Open Access

Promising benefit of resveratrol in preventing preterm birth: A systematic review

  • Authors:
    • Muhammad Habiburrahman
    • Muhammad Ilham Dhiya Rakasiwi
    • Afid Brilliana Putra
  • View Affiliations

  • Published online on: January 22, 2024     https://doi.org/10.3892/wasj.2024.225
  • Article Number: 10
  • Copyright : © Habiburrahman et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Prematurity is a matter of utmost concern in pregnancies due to its associated complications, mortality, disability and significant economic burden. However, the current tocolytics used for preventing preterm birth have limitations, such as side‑effects and unclear inhibitory pathway mechanisms. Hence, it is critical to explore alternative tocolytics, such as resveratrol. The present study, by performing a systematic review, aimed to investigate the potential of resveratrol in preventing prematurity and its therapeutic applications. A systematic search of the relevant literature was conducted using advanced search engines (PubMed, ProQuest, EBSCOhost and EMBASE), employing specific key words and predefined eligibility criteria to identify supporting articles, which were subsequently synthesized into the systematic review. Of note, four critical lines of evidence, supported by multiple studies, highlight the promising benefits of resveratrol in preventing preterm birth. In a mouse model of preterm birth, resveratrol has demonstrated the ability to: i) Reduce prematurity rates by 49‑64%; ii) inhibit inflammatory mediators and cytokines (nitric oxide synthase, inducible nitric oxide synthase, cyclooxygenase (COX)‑2 in the uterus, TNF‑α, IL‑1β, COX‑2 in peritoneal macrophages and the cervixs; iii) decrease prostaglandin E2 (PGE2) and prostaglandin F2α (PGF2α) levels in the uterus; and iv) regulate endocannabinoids (anandamide and 2‑arachidonoylglycerol) and endocannabinoid‑like lipids. By reducing the levels of these substances, resveratrol has the potential to prevent cervical ripening, membrane rupture and myometrial contractility associated with preterm birth. In summary, resveratrol offers multiple pathways to inhibit inflammation‑induced prematurity, rendering it a promising candidate as a novel tocolytic agent.

Introduction

Prematurity, defined as live birth before 37 weeks of gestation by the World Health Organization (WHO), poses a global health challenge (1). In 2020, there were 13.4 million cases of preterm birth (PTB), with approximately one million resulting in mortality, as reported by the United Nations (UN). These numbers indicate that 1 in 10 babies worldwide are born prematurely (2). The number of PTBs has not improved over the last decade, with 152 million PTBs occurring from 2010 to 2020. The majority of PTBs have occurred in sub-Saharan Africa and Southern Asia, accounting for >65% of the cases globally (2). Worldwide, there has been a 5.26% reduction in newborns born prematurely, decreasing from 16.06 million in 1990 to 15.22 million in 2019. Furthermore, deaths among these newborns have significantly decreased by 47.71%, decreasing from 1.27 million in 1990 to 0.66 million in 2019(3). Prematurity exposes infants to various health risks, such as neurocognitive deficits, pulmonary issues, heart rate irregularities, eye disorders, cerebral palsy, anemia, neonatal sepsis and intraventricular hemorrhage (4).

PTB not only has significant medical implications, but also poses substantial economic and social challenges for families. In a study conducted in the USA, researchers examined the healthcare costs associated with preterm and low-birth-weight (LBW) infants (5). That study was a retrospective cohort study using a national claims database of individuals covered by Aetna, Inc. during the first 6 months of life. The study included a total of 763,566 infants with a combined healthcare expense of approximately $8.4 billion. Preterm infants (n=50,511) had an average medical cost of $76,153, while LBW infants had an average cost of $114,437. Among infants born at 24 weeks of gestation (n=418), the average cost per infant was the highest at $603,778(5).

The pathophysiology of PTB involves various factors, such as genetics, infection/inflammation, environment, oxidative stress, progesterone resistance and an advanced maternal age (6). Infection-related inflammation is the primary cause, accounting for 40% of cases (7). Experimental mouse models are crucial for studying the mechanisms and potential therapies for prematurity. Maternal inflammation in these models is induced using bacterial lipopolysaccharide (LPS) endotoxin, which triggers the release of cytokines, the infiltration of leukocytes and the production of cyclooxygenase (COX)-induced prostaglandins (PGs), endocannabinoids, reactive oxygen and nitrogen species (RONS) and metalloproteinases (8). This immune response leads to uterine contractions, membrane rupture and cervical ripening (9). Mice with a genetic predisposition to PTB (e.g., Trp53 deficiency) also exhibit inflammation when exposed to LPS. These models provide valuable tools for evaluating novel treatments for the challenges associated with PTB (10,11).

The clinical management of preterm labor is challenging as no single drug effectively addresses all its mechanisms. Current therapeutics involve the use of tocolytics, which delay preterm labor and enable the administration of corticosteroids for neonatal lung maturation (12). Tocolytics include magnesium sulfate, β-mimetics, calcium channel blockers, PG inhibitors and oxytocin receptor antagonists (13). However, these drugs have limitations and potential side-effects, and there is insufficient evidence for significant neonatal benefits (14). Existing tocolytics also have limitations and lack strong evidence for maternal or neonatal outcomes (15,16). Consequently, efforts are underway to discover novel tocolytic agents that are more effective and safer. One such promising candidate is resveratrol.

Resveratrol (3,5,4'-trihydroxy-trans-stilbene) is a natural polyphenol known for its beneficial effects on inducing preterm labor (11,17) and other pregnancy-related complications (18,19). It exhibits anti-inflammatory and anti-aging properties, rendering it a potential therapeutic target for PTB (8,17). However, there is a lack of comprehensive studies on the specific benefits of resveratrol in preventing prematurity. Therefore, the present systematic review aimed to explore the potential of resveratrol in preventing prematurity, including its pharmacological profile, content in various plant species and therapeutic applications.

Data and methods

Protocol

The present descriptive qualitative systematic review followed the Preferred Reporting Items for Systematic Review and Meta-Analyses (PRISMA) statement (20).

Information sources and search strategy

The search, conducted in July-August 2023, aimed to identify studies on the mechanisms of resveratrol in preventing PTB in animal models. The present systematic review followed the PICOS format: Population (P)-Pregnancy with induced PTB; Intervention (I)-Resveratrol; Comparison (C)-Negative control (untreated with resveratrol); Outcome (O)-Prevention of PTB (tocolytic effect); Study Design (S)-Preclinical studies. Specific search strategies were used for various databases, including MEDLINE via PubMed, ProQuest, EBSCOhost and EMBASE. The search queries and MESH keywords combined with Boolean operators were used, such as ‘Resveratrol’ (Mesh) AND [‘Premature Birth’ (Mesh) OR ‘Obstetric Labor, Premature’ (Mesh)].

Study selection and data collection process

The studies were selected based on the eligibility criteria using the PICOS strategy. The inclusion criteria were as follows: i) English-language articles; ii) primary experimental studies utilizing resveratrol (in vitro and in vivo); and iii) outcomes related to PTB. The exclusion criteria encompassed the following: i) Duplicated articles; ii) abstracts and reviews lacking specific data; iii) clinical studies, systematic reviews, case reports, retrospective studies, theses, letters, editorials, opinions, surveys, guidelines, conferences, abstracts and commentary articles. Initially, a search was performed for human population studies; however, as none were found, a focus was placed solely on reviewing experimental animal studies. Articles focusing solely on the benefits of resveratrol for preterm infants after birth were also excluded. In total, the authors (MH, MIDR and ABP) independently screened titles and abstracts, removing duplicates. Eligible articles had their full texts examined for confirmation. No restrictions on publication dates were applied to the systematic review. The search process is illustrated in Fig. 1.

Quality assessment and risk of bias

To assess the quality of the studies, the ToxRTool (Toxicological Data Reliability Assessment Tool), developed by ECVAM (European Centre for the Validation of Alternative Methods) (21) was used. This tool provides criteria for evaluating the quality of pharmacology research using in vitro and in vivo experimental study designs. Previous systematic reviews that utilized the ToxRTool were also considered to determine the methodological reliability and inherent flaws in the studies (22-24). The ToxRTool consists of a 21-point rating checklist that assesses the methodological aspects of each study across five categories (25). These categories cover various aspects, including Category 1 (test substance identification), which includes details about (1.1) the substance, (1.2) purity, (1.3) source, and (1.4) nature and physico-chemical properties; Category 2 (test organism characterization), which includes (2.1) species (for in vivo studies) or test system (for in vitro studies), (2.2) sex (for in vivo studies) or the origin or source of the test system with the corresponding sex characteristic (for in vitro studies), (2.3) strain of test animals (for in vivo studies) or the specification of cell/tissue culture (for in vitro studies), (2.4) age or body weight of test organisms at the start of the study (for in vivo studies) or relevant clinical characteristics of human donors for cell/tissue cultivation (for in vitro studies), and (2.5) housing or feeding conditions in repeated dose studies (for in vivo studies) or cultivation process maintenance (for in vitro studies); Category 3 (study design description), which includes (3.1) administration method, (3.2) administered doses or concentration in the application media, (3.3) exposure frequency, duration, and explanation of observation time-points, (3.4) inclusion of negative and positive controls, (3.5) the number of animals (for in vivo studies) or the amount of cell/tissue culture per group (for in vitro studies), (3.6) administration scheme details for study evaluation, and (3.7) verification of achieved concentrations or substance stability in repeated dose studies; Category 4 (study endpoints), which includes (4.1) clear description and determination methods of endpoints, (4.2) transparent and comprehensive results description for all endpoints, and (4.3) transparent statistical methods for data analysis; and Category 5 (study design appropriateness and reliability), which includes (5.1) appropriate choice of study design for substance-specific data and (5.2) reliability of quantitative results. Studies with <13 points are deemed unreliable, those with 13-17 points are considered reliable with potential restrictions, and those scoring 18-21 points are deemed reliable without restrictions.

In addition, a risk of bias (RoB) assessment tool based on the Cochrane Handbook (26) was used with modifications that integrated the Office of Health Assessment and Translation, National Institute of Health (OHAT-NIH), and the National Toxicological Program tools, as used in previous studies (27-29). Three authors independently assessed all included studies for bias risk across nine parameters (randomization, allocation concealment, experimental conditions, blinding during the study, data completeness, exposure characterization, outcome assessment, selective reporting and data sufficiency). The responses were categorized as low risk (yes, provided information), unclear risk, or high risk (no, absent information) of bias following predefined protocol criteria. Any disagreements, conflicts, or discrepancies regarding the eligibility of studies were resolved through discussion and consensus to determine inclusion.

Data extraction

Data extraction was performed using Microsoft Office Excel 365. Three reviewers independently organized the data into variables based on the review's aims and topics. These variables included publication year, author(s), first author's country, study design, level of evidence (LoE) according to the Oxford Centre of Evidence-based Medicine (CEBM) criteria (30), preterm model sample, sample size, induced diseases and definitions. The outcomes data included the dose of resveratrol and administration details, treatment duration and frequency, additional drugs used, and associated study outcomes.

Synthesis of results

The relevant data of interest regarding the aforementioned variables were collected and organized into summary tables for study characteristics and outcomes using an electronic spreadsheet. The studies were further grouped based on similarities in reducing preterm rates and common underlying mechanisms.

