Posadino AM, Giordo R, Fenu G, Fardoun M, Iratni R, Eid AH, Zayed H, Pintus G. Effect of Resveratrol on Pregnancy, Prenatal Complications and Pregnancy-Associated Structure Alterations. Antioxidants. 2023; 12(2):341. https://doi.org/10.3390/antiox12020341

Chicago/Turabian Style

Ramli, Iman, Anna Maria Posadino, Roberta Giordo, Grazia Fenu, Manal Fardoun, Rabah Iratni, Ali H. Eid, Hatem Zayed, and Gianfranco Pintus. 2023. "Effect of Resveratrol on Pregnancy, Prenatal Complications and Pregnancy-Associated Structure Alterations" Antioxidants 12, no. 2: 341. https://doi.org/10.3390/antiox12020341

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