Results

Study selection

The PRISMA flow diagram was followed for allocation concealment, blinding, article review, and data extraction. Initially, 3,597 results were retrieved from all databases, which were reduced to 2,791 after removing duplicates. Screening based on titles and abstracts resulted in 1,103 articles, and after applying inclusion and exclusion criteria, 20 full-text articles were assessed for eligibility. Ultimately, only four articles were eligible for analysis and review. Additionally, searches were conducted on Google Scholar and various Gray Literature websites (http://www.opengrey.eu/search/, https://v2.sherpa.ac.uk/opendoar/, https://www.worldcat.org/, and https://ukhsalibrary.koha-ptfs.co.uk/greylit/). However, no additional results were obtained beyond those included in the initial selection from the central journal databases.

Study exclusion

A full-text assessment was conducted after removing all duplicate references using Zotero version 6.0.26 as the reference manager. The exclusion criteria included studies that focused on the effects of resveratrol on neonates as opposed to maternal conditions, such as retinopathy, lung injury, vascular dysfunction, embryopathy and oxidative stress-related disorders (n=7). Studies that discussed the benefits of resveratrol for cellular aging and senescence in cancer were also excluded (n=6). Additionally, a study that assessed the effects of resveratrol on non-pregnant myometrium was excluded (n=1), as well as narrative reviews (n=2).

Reliability and quality assessment

As regards the reliability assessment (Table I), two studies [Bariani et al (8) and Novaković et al (31)] were deemed fully reliable and were included in the systematic review. However, two other studies [Deng et al (11) and Furuya et al (17)] were labeled as ‘reliable with restrictions’ due to methodological uncertainties and the absence of critical quality measurement categories. Specifically, the study by Deng et al (11) lacked information on the purity and origin of resveratrol, the age and other characteristics of the mice used, the environmental conditions for the mice, positive controls, a precise number of samples, and details on the stabilization and extraction/obtaining of resveratrol. Similarly, the study by Furuya et al (17) had limitations in sample characteristics and conditioning during the experiment, as well as a lack of a positive control and information on the stabilization of resveratrol. Detailed assessments for each category are provided in Table SI, Table SII and Table SIII.

Table I

Reliability assessment of the selected articles according to the ToxRTool in vivo and in vitro criteria.

Table I

Reliability assessment of the selected articles according to the ToxRTool in vivo and in vitro criteria.

 Studies included in the systematic review [Authors/(Refs.), year of publication
Reliability of study assessmentBariani et al (8), 2017Deng et al (11), 2016Furuya et al (17), 2015Novaković et al (31), 2015
Criteria I: Test substance identification (maximum point 4)4244
Criteria II: Test organism characterization (maximum point 5)4334
Criteria III: Study design description (maximum point 7)5455
Criteria IV: Study results documentation (maximum point 3)3333
Criteria V: Plausibility of study design and data (maximum point 2)2122
Total score18131718
Reliability classificationReliable without restrictionsReliable with restrictionsReliable with restrictionsReliable without restrictions

[i] The scoring system was as follows: 18-21, reliable without restrictions, and thus being useful for the intended purpose; 13-17, reliable with restrictions, and thus being potentially useful for the intended purpose; <13 or not all critical criteria met, generally not to be used as a crucial study, but depending on the shortcomings of the study, it may still be helpful in weight-of-evidence approaches or as supportive information.

In the risk of bias summary (Fig. 2), Bariani et al (8) had a low risk of bias in seven out of nine domains in each RoB category, with two aspects rated as high risk. Novaković et al (31) had a low risk of bias in five of nine domains in each RoB category, with one aspect considered unclear risk and the remaining three as high risk. Deng et al (11) and Furuya et al (17) each demonstrated a low risk of bias in three domains, an unclear risk in three other domains, and three domains with high risk. Despite all the limitations in the reliability and quality assessment, all studies were eligible to be assessed and reviewed in the present systematic review. According to the CEBM levels, they all fall under the fifth level of EBM (1 to 5).

Study design and outcomes measurement

The present systematic review included two experimental in vivo studies [Bariani et al (8) and Furuya et al (17)] and one study combining animal and cell experiments [Deng et al (11)]. All these studies investigated the effect of resveratrol on preventing PTB in animal models. Additionally, one in vitro study [Novaković et al 2015(31)] focused on the tocolytic and myometrium relaxant effects of resveratrol using human myometrium tissue to determine the preterm mechanisms. In this section, several studies that examined the preventive effects of resveratrol on PTB are presented. These studies include information about its characteristics (Table II), as well as its beneficial preventive effects on PTB (Table III).

Table II

Characterization of the articles included in the systematic review.

Table II

Characterization of the articles included in the systematic review.

Authors, year of publicationRegionStudy DesignLoESampleSample sizePreterm induction modelDisease induction and operational definition(Refs.)
Bariani et al, 2017ArgentinaIn vivo515-days pregnant BALB/c mice±35-41 miceInflammatory induction: LPS i.p. (0.17 mg/kg and 0.5 mg/kg after 3 h) on day 15• LPS-induced preterm labor • Preterm labor was defined as labor occurring 19 days(8)
Deng et al, 2016USAIn vivo and in vitro5Trp53fl/fl PgrCre/+ mice (p53d/d mice) and floxed p53 mice (p53fl/fl), which are genetically predisposed to PTB • Human uterine fibroblast (HuF) cells, which were decidualized in vitro and placenta obtained from term and pre- term (at 23 weeks) vaginal delivery. • p53fl/fl and p53d/d uterine stromal cells, which were decidualized in vitro obtained from 4-days pregnant mice • Mouse embryonic fibroblasts (MEFs) from Prkaa1-/- Prkaa2-/- double knockout (AMPK dKO), WT, and p53-/ mice• The exact number of mice was not mentioned • Samples from 3 women were used for human cell studiesIn vivo model: conditional uterine deletion of the tumor suppressor gene p53 • Superimposed inflammatory insult: LPS (10 µg) administered 4 h following a progesterone injection (1-2 mg/mouse) on day 16 • In vitro model: Hormone treatment to induce decidualization in cultured uterine stromal cells• Spontaneous PTB in p53d/d females, both with and without dystocia • Preterm labor was defined as labor occurring before 19 days • Dystocia was defined as a problematic delivery lasting >12 h(11)
Furuya et al, 2015JapanIn vivo57-days pregnant C57BL6 mice12 miceInflammatory induction: Transvaginal administration of LPS (10 µg in 200 ml saline) to the cervix and uterus on day 15• LPS-induced preterm labor • Preterm labor was defined as labor occurring before 19 days. • PTB occurred within 48 h(17)
Novaković et al, 2015SerbiaIn vitro5Myometrial samples were obtained from females undergoing an IVF program and currently scheduled for elective C-section (due to cephalopelvic disproportion) in the third trimester of pregnancy. Biopsies were excised from the mid-line portion of the upper lip of the incision in the lower uterine segmentMyometrial tissue samples were collected from 42 non-laboring womenContraction inducer: Oxytocin (20 nM)• Oxytocin-induced phasic contractions of constant amplitude and frequency in human-term pregnant myometrium • This study did not specifically focus on preterm labor, but the outcomes are still relevant in the context of tocolytic research(31)

[i] AE, N-acyl ethanolamine; AMPK, AMP-activated protein kinase; C-section, cesarean section; CPD, cephalopelvic disproportion; HuF, human uterine fibroblast; IVF, in vitro fertilization; LoE, level of evidence; LPS, lipopolysaccharide; MEF, mouse embryonic fibroblasts; mg, milligram; ml, milliliter; nM, nanomolar; PTB, preterm birth; WT, wild type; µg, microgram.

Table III

Summary of the articles analyzed in the systematic review focusing on the effect and potential benefit of resveratrol in preventing preterm birth.

Table III

Summary of the articles analyzed in the systematic review focusing on the effect and potential benefit of resveratrol in preventing preterm birth.

Authors, year of publicationResveratrol dose and routeDuration frequencyAdditional drugOutcomes(Refs.)
Bariani et al, 20173 mg/kg, POOn day 15, per 8 hN/A• Resveratrol reduced the proportion of PTB compared to the control group (64% vs. 85%) • Resveratrol reduced stillbirth compared to the negative control group (stillbirth, 34 vs. 62%) • Resveratrol prevented antepartum hemorrhage based on macroscopic examination. • Resveratrol may protect against pathological PTB by reducing uterine NOS activity, the expression of iNOS, COX-2, PGE2, and AEA profiling (P<0.05) • Resveratrol altered the uterine endocannabinoid profiling that was previously affected by LPS(8)
Deng et al, 201630 mg/kg POOn days 8,10,12 and 14• A single injection of progesterone (1-2 mg/mouse) was administered 4 h before LPS induction • Metformin 1 mg/kg was administered on days 8, 10 and 12 separately (not combined with resveratrol)• The combination of resveratrol + progesterone is an effective approach for targeting both decidual health and compensating ovarian luteolysis • Resveratrol or progesterone alone could not prevent the incidence of PTB and did not increase the number of live mouse pups • Resveratrol reduced PGE2 levels with limited effects on PGF2α levels in both p53fl/fl and p53d/d decidual cells, as indicated by lower expression of Ptgs2 (encoding COX2) in mice and human decidual cells treated with resveratrol • Resveratrol has an inverse regulatory effect on AMPK and mTORC1 signaling in decidual cells, controlling parturition timing to prevent PTB • Resveratrol did not appear to have detrimental effects on pregnancy outcomes • Progesterone injection alone showed some adverse effects on pup viability, likely due to the normal decline in progesterone levels approaching parturition(11)
Furuya et al, 201520 and 40 mg/kg POOn days 12-14 1x/day; On day 15 2x/day (at 6 to 12 h after LPS injection)N/A• Resveratrol decreased the rate of PTB compared to the control group (48.6±19.4 vs. 97.8±1.9%) • Resveratrol may protect against pathological PTB by suppressing the elevated levels of pro-inflammatory cytokines TNF-α (16.7 vs. 53.9 pg/ml) and IL-1β (88.4 vs. 403 pg/ml), but not IL-6 levels in peritoneal washes • Resveratrol inhibited mRNA expression of TNF-α in uterine cervices, but not IL-1β and IL-6 levels • Resveratrol dose-dependently reduced mRNA expression of TNF-α and IL-1β, but not IL-6 levels, in peritoneal macrophages • Resveratrol suppressed the pro-inflammatory cytokine-mediated elevation of COX-2 mRNA levels produced by peritoneal macrophages(17)
Novaković et al, 2015A range between 1-100 µM POResveratrol was incrementally added to the bathing solution, allowing for equilibrium response to be achieved within ~20 min• Glibenclamide (10 µM) • Iberiotoxin (100 nM) 4-Amino- pyridine (1 mM)• Resveratrol with the concentration of 1-100 µM might inhibit the amplitude of oxytocin-induced contractions in a concentration-dependent manner (pD2 4.52+0.11, maximal responses: 82.25+1.50%) • Resveratrol significantly reduced spontaneous rhythmic contractions and the amplitude of phasic contractions induced by oxytocin but had no effect on tonic contractions induced by oxytocin • Resveratrol suppressed the contractility of human term pregnant myometrium by modulating different myometrial K+ channels, including the activation of KATP channels, Kv channels, and BKCa channels through an increase in intracellular Ca2+ • Resveratrol may induce relaxation of pregnant myometrium, at least partially, through the activation of Kir6.2/SUR1 channels. Resveratrol at a concentration of 10 mM showed insensitivity to all K+ channel blockers(31)

[i] AEA, N-arachidonoylethanolamine; AMPK, AMP-activated protein kinase; COX-2, cyclooxygenase 2; kg, IL, interleukin; iNOS, inducible nitric oxide synthase; kilogram; Kv channel, Voltage-gated potassium channel; LPS, lipopolysaccharide; mg, milligram; ml, milliliter; mRNA, messenger RNA; mTORC 1, mammalian target of rapamycin complex 1; nM, nanomolar; NOS, nitric oxide synthase; pg, picogram; PGE2, prostaglandin E2; PGF2α, prostaglandin F2α; TNF-α, tumor necrosis factor α; PO, per oral; PTB, preterm birth; µM, micromolar.

Discussion

Complications arising from PTB are a major contributor to neonatal mortality and the second leading cause of mortality among children under the age of 5 years. Surviving children often experience lifelong disabilities. The reduction of PTB rates is a global priority, due to its significant impact on healthcare and the economy (5). Addressing prematurity continues to be a significant challenge in the field of maternal and child health, as there is no single drug that effectively targets all the underlying pathways leading to prematurity. Each tocolytic agent has its own limitations and potential side-effects, with limited evidence of efficacy in improving neonatal outcomes (32).

The present systematic review examined four articles that explored the role of resveratrol in cases of prematurity. The studies were conducted between 2015 and 2017, with two taking place in developing countries (Argentina and Serbia) (8,31) and the other two in developed countries (USA and Japan) (11,17). The analysis included two in vivo studies conducted on mice (8,17), one in vitro study using human myometrial tissue (31), and one study that combined both in vivo and in vitro research (11). In the in vivo studies, PTB was induced using LPS, and resveratrol was administered orally (8,11,17). However, the dosage and timing of resveratrol varied, ranging from 3 to 40 mg/kg on days 8 to 15. In the in vitro studies, resveratrol was applied as a bath to cell tissue that had been previously exposed to oxytocin (11,31). To the best of our knowledge, there is no published study available on the potential impact of resveratrol dosages on human tissues and cells. However, the available studies have indicated that when resveratrol is orally administered to mice, the compound remains unaltered and is retained within the tissues (33).

Following oral administration, low concentrations of resveratrol have been detected in both the plasma and tissues of humans and experimental animals (34). Studies that involved the incremental dosing of resveratrol from 25 to 5,000 mg consistently demonstrated an increase in plasma concentrations without any signs of metabolic saturation. However, even at the highest dose of 5,000 mg, the observed peak plasma levels only reached ~500 ng/ml. There is a possibility of metabolic saturation with prolonged or repeated dosing, which may result in elevated resveratrol levels in plasma and tissues (35). Although it remains a hypothesis, studies have suggested that it may be possible to achieve biologically active concentrations of resveratrol and/or its metabolites in human subjects through chronic dosing (36,37).

Bariani et al (8) and Furuya et al (17) demonstrated that resveratrol effectively reduced the incidence of PTB in mice through the reduction of pro-inflammatory cytokines (e.g., TNF-α and IL-1β). Novaković et al (31) demonstrated that resveratrol significantly decreased both rhythmic contractions and contraction amplitude. However, Deng et al (11) argued that resveratrol alone may not be sufficient in preventing PTB events. By analyzing these four articles alongside supporting literature, the authors compiled a comprehensive understanding of the risk factors and pathophysiology of prematurity. Additionally, the present study explored the mechanisms of action through which resveratrol targets prematurity mechanisms, the efficacy of resveratrol as a preventive agent for PTB, and the pharmacological characteristics of resveratrol, from experimental settings to clinical applications.

Pathophysiology of prematurity in cellular and animal studies

The limited understanding of the complex etiology of PTB is a barrier to the development of effective strategies for its prevention. Recognized risk factors for PTB include multiple pregnancies, chorioamnionitis, maternal diseases, genetic factors, previous PTB and uterine abnormalities. However, not all cases of PTB have identifiable risk factors. The pathophysiology of PTB involves various processes, including myometrial contraction and extracellular matrix degradation. Among these processes, inflammation, which is initiated following implantation, plays a crucial role in PTB (37,38). Experimental animals, primarily mice or rats, play a critical role in studying the pathophysiology of PTB. The commonly used inflammation-inducing agent is LPS, derived from Escherichia coli endotoxins, administered intrauterine or intraperitoneally (39). In addition to LPS, various methods, including hormonal or immune agents, can induce PTB in mice (39).

Immunity, both innate and adaptive, plays a crucial role in the incidence of PTB. In mouse studies, macrophages induce PTB by producing pro-inflammatory cytokines such as matrix metallopeptidases, IL-1, IL-6 and IL-8. In the mouse model, PTB can be prevented by inhibiting the IL-1 receptor via the NF-κβ pathway (40). IL6-deficient mice exhibit resistance to LPS-induced PTB (41). Complement activation is also significant in preventing PTB, as women with spontaneous preterm labor have increased levels of complement C3a, C4a and C5a in infection-associated cases. Mice lacking the C5aR complement receptor demonstrate resistance to LPS-induced PTB in vitro (42).

In addition to the role of the innate immune system in PTB, the adaptive immune response is significant. PTB often occurs due to maternal intolerance to fetal antigens. Regulatory T-cells (Tregs) and effector T-cells (Teffs) undergo changes in frequency and phenotype. Tregs in women in preterm labor exhibit differential activation and reduced suppressive capacity (6). Toll-like receptors (TLRs) are another crucial component of the immune response. They activate signaling pathways that lead to cytokine and chemokine secretion by innate immune cells. TLR activation initiates inflammatory pathways that result in vaginal and preterm labor, immune cell recruitment, PG and MMP production, cervical ripening, and uterine contractions. TLR4 and TLR2 influence the timing of delivery during pregnancy (43,44). In addition to maternal TLR expression, fetal TLR expression may impact birth timing, with polymorphic alleles of fetal TLR4 and TLR2 associated with prematurity (45).

Current research explores the role of the microbiome role in modulating the risk of PTB. The composition of the vaginal microbiota influences the risk of PTB, with dysbiosis linked to increased levels of pro-inflammatory cytokines, such as CXCL10, in early pregnancy (46). However, administering pre-conception antibiotics to women with a history of PTB does not reduce the rates of PTB; instead, it is associated with a lower occurrence of LBW babies and early labor (47).

Among the various mechanisms associated with PTB, including innate immunity, adaptive immunity and dysbiosis, there is a notable interest in elucidating the mechanisms through which resveratrol intervenes in these processes. Resveratrol demonstrates antimicrobial properties against various pathogenic microorganisms, encompassing both Gram-positive and Gram-negative bacteria, as well as fungi. It can inhibit the growth of specific bacterial species at concentrations below 100 µg/ml, although higher concentrations are required to inhibit the growth of many bacterial organisms (48). The mechanism of action of resveratrol involves multiple pathways. It inhibits the electron transport chain and F0F1-ATPase, resulting in reduced cellular energy production. Resveratrol also interferes with DNA through the formation of a Cu(II)-peroxide complex and suppresses cell division by targeting the FtsZ gene. Moreover, it is effective in preventing biofilm formation and acts as both an antibiofilm and an anti-quorum sensing agent (49). The additional proposed mechanisms of action of resveratrol have been succinctly outlined in Fig. 3.

Mechanisms of action: Resveratrol targeting the mechanism of prematurity. Reduction of inflammatory mediators [nitric oxide (NO) synthase (NOS) and inducible NOS (iNOS)] in the uterus

The beneficial effects of resveratrol in inflammation-induced preterm delivery involve multiple pathways. One of these pathways is the decrease in the activity of uterine NOS and the protein expression of uterine iNOS. The excessive production of NO leads to the formation of RONS, which are associated with tissue damage, oxidative and nitrative stress (50). To assess the activity of NOS and the protein expression of iNOS in the uterus, measurements were taken 5 h following the injection of LPS. The results indicated an increase in NOS and iNOS activity in mice treated with LPS alone. Bu contrast, mice treated with resveratrol after LPS exposure showed no significant increase in NOS activity (51). This reduction in uterine NOS and iNOS activity is beneficial as it prevents excessive production of NO and its associated adverse effects, such as damage to reproductive tissues and oxidative stress (51).

Reduction of levels of inflammatory cytokines (TNF-α, IL-1β and IL-6) in the cervix and peritoneum. Resveratrol is known to inhibit the activity of NF-κβ, a transcription factor involved in the expression of pro-inflammatory cytokines, in various immune response-related disease models (52). This factor operates not only in immune cells but also in the uterine myometrium, decidual stroma and amniotic cells (17). It promotes the production of cytokines, such as TNF-α, IL-1β, IL-6 and IL-8, which are crucial for inducing labor, particularly in preterm cases (53,54). Resveratrol appears to inhibit this NF-κβ/cytokine loop (17).

Furuya et al (17) focused on murine pro-inflammatory cytokines and observed significant increases in the levels of TNF-α, IL-1β and IL-6 in peritoneal washes and cervical tissues following exposure to LPS. In their in vivo model, resveratrol effectively suppressed the elevation of TNF-α and IL-1β levels induced by LPS, with a slight reduction in IL-6 levels (17). Although IL-6, produced by various stromal cells, was not as markedly suppressed, it may be due to its induction by other pro-inflammatory cytokines and direct stimulation by LPS (55). Since the preterm delivery model is initiated by the stimulation of TLRs in macrophages by LPS, inhibiting the expression of TNF-α and IL-1β in peritoneal washes and the uterine cervix directly suppresses this signaling pathway (17).

Resveratrol effectively suppresses the production of pro-inflammatory cytokines in the cervix, which is a key factor in the pathogenesis of PTB. PTB often involves local inflammation caused by microbial infection, resulting in elevated levels of cytokines, PGs, uterine contractions, and cervical ripening (56). In the study by Furuya et al (17), cervical tissues exposed to LPS in the control group exhibited significantly higher mRNA levels of TNF-α, IL-1β and IL-6 compared to the group treated with resveratrol(17). Resveratrol notably reduced the peak levels of TNF-α and IL-1β mRNA, particularly resulting in significantly lower TNF-α levels (0.54±0.09) in the resveratrol-treated group compared to the control group (2.45±0.93) (17). Resveratrol also demonstrates anti-inflammatory effects on placental tissues in pregnant women by downregulating the sirtuin-1 gene post-delivery (57). Its mechanism involves inhibiting the transcription activity of NF-κB, reducing the expression of the NF-κβ sub-unit RelA/p65, and stabilizing inhibitory I-κβ (57,58). Previous studies have demonstrated that resveratrol can suppress inflammatory responses in endometrial stromal cells in patients with endometriosis. The excessive production of pro-inflammatory cytokines can further stimulate the overproduction of PG, leading to increased uterine contractions and cervical maturation (59). Therefore, the ability of resveratrol to suppress pro-inflammatory cytokines in the cervix holds promising therapeutic implications for preventing PTB.

The pathogenesis of PTB involves both the cervix and the peritoneal area. In the study by Furuya et al (17), the injection of LPS into the cervix significantly increased the levels of pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6) in peritoneal washes, with IL-1β spiking to levels 480 times higher (17). This led to intrauterine inflammation and PTB in the experimental model. Peritoneal washes assessed the local immune responses to LPS, and the administration of resveratrol markedly decreased the levels of TNF-α and IL-1β. TNF-α levels were reduced to one-third, and IL-1β to about one-fourth, in mice treated with resveratrol compared to controls. However, there was no significant difference in IL-6 levels between the groups (17). Furuya et al (17) also demonstrated that resveratrol prevented the increased production of TNF-α and IL-1β by peritoneal macrophages in response to LPS. Comparing the local immune responses within 4 h of the injection, the results of peritoneal lavage revealed a significant decrease in TNF-α and IL-1β levels following the administration of resveratrol (17). TNF-α levels were approximately one-third in mice treated with resveratrol (16.7 vs. 53.9 pg/m), while IL-1β levels were approximately one-quarter (88.4 vs. 403 pg/ml, P<0.01). However, there was no significant difference in IL-6 levels between the resveratrol-treated mice and the controls (17).

Reducing inflammatory mediators from peritoneal macrophages. COX-2 enzymes are upregulated in response to pro-inflammatory cytokines, particularly in macrophages. This leads to elevated cytokine-mediated PGs, including PGE2 and PGF2α, which are significant in the mechanisms of PTB (60,61). TNF-α and IL-1β are central to the COX-2 pathway, and macrophages, responsible for their production, are suspected to play a role in the local inflammatory mechanism of PTB (8). In the study by Furuya et al (17), LPS-exposed peritoneal macrophages exhibited a significant increase in COX-2 mRNA levels for TNF-α and IL-1β. Resveratrol treatment effectively suppressed COX-2 production in these macrophages. This aligns with prior observations in peritoneal washes and cervical tissues by Furuya et al (17). Resveratrol reduced TNF-α, IL-1β, and COX-2 mRNA levels in macrophages induced by LPS in a concentration-dependent manner (17). Macrophages play a crucial role in COX-2 production and subsequent PGE2 and PGF2α production. A previous study demonstrated that omega-3 fatty acids, which also have antioxidant and anti-inflammatory properties, can suppress macrophage function and the elevation of PGE2 and PGF2α in gestational tissues induced by LPS (62). These findings collectively suggest that resveratrol primarily targets uterine-associated macrophages, offering the potential to modulate the local inflammatory response in PTB.

Reduction of COX-2 and PGs in the uterus. COX-2 produces PGE2 and PGF2α, potent inducers of uterine contractions and cervical ripening (19). PGs play a pivotal role in cervical maturation and uterine contractions, increasing amniotic fluid during labor (63,64), an essential process in childbirth (65). The upregulation of COX-2 is associated with PTB pathology in both mouse models and humans. The study by Bariani et al (8) revealed that LPS-induced uterine inflammation increases COX-2 expression, initiating preterm labor. This upregulation occurs in tissues such as the amnion, choriodecidua, and myometrium, linking it to PTB pathology (66).

The ability of resveratrol to modulate COX-2 expression, a key player in PG production, led to the assessment of uterine PG levels, PGE2, PGF2α, 6-keto-PGF1α and PGI2(8). Resveratrol has been shown to effectively suppress the LPS-induced elevation of COX-2 in gestational tissues (8). Its administration results in the downregulation of both COX-2 mRNA and protein expression in the uterus, preventing COX-2 gene and protein overexpression induced by LPS (8,67). Notably, LPS alone or in combination with resveratrol does not alter the levels of 15-hydroxyprostaglandin dehydrogenase (15-Pgdh) mRNA and COX-1 protein levels (8), which decrease PG production, counteracting the effects of COX-2(68). In the study by Bariani et al (8), resveratrol administration prevented the increase in uterine PGE2 levels and partially affected PGF2α levels, with no significant impact on the 6-keto-PGF1α concentration (8). The reduction in pro-inflammatory cytokines by resveratrol may explain the lower PG expression, as TNF-α and IL-1β induce PGE2 and PGF2α in choriodecidual tissues (69). In summary, by decreasing PG synthesis, resveratrol exhibits potential in helping to prevent preterm labor.

Reduction of endocannabinoid levels. The endocannabinoid system (ECS) consists of cannabinoid receptors CB1 and CB2, endogenous lipid ligands called endocannabinoids, and regulating enzymes that synthesize and break down these lipids. All endocannabinoids belong to the eicosanoid class (70). Endocannabinoids are unsaturated fatty acid derivatives that interact with this system, eliciting specific responses (71). Anandamide (AEA) and 2-arachidonoylglycerol (2-AG) are key endocannabinoids that are released enzymatically from cell membrane phospholipid precursors in response to various stimuli, including neurotransmitters, depolarizing agents, and hormones (71). AEA is present in the uterus, and its levels in the blood increase during normal labor, suggesting the involvement of the ECS in reproductive and preterm labor processes (72-74). The study by Bariani et al (8) demonstrated that LPS and/or resveratrol affected uterine endocannabinoid levels. LPS did not affect the 2-AG levels, but resveratrol alone reduced its levels. AEA increased with LPS, but resveratrol prevented this increase.

The effects of AEA on the uterus are conflicting, with some studies demonstrating the relaxation of the pregnant myometrium (75), while others have reported that the activation of CB1 receptors leads to the increased production of PGE2 in the amnion and choriodecidua (76). CB1 receptor activation is associated with the increased production of PGF2α in inflammation-induced preterm labor (77). Prolonged endocannabinoid signaling is linked to adverse pregnancy outcomes (78). Bariani et al (8) found that LPS altered AEA and related endocannabinoid-like lipid levels in the uterus, consistent with previous research (75,76). LPS-induced cytokines can affect endocannabinoid production by modulating the expression and activity of fatty acid amide hydrolase (FAAH) (79,80). In the study by Bariani et al (8) treatment with resveratrol restored AEA and other lipid levels to the control levels, although the exact mechanisms involved remain unclear. The increased AEA production induced by LPS was associated with a higher incidence of preterm labor, while the tocolytic effects of resveratrol correlated with reduced uterine AEA content (8).

Bariani et al (8) discovered that LPS increased the AEA levels in the uterus, resulting in a greater likelihood of preterm labor. However, resveratrol prevented this increase, indicating its tocolytic effect in reducing uterine endocannabinoids during inflammation (8). In addition to endocannabinoids, endocannabinoid-like lipids from the N-acyl ethanolamine, N-arachidonoyl glycine (NAGlys), N-arachidonoyl serine (NASers) and 2-acyl-sn-glycerols families can undergo changes in composition during LPS-induced pregnancy inflammation (8). The specific molecular mechanism through which resveratrol modifies uterine endocannabinoids and lipid composition remains unclear. However, it is known that NAGlys and NASers, which are part of these lipids, do not activate CB1 or CB2 receptors (81). NASers have been observed to regulate calcium-activated potassium channels (82) and N-type calcium channels (83). Additionally, NAGlys can be metabolized by COX-2 and FAAH, potentially functioning as endogenous enzyme inhibitors (84).

There was a 1.5-fold increase in the average plasma levels of AEA from 1.20±0.57 nM in non-labor conditions to 1.82±0.87 nM during labor (P<0.0001) (72). A plasma AEA concentration exceeding 1.095 nM was found to be a better predictor of PTB before 37 weeks of gestation compared to currently used predictors, such as cervical length, oncofetal fibronectin and cervical insulin-like growth factor binding protein 1. It demonstrated 87.14% specificity, 25.93% sensitivity, a 70.24% negative predictive value (NPV), and a 61.2% positive predictive value (PPV), surpassing existing predictive tests (85). The average plasma AEA level significantly increased in women with a positive fetal fibronectin test compared to those with a negative result, providing additional evidence of the role of AEA in labor and its potential use as a predictor for PTB (85). AEA levels are regulated through cellular uptake by the AEA transporter and enzymatic degradation by FAAH on the cell membrane (86). LPS-induced cytokines influence endocannabinoid production by affecting FAAH protein expression and enzymatic activity (87). Anti-inflammatory cytokines (IL-4 and IL-10) enhance FAAH activity, while pro-inflammatory cytokines (IL-12 and IFN-γ) reduce FAAH activity and protein expression in peripheral mononuclear cells (lymphocytes) (8,88). During pregnancy, FAAH levels in peripheral mononuclear cells show an inverse correlation with blood AEA levels (89). Low AEA levels with high FAAH are crucial for the successful development of pregnancy (90,91). The reduced activity and expression of anandamide hydrolase in peripheral lymphocytes can serve as an early indication of spontaneous abortion before eight weeks of gestation, offering potential diagnostic utility for pregnancy monitoring and fertility assessment (91).

Modulating 5' AMP-activated protein kinase (AMPK) and mammalian target of rapamycin complex (mTORC) pathways. Inflammation and oxidative stress are widely recognized risk factors for PTB. Studies conducted on mouse models, specifically those with spontaneous and inflammation-induced PTB with p53 deletion (p53d/d females), have indicated the involvement of mTOR activation in decidual senescence and the excessive production of PGs derived from COX2, ultimately leading to PTB (11). In female mice with the p53d/d phenotype, a noticeable decrease in the growth of uterine decidual cells due to accelerated senescence has been observed (92,93). This condition is accompanied by elevated levels of pAkt, p21, IL-8 and various CXCL cytokines, collectively contributing to the senescence-associated secretory phenotype, which further promotes decidual senescence and PTB (93). Additionally, this p53 deletion has been associated with PTB, which can also be influenced by factors, such as increased ROS production due to conditions such as pre-eclampsia, as well as environmental factors such as smoking and pollution (93).

Deng et al (11) demonstrated that resveratrol, an antioxidant and anti-aging agent, activated AMPK signaling and inhibited mTORC1 signaling in decidual cells without causing any adverse effects on the mother or offspring. Consequently, resveratrol offers protection against both spontaneous and inflammation-induced PTB in p53d/d females. Similar beneficial effects have also been observed with the use of AMPK activators such as metformin, which enhances decidual health and reduces PTB rates in mouse models (11). Human fetal membranes contain both AMPK and its phosphorylated form (p-AMPK). Membranes obtained from spontaneous term labor and pre-labor rupture display significantly lower levels of p-AMPK compared to intact membranes (11). Pre-treatment of human fetal membranes with AMPK activators reduces the production of inflammatory cytokines in response to LPS, indicating the anti-inflammatory effects of p-AMPK on fetal membranes (94). Resveratrol decreases PGE2 levels by suppressing Ptgs2, the COX2-encoding gene, in decidual cells. It also inhibits hyperactive mTORC1 through both AMPK-dependent and/or AMPK-independent pathways, underscoring its role in the regulation of AMPK and mTORC1 signaling in an inverse manner (95).

Other potential benefits of resveratrol: Smooth muscle relaxation. Resveratrol has the ability to relax smooth muscle, which may have an impact on PTB by targeting potassium (K+) channels (31,96-98). The proper functioning of K+ channels is crucial for maintaining a relaxed uterine state during pregnancy by aiding in membrane repolarization (99). The dysregulation of K+ channels can lead to abnormal uterine activity, contributing to conditions such as preterm labor (31). Poorly functioning or less active K+ channels can decrease the repolarizing current in myometrial smooth muscle cells, resulting in untimely uterine contractions and preterm delivery. Conversely, excessive expression of K+ channels in the later stages of pregnancy may hinder synchronized uterine muscle activity needed for full-term labor (98). Various types of K+ channels, including big Ca2+-sensitive K+ (BKCa) channels, ATP-sensitive K+ (KATP) channels, Kv channels and SK channels, induce relaxation in both non-pregnant and pregnant myometrium, demonstrating the complex regulation of uterine tone (31,96-98). BKCa channels, which hold a particular influence on uterine smooth muscle, play a significant role in inducing smooth muscle relaxation and repolarizing K+ current (99). Their activity varies throughout pregnancy, and their modulation is regulated by 17b-estradiol (31). BKCa channels have a more pronounced relaxant effect during mid-gestation (100,101), indicating their importance in maintaining uterine quiescence (99).

The regulation of myometrial tone during pregnancy involves the modulation of ion channels. β-receptor agonists, used to delay preterm labor, interact with BKCa channels via ß2- and ß3-receptors, resulting in uterine relaxation (99). Human studies suggest increased levels of BKCa channel in late pregnant non-laboring myometrium but decreased expression in preterm and term laboring myometrium (99). KATP channels, composed of Kir6 channels and sulfonylurea receptors, play a role in myometrial quiescence; however, their specific function in the myometrium is not yet clear. The downregulation of KATP channels, particularly Kir6.1 and SUR2B subunits, may increase uterine excitability and labor contractions in term pregnancy (101). The regulation of KATP channels varies with the stage of gestation and the incidence of labor contractions in the human myometrium, necessitating further research for a comprehensive understanding (101). Aberrant K channel function or expression is associated with conditions such as PTB and preeclampsia (96,102). Resveratrol can modulate K channel activity, potentially offering new treatments for these conditions by reducing intracellular calcium levels and inhibiting various contractions (31). Resveratrol effectively inhibits uterine smooth muscle contractions, which can lead to infertility, endometriosis, miscarriage, or PTB (31,96,97,99). It targets KATP, BKCa and Kv channels to achieve relaxation effects on the uterus (97). This relaxation potential extends to pregnancy-related disorders, such as PTB, by relaxing the myometrial smooth muscle and fetoplacental blood vessels, inhibiting contractions induced by oxytocin, PGs and acetylcholine (96).

Efficacy of resveratrol in preventing preterm birth

Resveratrol shows promise as an herbal preventive agent for PTB. In the study by Bariani et al (8), the administration of resveratrol resulted in a 64% reduction in PTB rates and a 28% decrease in mortality rates. These effects may be attributed to resveratrol's impact on uterine NOS activity, iNOS expression, COX-2, PGE2, and AEA profiling. Additionally, this treatment led to a decrease in antepartum hemorrhage, fetal toxicity, and offspring mortality. Importantly, mice treated with resveratrol did not experience vaginal bleeding, and their uterine morphology improved (8). Deng et al (11) demonstrated that resveratrol has the potential to prevent PTB through its modulation of AMPK and mTORC1 signaling in decidual cells. The administration of resveratrol offers several advantages, including no adverse effects on pregnancy outcomes, effective targeting of decidual health, and compensation for ovarian luteolysis (11). The study conducted by Furuya et al (17) also supports the tocolytic effects of resveratrol by suppressing peritoneal macrophage-induced inflammation and preventing spontaneous PTB in a genetic model of accelerated decidual senescence.

Furthermore, Furuya et al (17) discovered that resveratrol had a significant impact on reducing the rate of PTB in mice. At a dosage of 20 mg/kg, the PTB rate was 48.6±19.4% compared to 97.8±1.9% in the control group (P=0.03). Morever, at a dosage of 40 mg/kg, the PTB rate was 57.1±13.8% compared to 83.7±13.2% in the control group (P=0.15). By orally administering resveratrol, the occurrence of LPS-induced prematurity in mice was reduced through the suppression of inflammatory mediators, PGs, peritoneal macrophages and endocannabinoid compounds in the maternal reproductive organs (17). Novaković et al (31) demonstrated that resveratrol effectively prevents PTB by inhibiting uterine contractions, including those stimulated by oxytocin. Resveratrol achieves this by regulating various myometrial K+ channels, such as KATP, Kv and BKCa channels, by increasing intracellular Ca2+ levels. Moreover, it induces partial relaxation of the myometrium by activating Kir6.2/SUR1 channels. Nonetheless, there are certain limitations to consider, such as the failure of a single resveratrol treatment to prevent PTB in the study conducted by Deng et al (11) and its lack of effectiveness against tonic contractions induced by oxytocin (31).

Pharmacological profile of resveratrol: From bench to bedside

Oral resveratrol is readily absorbed but undergoes rapid metabolism (102). Azachi et al (103) conducted a study in which red grape cell (RGC) resveratrol exhibited two plasma concentration peaks: One at 1 h following administration and another at 5 h following administration. The initial peak is likely a result of the extensive glucuronidation and sulfation of resveratrol in the enterocytes of the small intestine. By contrast, the second peak occurs due to the flow of bile-containing metabolites from the liver to the intestines, a normal occurrence after food consumption that may prolong the pharmacological effect of certain substances and their metabolites (103,104).

The median time to peak drug concentration (Tmax) for total resveratrol at a dose of 150 mg RGC is 4 h (range, 0.67-6.00), and at 50 mg it is 1 h (range, 0.33-8.00). Moreover, the median Tmax for free resveratrol at a dose of 150 mg RGC is 1 h (range, 0.33-4.00) (103). Resveratrol exhibits a high oral absorption but rapid metabolism, resulting in poor overall bioavailability. Even with a high dose of 5 g, it only reaches a peak plasma-free concentration of 538 ng/ml. This limited bioavailability may be attributed to low solubility, similar to other polyphenol aglycones, which struggle to form hydrogen bonds due to hydrophobic interactions with hydroxyl or aromatic groups (103). One approach to enhance solubility is through glycosylation of the resveratrol parent compound (103).

Resveratrol interacts with cytochrome P450 enzymes, inhibiting CYP3A4, CYP2D6 and CYP2C9, while inducing CYP1A2, potentially affecting drugs metabolized by these enzymes (105). Urinary excretion studies demonstrate high absorption rates (at least 70%) after oral resveratrol intake, with 53.4 to 84.9% of total radioactivity recovered in urine. However, rapid biotransformation leads to minimal unmodified resveratrol in circulation and relatively high levels (maximum, 2 µM) of resveratrol metabolites after a 25 mg oral dose, resulting in near-zero resveratrol bio-availability (104). The intravenous injection of a 0.2 mg dose shows no second peak and a rapid decline in plasma total radioactivity over the first hour, indicating extensive distribution. Plasma half-life durations are similar after both routes (9.2 h orally and 11.4 h intravenously), with dose-dependent resveratrol plasma levels reaching up to 530 ng/ml with higher doses (up to 5 g) (104). The administration of resveratrol during pregnancy appears to be relatively safe for both the mother and fetus, with no reported toxicity or adverse pregnancy outcomes (11,102).

Resveratrol is primarily sourced from grapes, grape skins, peanuts, red wine, cranberries and Japanese knotweed (Polygonum cuspidatum) (102,105). The quantity of resveratrol varies among these sources. For example, RGCs contain between 726 to 916 mg/kg (103), while agricultural red grapes range from 0 to 42.5 mg/kg (103). Other examples include jamun fruit with 11.19 to 34.87 mg/g of dry weight, mulberries with 35.8 to 50.61 mg/g, jackfruit with 0.07 to 3.56 mg/g (106), apples with 47 mg/g, broccoli with 18 mg/g, onions with 18 mg/g and black tea with 11.3 mg/g (107). Typical concentrations of resveratrol in various food products to meet the recommended daily allowance (RDA) of 1 g are as follows: Red grapes (92-1,604 mg/kg fresh weight), white grapes (59-1,759 mg/kg fresh weight), peanuts without seed coats (0.03-0.14 mg/g), red wines (0.361-1.972 mg/l), white wines (0-1.089 mg/l), rosé wines (0.29 mg/l), beers (1.34-77.0 mg/l), tomato skins (19 mg/g dry weight), dark chocolate (350 mg/kg), milk chocolate (100 mg/kg), Itadori tea (68 mg/100 ml) and apples (400 mg/kg fresh weight) (108).

Due to its limited solubility in water, resveratrol is primarily absorbed through passive diffusion in the colon and undergoes significant pre-systemic metabolism in the liver. This liver metabolism markedly reduces the levels of free resveratrol before it enters systemic circulation. Resveratrol is conjugated to more soluble glucuronides (e.g., resveratrol-3-O-glucuronide, resveratrol-4-O-glucuronide) and sulfates (e.g., resveratrol-trisulfate) in phase II metabolism or binds to albumin and lipoproteins. As a result, the therapeutic effectiveness of oral resveratrol is relatively low. Efforts to improve the bioavailability of resveratrol or delay its phase II metabolism through the use of resveratrol prodrugs are crucial for its biomedical applications (108,109).

The oral supplementation of 80 mg resveratrol in overweight pregnant women has demonstrated a reduction in the incidence of gestational diabetes mellitus, improvement in lipid profiles, and a decrease in glucose levels within a 60-day period. Additionally, resveratrol administration at a dose of 50 mg for up to 5 doses has been found to lower blood pressure in patients with preeclampsia (102). However, it should be noted that there is limited clinical research available on the use of resveratrol during pregnancy, with more extensive studies available for patients with diabetes, cardiovascular issues and metabolic disorders (105). Challenges in the clinical application of resveratrol include the wide range of dosages used (ranging from 5 mg to 5 g daily), the diverse dose-effect associations observed in the SIRT1 pathway, concerns regarding renal toxicity associated with micronized oral formulations such as SRT501 (commercially marketed as a supplement, rather than a definitive treatment drug) and gastrointestinal side-effects at high doses. Furthermore, it has been observed that resveratrol can activate estrogen-regulated genes that are associated with estrogen-dependent neoplasms (105).

Strength and limitations

This presented systematic review highlights a unique academic perspective on the prevention of PTB using resveratrol. It offers original contributions to the field of preclinical investigations of natural compound medicine for PTB. Notably, to the best of our knowledge, this systematic review is the first to specifically examine the impact of resveratrol in preventing PTB and highlights its promising benefits. The present study goes beyond a mere summary of the literature by providing a comprehensive framework that explains the preventive mechanisms of resveratrol. It also includes a visualized image that consolidates the scattered literature. By adopting this approach, the present study contributes significantly to the development of strategies to combat PTB. Furthermore, the present study stands out for its meticulous quality assessment, employing stringent predetermined criteria to critically evaluate the included studies.

In the present systematic review, the authors were not able to conduct a meta-analysis due to two main reasons. First, there was a limited number of articles included, which makes it impractical. Second, the data and results presented in these studies were presented in a heterogeneous manner, rendering it impossible to pool the data. The main limitation of this systematic review was the diverse nature of how data and results are presented across the included studies, which prevents a meta-analysis from being practically feasible. Additionally, while the significant prevalence of favorable outcomes in the present systematic review is encouraging, it is important to consider the possibility of publication bias, where studies with unfavorable results may remain unpublished. Furthermore, the strict inclusion criteria resulted in the exclusion of several studies that did not directly involve resveratrol in PTB. Nonetheless, the insights from these excluded studies are still integrated into the discussion section, adding to the broader conversation on the subject.

Despite the potential of resveratrol as a tocolytic agent for preventing PTB, particularly in cases involving intrauterine inflammation, there is a scarcity of human studies evaluating its effectiveness in this regard, and no current human clinical trials have been conducted, at least to the best of our knowledge. While mouse models provide valuable information, their limitations stem from significant anatomical and physiological differences compared to humans, and findings from in vitro and in vivo studies may not directly translate to human outcomes due to inherent species disparities. However, this juncture presents an opportune moment for researchers to validate the potential benefits of resveratrol through dedicated human studies. The current insights from various models offer valuable information about labor and delivery mechanisms, underscoring the need for well-designed prospective studies to assess the efficacy of resveratrol in human populations.

In conclusion, the findings presented herein indicate that resveratrol may have the ability to prevent preterm labor induced by LPS by inhibiting pro-inflammatory mediators, reducing PG levels, improving endocannabinoid profiles, relaxing smooth muscle and preventing excessive uterine contractions through ion channels. Considering its ability to cross the placenta, future studies are required to investigate its potential protective effects on embryos. Moreover, a better understanding of the role of resveratrol in human or similar uterine physiology necessitates further investigation. Urgent clinical trials are necessary to determine the optimal dosage of resveratrol for PTB therapy using standardized formulations. Given the low oral bioavailability of resveratrol, improving its pharmacological properties is pivotal. Structural optimization and the use of resveratrol-encapsulated nanoparticles can enhance efficacy, reduce dosages, minimize side-effects and target specific organs. Additionally, exploring the potential of locally derived resveratrol from plants and conducting oligomer research shows promise. Long-term studies are required to substantiate the scientific potential of resveratrol.

Supplementary Material

Details of reliability assessment according to ToxRtool (21,25).
Summary of scoring reliability assessment (21,25).
Interpretation of reliability assessment (21,25).

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

MH served as the principal investigator for the present study, conceiving the research and making the decision to publish. In the role of guarantor, MH took full responsibility for the work. MH, MIDR, and ABP jointly designed the methodology. MH, MIDR and ABP conducted the investigation, had complete access to the literature data, contributed to data analysis and curation, drafted the manuscript and secured funding. MH and ABP utilized software to create visualizations of the study findings and managed the project. Additionally, MH provided resources, validated all evidence analyses and supervised the study process meticulously. MH, MIDR and ABP confirm the authenticity of all the raw data. All authors have read and approved the final version for publication.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

World Health Organization (WHO): Factsheet of Preterm Birth. WHO, Geneva, 2018. https://www.who.int/news-room/fact-sheets/detail/preterm-birth.

2 

Pan American Health Organization (PAHO): 152 million babies born preterm in the last decade. PAHO, Washington, DC, 2013. https://www.paho.org/en/news/15-6-2023-152-million-babies-born-preterm-last-decade#:~:text=An%20estimated%2013.4%20million%20babies,37%20weeks%20of%20pregnancy)%20worldwide. Accessed Sep 28, 2023.

3 

Cao G, Liu J and Liu M: Global, regional, and national incidence and mortality of neonatal preterm birth, 1990-2019. JAMA Pediatr. 176(787)2022.PubMed/NCBI View Article : Google Scholar

4 

Bolisetty S, Dhawan A, Abdel-Latif M, Bajuk B, Stack J and Oei JL: New South Wales and Australian Capital Territory Neonatal Intensive Care Units' Data Collection. Intraventricular hemorrhage and neurodevelopmental outcomes in extreme preterm infants. Pediatrics. 133:55–62. 2014.PubMed/NCBI View Article : Google Scholar

5 

Beam AL, Fried I, Palmer N, Agniel D, Brat G, Fox K, Kohane I, Sinaiko A, Zupancic JAF and Armstrong J: Estimates of healthcare spending for preterm and low-birthweight infants in a commercially insured population: 2008-2016. J Perinatol. 40:1091–1099. 2020.PubMed/NCBI View Article : Google Scholar

6 

Romero R, Dey SK and Fisher SJ: Preterm labor: One syndrome, many causes. Science. 345:760–765. 2014.PubMed/NCBI View Article : Google Scholar

7 

Bastek JA, Gómez LM and Elovitz MA: The role of inflammation and infection in preterm birth. Clin Perinatol. 38:385–406. 2011.PubMed/NCBI View Article : Google Scholar

8 

Bariani MV, Correa F, Leishman E, Domínguez Rubio AP, Arias A, Stern A, Bradshaw HB and Franchi AM: Resveratrol protects from lipopolysaccharide-induced inflammation in the uterus and prevents experimental preterm birth. Mol Hum Reprod. 23:571–581. 2017.PubMed/NCBI View Article : Google Scholar

9 

Tency I: Inflammatory response in maternal serum during preterm labour. Facts Views Vis Obgyn. 6:19–30. 2014.PubMed/NCBI

10 

Elovitz MA, Wang Z, Chien EK, Rychlik DF and Phillippe M: A new model for Inflammation-Induced preterm birth. Am J Pathol. 163:2103–2111. 2003.PubMed/NCBI View Article : Google Scholar

11 

Deng W, Cha J, Yuan J, Haraguchi H, Bartos A, Leishman E, Viollet B, Bradshaw HB, Hirota Y and Dey SK: p53 coordinates decidual sestrin 2/AMPK/mTORC1 signaling to govern parturition timing. J Clin Invest. 126:2941–2954. 2016.PubMed/NCBI View Article : Google Scholar

12 

Simhan HN and Caritis SN: Prevention of preterm delivery. N Engl J Med. 357:477–487. 2007.PubMed/NCBI View Article : Google Scholar

13 

Abramovici A, Cantu J and Jenkins SM: Tocolytic therapy for acute preterm labor. Obstet Gynecol Clin North Am. 39:77–87. 2012.PubMed/NCBI View Article : Google Scholar

14 

Alfirevic Z: Tocolytics: Do they actually work? BMJ. 345:e6531. 2012.PubMed/NCBI View Article : Google Scholar

15 

Croen LA, Connors SL, Matevia M, Qian Y, Newschaffer C and Zimmerman AW: Prenatal exposure to β2-adrenergic receptor agonists and risk of autism spectrum disorders. J Neurodev Disord. 3:307–315. 2011.PubMed/NCBI View Article : Google Scholar

16 

Gidaya NB, Lee BK, Burstyn I, Michael Y, Newschaffer CJ and Mortensen EL: In utero exposure to β-2-Adrenergic receptor agonist drugs and risk for autism spectrum disorders. Pediatrics. 137(e20151316)2016.PubMed/NCBI View Article : Google Scholar

17 

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 Cie. 22:1561–1568. 2015.PubMed/NCBI View Article : Google Scholar

18 

Bourque SL, Dolinsky VW, Dyck JRB and Davidge ST: Maternal resveratrol treatment during pregnancy improves adverse fetal outcomes in a rat model of severe hypoxia. Placenta. 33:449–452. 2012.PubMed/NCBI View Article : Google Scholar

19 

Poudel R, Stanley JL, Rueda-Clausen CF, Andersson IJ, Sibley CP, Davidge ST and Baker PN: Effects of resveratrol in pregnancy using murine models with reduced blood supply to the uterus. PLoS One. 8(e64401)2013.PubMed/NCBI View Article : Google Scholar

20 

Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, Shamseer L, Tetzlaff JM, Akl EA, Brennan SE, et al: The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ. 372(n71)2021.PubMed/NCBI View Article : Google Scholar

21 

Schneider K, Schwarz M, Burkholder I, Kopp-Schneider A, Edler L, Kinsner-Ovaskainen A, Hartung T and Hoffmann S: ‘ToxRTool’, a new tool to assess the reliability of toxicological data. Toxicol Lett. 189:138–144. 2009.PubMed/NCBI View Article : Google Scholar

22 

Valenti C, Billi M, Pancrazi GL, Calabria E, Armogida NG, Tortora G, Pagano S, Barnaba P and Marinucci L: Biological effects of cannabidiol on human cancer cells: Systematic review of the literature. Pharmacol Res. 181(106267)2022.PubMed/NCBI View Article : Google Scholar

23 

Moser VC, Morris-Schaffer K, Richardson JR and Li AA: Glyphosate and neurological outcomes: A systematic literature review of animal studies. J Toxicol Environ Health B Crit Rev. 25:162–209. 2022.PubMed/NCBI View Article : Google Scholar

24 

Gupta M, Singh D, Rastogi S, Siddique HR, Al-Dayan N, Ahmad A, Sikander M and Sarwat M: Anti-cancer activity of guggulsterone by modulating apoptotic markers: A systematic review and meta-analysis. Front Pharmacol. 14(1155163)2023.PubMed/NCBI View Article : Google Scholar

25 

EU Science Hub: ToxRTool-Toxicological data reliability assessment tool. In: European Commission for Scientific Tools and Databases. European Commission, 2023. https://joint-research-centre.ec.europa.eu/scientific-tools-and-databases/toxrtool-toxicological-data-reliability-assessment-tool_en. Accessed Sep 21, 2023.

26 

University of Pittsburgh: Health Research Reporting Guidelines, Study Execution Manuals, Critical Appraisal, Risk of Bias, and Non-reporting Biases: From Cochrane Handbook for Systematic Reviews of Interventions [Internet]. University of Pittsburgh: Health Sciences Library System Guides, 2023. https://hsls.libguides.com/reporting-study-tools/risk-of-bias. Accessed Sep 21, 2023.

27 

Osmanovic A, Halilovic S, Kurtovic-Kozaric A and Hadziabdic N: Evaluation of periodontal ligament cell viability in different storage media based on human PDL cell culture experiments-A systematic review. Dental Traumatol. 34:384–393. 2018.PubMed/NCBI View Article : Google Scholar

28 

National Toxicology Program: Handbook for conducting a literature-based health assessment using OHAT approach for systemic review and evidence integration. National Institute of Environmental Health Science, pp1-102, 2019.

29 

Puidokas T, Kubilius M, Stumbras A and Juodzbalys G: Effect of leukocytes included in platelet concentrates on cell behaviour. Platelets. 30:937–945. 2019.PubMed/NCBI View Article : Google Scholar

30 

Center for Evidence-Based Medicine (CEBM): OCEMB Levels of Evidence. CEBM, Oxford, 2011. https://www.cebm.ox.ac.uk/resources/levels-of-evidence/ocebm-levels-of-evidence. Accessed Sep 21, 2023.

31 

Novaković R, Radunović N, Marković-Lipkovski J, Ćirović S, Beleslin-Čokić B, Ilić B, Ivković B, Heinle H, Živanović V and Gojković-Bukarica LJ: Effects of the polyphenol resveratrol on contractility of human term pregnant myometrium. Mol Hum Reprod. 21:545–551. 2015.PubMed/NCBI View Article : Google Scholar

32 

Wilson A, Hodgetts-Morton VA, Marson EJ, Markland AD, Larkai E, Papadopoulou A, Coomarasamy A, Tobias A, Chou D, Oladapo OT, et al: Tocolytics for delaying preterm birth: A network meta-analysis. Cochrane Database Syst Rev. 8(CD014978)2022.PubMed/NCBI View Article : Google Scholar

33 

Abd El-Mohsen M, Bayele H, Kuhnle G, Gibson G, Debnam E, Kaila Srai S, Rice-Evans C and Spencer JP: Distribution of [3H]trans-resveratrol in rat tissues following oral administration. Br J Nutr. 96:62–70. 2006.PubMed/NCBI View Article : Google Scholar

34 

Walle T, Hsieh F, DeLegge MH, Oatis JE and Walle UK: High absorption but very low bioavailability of oral resveratrol in humans. Drug Metab Dispos. 32:1377–1382. 2004.PubMed/NCBI View Article : Google Scholar

35 

Walle T: Bioavailability of resveratrol. Ann N Y Acad Sci. 1215:9–15. 2011.PubMed/NCBI View Article : Google Scholar

36 

la Porte C, Voduc N, Zhang G, Seguin I, Tardiff D, Singhal N and Cameron DW: Steady-State pharmacokinetics and tolerability of Trans-Resveratrol 2000 mg twice daily with food, quercetin and alcohol (Ethanol) in healthy human subjects. Clin Pharmacokinet. 49:449–454. 2010.PubMed/NCBI View Article : Google Scholar

37 

Brown VA, Patel KR, Viskaduraki M, Crowell JA, Perloff M, Booth TD, Vasilinin G, Sen A, Schinas AM, Piccirilli G, et al: Repeat dose study of the cancer chemopreventive agent resveratrol in healthy volunteers: Safety, pharmacokinetics, and effect on the Insulin-like growth factor axis. Cancer Res. 70:9003–9011. 2010.PubMed/NCBI View Article : Google Scholar

38 

Gracie S, Pennell C, Ekman-Ordeberg G, Lye S, McManaman J, Williams S, Palmer L, Kelley M, Menon R and Gravett M: PREBIC ‘-Omics’ Research Group. An integrated systems biology approach to the study of preterm birth using ‘-omic’ technology-a guideline for research. BMC Pregnancy Childbirth. 11(71)2011.PubMed/NCBI View Article : Google Scholar

39 

Green ES and Arck PC: Pathogenesis of preterm birth: Bidirectional inflammation in mother and fetus. Semin Immunopathol. 42:413–429. 2020.PubMed/NCBI View Article : Google Scholar

40 

McCarthy R, Martin-Fairey C, Sojka DK, Herzog ED, Jungheim ES, Stout MJ, Fay JC, Mahendroo M, Reese J, Herington JL, et al: Mouse models of preterm birth: Suggested assessment and reporting guidelines. Biol Reprod. 99:922–937. 2018.PubMed/NCBI View Article : Google Scholar

41 

Romero R and Tartakovsky B: The natural interleukin-1 receptor antagonist prevents interleukin-l-induced preterm delivery in mice. Am J Obstet Gynecol. 167:1041–1045. 1992.PubMed/NCBI View Article : Google Scholar

42 

Robertson SA, Christiaens I, Dorian CL, Zaragoza DB, Care AS, Banks AM and Olson DM: Interleukin-6 is an essential determinant of on-time parturition in the mouse. Endocrinology. 151:3996–4006. 2010.PubMed/NCBI View Article : Google Scholar

43 

Montalbano AP, Hawgood S and Mendelson CR: Mice deficient in surfactant protein A (SP-A) and SP-D or in TLR2 manifest delayed parturition and decreased expression of inflammatory and contractile genes. Endocrinology. 154:483–498. 2013.PubMed/NCBI View Article : Google Scholar

44 

Gonzalez JM, Franzke CW, Yang F, Romero R and Girardi G: Complement activation triggers metalloproteinases release inducing cervical remodeling and preterm birth in mice. Am J Pathol. 179:838–849. 2011.PubMed/NCBI View Article : Google Scholar

45 

Wahid HH, Dorian CL, Chin PY, Hutchinson MR, Rice KC, Olson DM, Moldenhauer LM and Robertson SA: Toll-Like Receptor 4 Is an essential upstream regulator of On-Time parturition and perinatal viability in mice. Endocrinology. 156:3828–3841. 2015.PubMed/NCBI View Article : Google Scholar

46 

Krediet TG, Wiertsema SP, Vossers MJ, Hoeks SBEA, Fleer A, Ruven HJ and Rijkers GT: Toll-like receptor 2 polymorphism is associated with preterm birth. Pediatr Res. 62:474–476. 2007.PubMed/NCBI View Article : Google Scholar

47 

Bayar E, Bennett PR, Chan D, Sykes L and MacIntyre DA: The pregnancy microbiome and preterm birth. Semin Immunopathol. 42:487–499. 2020.PubMed/NCBI View Article : Google Scholar

48 

Andrews WW, Goldenberg RL, Hauth JC, Cliver SP, Copper R and Conner M: Interconceptional antibiotics to prevent spontaneous preterm birth: A randomized clinical trial. Am J Obstet Gynecol. 194:617–623. 2006.PubMed/NCBI View Article : Google Scholar

49 

Abedini E, Khodadadi E, Zeinalzadeh E, Moaddab SR, Asgharzadeh M, Mehramouz B, Dao S and Samadi Kafil H: A comprehensive study on the antimicrobial properties of resveratrol as an alternative therapy. Evid Based Complement Alternat Med. 2021(8866311)2021.PubMed/NCBI View Article : Google Scholar

50 

Ma DSL, Tan LTH, Chan KG, Yap WH, Pusparajah P, Chuah LH, Ming LC, Khan TM, Lee LH and Goh BH: Resveratrol-Potential antibacterial agent against foodborne pathogens. Front Pharmacol. 9(102)2018.PubMed/NCBI View Article : Google Scholar

51 

Roberts RA, Laskin DL, Smith CV, Robertson FM, Allen EMG, Doorn JA and Slikker W: Nitrative and oxidative stress in toxicology and disease. Toxicol Sci. 112:4–16. 2009.PubMed/NCBI View Article : Google Scholar

52 

Meng T, Xiao D, Muhammed A, Deng J, Chen L and He J: Anti-Inflammatory action and mechanisms of resveratrol. Molecules. 26(229)2021.PubMed/NCBI View Article : Google Scholar

53 

Gómez-Chávez F, Correa D, Navarrete-Meneses P, Cancino-Diaz JC, Cancino-Diaz ME and Rodríguez-Martínez S: NF-κB and its regulators during pregnancy. Front Immunol. 12(679106)2021.PubMed/NCBI View Article : Google Scholar

54 

Malaguarnera. Influence of resveratrol on the immune response. Nutrients. 11(946)2019.PubMed/NCBI View Article : Google Scholar

55 

Liu T, Zhang L, Joo D and Sun SC: NF-κB signaling in inflammation. Signal Transduct Target Ther. 2(17023)2017.PubMed/NCBI View Article : Google Scholar

56 

Klawitter M, Hakozaki M, Kobayashi H, Krupkova O, Quero L, Ospelt C, Gay S, Hausmann O, Liebscher T, Meier U, et al: Expression and regulation of toll-like receptors (TLRs) in human intervertebral disc cells. Eur Spine J. 23:1878–1891. 2014.PubMed/NCBI View Article : Google Scholar

57 

Lappas M, Mitton A, Lim R, Barker G, Riley C and Permezel M: SIRT1 Is a novel regulator of key pathways of human labor1. Biol Reprod. 84:167–178. 2011.PubMed/NCBI View Article : Google Scholar

58 

Manna SK, Mukhopadhyay A and Aggarwal BB: Resveratrol suppresses TNF-induced activation of nuclear transcription factors NF-kappa B, activator protein-1, and apoptosis: Potential role of reactive oxygen intermediates and lipid peroxidation. J Immunol. 164:6509–6519. 2000.PubMed/NCBI View Article : Google Scholar

59 

Zhu X, Liu Q, Wang M, Liang M, Yang X, Xu X, Zou H and Qiu J: Activation of sirt1 by resveratrol inhibits TNF-α induced inflammation in fibroblasts. PLoS One. 6(e27081)2011.PubMed/NCBI View Article : Google Scholar

60 

Taguchi A, Yamashita A, Kawana K, Nagamatsu T, Furuya H, Inoue E, Osuga Y and Fujii T: Recent progress in therapeutics for inflammation-associated preterm birth: A review. Reprod Sci. 24:7–18. 2017.PubMed/NCBI View Article : Google Scholar

61 

Taguchi A, Wada-Hiraike O, Kawana K, Koga K, Yamashita A, Shirane A, Urata Y, Kozuma S, Osuga Y and Fujii T: Resveratrol suppresses inflammatory responses in endometrial stromal cells derived from endometriosis: A possible role of the sirtuin 1 pathway. J Obstet Gynaecol Res. 40:770–778. 2014.PubMed/NCBI View Article : Google Scholar

62 

Vidal MS, Lintao RCV, Severino MEL, Tantengco OAG and Menon R: Spontaneous preterm birth: Involvement of multiple feto-maternal tissues and organ systems, differing mechanisms, and pathways. Front Endocrinol (Lausanne). 13(1015622)2022.PubMed/NCBI View Article : Google Scholar

63 

Wood EM, Hornaday KK and Slater DM: Prostaglandins in biofluids in pregnancy and labour: A systematic review. PLoS One. 16(e0260115)2021.PubMed/NCBI View Article : Google Scholar

64 

Yamashita A, Kawana K, Tomio K, Taguchi A, Isobe Y, Iwamoto R, Masuda K, Furuya H, Nagamatsu T, Nagasaka K, et al: Increased tissue levels of omega-3 polyunsaturated fatty acids prevents pathological preterm birth. Sci Rep. 3(3113)2013.PubMed/NCBI View Article : Google Scholar

65 

Li WJ, Lu JW, Zhang CY, Wang WS, Ying H, Myatt L and Sun K: PGE2 vs PGF2α in human parturition. Placenta. 104:208–219. 2021.PubMed/NCBI View Article : Google Scholar

66 

Alfirevic Z, Keeney E, Dowswell T, Welton NJ, Dias S, Jones LV, Navaratnam K and Caldwell DM: Labour induction with prostaglandins: A systematic re view and network meta-analysis. BMJ. 350(h217)2015.PubMed/NCBI View Article : Google Scholar

67 

Sykes L, MacIntyre DA, Teoh TG and Bennett PR: Anti-inflammatory prostaglandins for the prevention of preterm labour. Reproduction. 148:R29–R40. 2014.PubMed/NCBI View Article : Google Scholar

68 

Challis JR, Lockwood CJ, Myatt L, Norman JE, Strauss JF and Petraglia F: Inflammation and Pregnancy. Reprod Sci. 16:206–215. 2009.PubMed/NCBI View Article : Google Scholar

69 

Zhang Y, Desai A, Yang SY, Bae KB, Antczak MI, Fink SP, Tiwari S, Willis JE, Williams NS, Dawson DM, et al: Inhibition of the prostaglandin-degrading enzyme 15-PGDH potentiates tissue regeneration. Science. 348(aaa2340)2015.PubMed/NCBI View Article : Google Scholar

70 

Mitchell MD, Rice GE, Vaswani K, Kvaskoff D and Peiris HN: Differential regulation of eicosanoid and endocannabinoid production by inflammatory mediators in human choriodecidua. PLoS One. 11(e0148306)2016.PubMed/NCBI View Article : Google Scholar

71 

Maia J, Fonseca B, Teixeira N and Correia-da-Silva G: The fundamental role of the endocannabinoid system in endometrium and placenta: Implications in pathophysiological aspects of uterine and pregnancy disorders. Hum Reprod Update. 26:586–602. 2020.PubMed/NCBI View Article : Google Scholar

72 

Nallendran V, Lam PM, Marczylo TH, Bankart MJ, Taylor AH, Taylor DJ and Konje JC: The plasma levels of the endocannabinoid, anandamide, increase with the induction of labour. BJOG. 117:863–869. 2010.PubMed/NCBI View Article : Google Scholar

73 

Bariani MV, Domínguez Rubio AP, Cella M, Burdet J, Franchi AM and Aisemberg J: Role of the endocannabinoid system in the mechanisms involved in the LPS-induced preterm labor. Reproduction. 150:463–472. 2015.PubMed/NCBI View Article : Google Scholar

74 

Komarnytsky S, Rathinasabapathy T, Wagner C, Metzger B, Carlisle C, Panda C, Le Brun-Blashka S, Troup JP and Varadharaj S: Endocannabinoid system and its regulation by polyunsaturated fatty acids and full spectrum hemp oils. Int J Mol Sci. 22(5479)2021.PubMed/NCBI View Article : Google Scholar

75 

Rava A and Trezza V: Emerging roles of endocannabinoids as key lipid mediators for a successful pregnancy. Int J Mol Sci. 24(5220)2023.PubMed/NCBI View Article : Google Scholar

76 

Dennedy MC, Friel AM, Houlihan DD, Broderick VM, Smith T and Morrison JJ: Cannabinoids and the human uterus during pregnancy. Am J Obstet Gynecol. 190:2–9. 2004.PubMed/NCBI View Article : Google Scholar

77 

Mitchell MD, Sato TA, Wang A, Keelan JA, Ponnampalam AP and Glass M: Cannabinoids stimulate prostaglandin production by human gestational tissues through a tissue- and CB1-receptor-specific mechanism. Am J Physiol Endocrinol Metab. 294:E352–E356. 2008.PubMed/NCBI View Article : Google Scholar

78 

Kozakiewicz ML, Grotegut CA and Howlett AC: Endocannabinoid system in pregnancy maintenance and labor: A mini-review. Front Endocrinol (Lausanne). 12(699951)2021.PubMed/NCBI View Article : Google Scholar

79 

Booker L, Kinsey SG, Abdullah RA, Blankman JL, Long JZ, Ezzili C, Boger DL, Cravatt BF and Lichtman AH: The fatty acid amide hydrolase (FAAH) inhibitor PF-3845 acts in the nervous system to reverse LPS-induced tactile allodynia in mice. Br J Pharmacol. 165:2485–2496. 2012.PubMed/NCBI View Article : Google Scholar

80 

Sun X, Deng W, Li Y, Tang S, Leishman E, Bradshaw HB and Dey SK: Sustained endocannabinoid signaling compromises decidual function and promotes inflammation-induced preterm birth. J Biol Chem. 291:8231–8240. 2016.PubMed/NCBI View Article : Google Scholar

81 

Donvito G, Nass SR, Wilkerson JL, Curry ZA, Schurman LD, Kinsey SG and Lichtman AH: The endogenous cannabinoid system: A budding source of targets for treating inflammatory and neuropathic pain. Neuropsychopharmacology. 43:52–79. 2018.PubMed/NCBI View Article : Google Scholar

82 

Connor M, Vaughan CW and Vandenberg RJ: N-Acyl amino acids and N-acyl neurotransmitter conjugates: Neuromodulators and probes for new drug targets. Br J Pharmacol. 160:1857–1871. 2010.PubMed/NCBI View Article : Google Scholar

83 

Godlewski G, Offertáler L, Osei-Hyiaman D, Mo FM, Harvey-White J, Liu J, Davis MI, Zhang L, Razdan RK, Milman G, et al: The endogenous brain constituent N-Arachidonoyl l-Serine Is an activator of large conductance Ca 2+-Activated K+ channels. J Pharmacol Exp Ther. 328:351–361. 2009.PubMed/NCBI View Article : Google Scholar

84 

Guo J, Williams DJ and Ikeda SR: N-Arachidonoyl L-Serine, a putative endocannabinoid, alters the activation of N-Type Ca2+ Channels in sympathetic neurons. J Neurophysiol. 100:1147–151. 2008.PubMed/NCBI View Article : Google Scholar

85 

Grazia Cascio M, Minassi A, Ligresti A, Appendino G, Burstein S and Di Marzo V: A structure-activity relationship study on N-arachidonoyl-amino acids as possible endogenous inhibitors of fatty acid amide hydrolase. Biochem Biophys Res Commun. 314:192–196. 2004.PubMed/NCBI View Article : Google Scholar

86 

Bachkangi P, Taylor AH, Bari M, Maccarrone M and Konje JC: Prediction of preterm labour from a single blood test: The role of the endocannabinoid system in predicting preterm birth in high-risk women. Eur J Obstet Gynecol Reprod Biol. 243:1–6. 2019.PubMed/NCBI View Article : Google Scholar

87 

Deutsch DG: A personal retrospective: Elevating anandamide (AEA) by targeting fatty acid amide hydrolase (FAAH) and the fatty acid binding proteins (FABPs). Front Pharmacol. 7(370)2016.PubMed/NCBI View Article : Google Scholar

88 

Liu J, Bátkai S, Pacher P, Harvey-White J, Wagner JA, Cravatt BF, Gao B and Kunos G: Lipopolysaccharide induces anandamide synthesis in macrophages via CD14/MAPK/phosphoinositide 3-kinase/NF-kappaB independently of platelet-activating factor. J Biol Chem. 278:45034–45039. 2003.PubMed/NCBI View Article : Google Scholar

89 

Nagarkatti P, Pandey R, Rieder SA, Hegde VL and Nagarkatti M: Cannabinoids as novel anti-inflammatory drugs. Future Med Chem. 1:1333–1349. 2009.PubMed/NCBI View Article : Google Scholar

90 

Maccarrone M, Valensise H, Bari M, Lazzarin N, Romanini C and Finazzi-Agrò A: Relation between decreased anandamide hydrolase concentrations in human lymphocytes and miscarriage. Lancet. 355:1326–1329. 2000.PubMed/NCBI View Article : Google Scholar

91 

Dong C, Chen J, Harrington A, Vinod KY, Hegde ML and Hegde VL: Cannabinoid exposure during pregnancy and its impact on immune function. Cell Mol Life Sci. 76:729–743. 2019.PubMed/NCBI View Article : Google Scholar

92 

Hirota Y, Cha J, Yoshie M, Daikoku T and Dey SK: Heightened uterine mammalian target of rapamycin complex 1 (mTORC1) signaling provokes preterm birth in mice. Proc Natl Acad Sci USA. 108:18073–18078. 2011.PubMed/NCBI View Article : Google Scholar

93 

Wolfson ML, Aisemberg J, Correa F and Franchi AM: Peripheral blood mononuclear cells infiltration downregulates decidual FAAH activity in an LPS-Induced embryo resorption model. J Cell Physiol. 232:1441–1447. 2017.PubMed/NCBI View Article : Google Scholar

94 

Cox LS and Redman C: The role of cellular senescence in ageing of the placenta. Placenta. 52:139–145. 2017.PubMed/NCBI View Article : Google Scholar

95 

Singh M, Parent S, Leblanc V and Asselin E: Resveratrol modulates the expression of PTGS2 and cellular proliferation in the normal rat endometrium in an AKT-Dependent Manner1. Biol Reprod. 84:1045–1052. 2011.PubMed/NCBI View Article : Google Scholar

96 

Rajkovic J, Djokic V, Gostimirovic M, Gojkovic-Bukarica L, Martorell M, Sharifi-Rad J and Novakovic R: Potassium channels on smooth muscle as a molecular target for plant-derived Resveratrol. Cell Mol Biol (Noisy-le-Grand). 66:133–144. 2020.PubMed/NCBI

97 

Carreiro JN, Magnani M, Jobling P, van Helden DF, Nalivaiko E and Braga VA: Resveratrol restores uterine contractions during hypoxia by blockade of ATP-sensitive potassium channels. J Funct Foods. 33:307–313. 2017.

98 

Lim R, Barker G and Lappas M: Activation of AMPK in human fetal membranes alleviates infection-induced expression of pro-inflammatory and pro-labour mediators. Placenta. 36:454–462. 2015.PubMed/NCBI View Article : Google Scholar

99 

Novakovic R, Ilic B, Beleslin-Cokic B, Radunovic N, Heinle H, Scepanovic R and Gojkovic-Bukarica L: The effect of resveratrol on contractility of non-pregnant rat uterus: The contribution of K(+) channels. J Physiol Pharmacol. 64:795–805. 2013.PubMed/NCBI

100 

Okawa T, Longo M, Vedernikov YP, Chwalisz K, Saade GR and Garfield RE: Role of nucleotide cyclases in the inhibition of pregnant rat uterine contractions by the openers of potassium channels. Am J Obstet Gynecol. 182:913–918. 2000.PubMed/NCBI View Article : Google Scholar

101 

Brainard AM, Korovkina VP and England SK: Potassium channels and uterine function. Semin Cell Dev Biol. 18:332–339. 2007.PubMed/NCBI View Article : Google Scholar

102 

Zheng S, Feng Q, Cheng J and Zheng J: Maternal resveratrol consumption and its programming effects on metabolic health in offspring mechanisms and potential implications. Biosci Rep. 38(BSR20171741)2018.PubMed/NCBI View Article : Google Scholar

103 

Azachi M, Yatuv R, Katz A, Hagay Y and Danon A: A novel red grape cells complex: Health effects and bioavailability of natural resveratrol. Int J Food Sci Nutr. 65:848–855. 2014.PubMed/NCBI View Article : Google Scholar

104 

Wang P and Sang S: Metabolism and pharmacokinetics of resveratrol and pterostilbene. BioFactors. 44:16–25. 2018.PubMed/NCBI View Article : Google Scholar

105 

Pollack RM and Crandall JP: Resveratrol: Therapeutic potential for improving cardiometabolic health. Am J Hypertens. 26:1260–1268. 2013.PubMed/NCBI View Article : Google Scholar

106 

Shrikanta A, Kumar A and Govindaswamy V: Resveratrol content and antioxidant properties of underutilized fruits. J Food Sci Technol. 52:383–390. 2015.PubMed/NCBI View Article : Google Scholar

107 

Cione E, La Torre C, Cannataro R, Caroleo MC, Plastina P and Gallelli L: Quercetin, Epigallocatechin gallate, curcumin, and resveratrol: From dietary sources to human MicroRNA modulation. Molecules. 25(63)2019.PubMed/NCBI View Article : Google Scholar

108 

Weiskirchen S and Weiskirchen R: Resveratrol: How much wine do you have to drink to stay healthy? Adv Nutr. 7:706–718. 2016.PubMed/NCBI View Article : Google Scholar

109 

Kuršvietienė L, Stanevičienė I, Mongirdienė A and Bernatonienė J: Multiplicity of effects and health benefits of resveratrol. Medicina (Kaunas). 52:148–155. 2016.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

March-April 2024
Volume 6 Issue 2

Print ISSN: 2632-2900
Online ISSN:2632-2919

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Habiburrahman M, Rakasiwi MI and Putra AB: Promising benefit of resveratrol in preventing preterm birth: A systematic review. World Acad Sci J 6: 10, 2024
APA
Habiburrahman, M., Rakasiwi, M.I., & Putra, A.B. (2024). Promising benefit of resveratrol in preventing preterm birth: A systematic review. World Academy of Sciences Journal, 6, 10. https://doi.org/10.3892/wasj.2024.225
MLA
Habiburrahman, M., Rakasiwi, M. I., Putra, A. B."Promising benefit of resveratrol in preventing preterm birth: A systematic review". World Academy of Sciences Journal 6.2 (2024): 10.
Chicago
Habiburrahman, M., Rakasiwi, M. I., Putra, A. B."Promising benefit of resveratrol in preventing preterm birth: A systematic review". World Academy of Sciences Journal 6, no. 2 (2024): 10. https://doi.org/10.3892/wasj.2024.225