Next Article in Journal
Focus of Sustainable Healthy Diets Interventions in Primary School-Aged Children: A Systematic Review
Next Article in Special Issue
Raspberry Leaves and Extracts-Molecular Mechanism of Action and Its Effectiveness on Human Cervical Ripening and the Induction of Labor
Previous Article in Journal
Muscle Quality Index in Morbidly Obesity Patients Related to Metabolic Syndrome Markers and Cardiorespiratory Fitness
Previous Article in Special Issue
Reprogramming Effects of Postbiotic Butyrate and Propionate on Maternal High-Fructose Diet-Induced Offspring Hypertension
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Understanding PPARγ and Its Agonists on Trophoblast Differentiation and Invasion: Potential Therapeutic Targets for Gestational Diabetes Mellitus and Preeclampsia

1
Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
2
Department of Biology, Texas A&M University, College Station, TX 77843, USA
3
Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(11), 2459; https://doi.org/10.3390/nu15112459
Submission received: 17 April 2023 / Revised: 16 May 2023 / Accepted: 22 May 2023 / Published: 25 May 2023
(This article belongs to the Special Issue Nutrition and Supplements during Pregnancy)

Abstract

:
The increasing incidence of pregnancy complications, particularly gestational diabetes mellitus (GDM) and preeclampsia (PE), is a cause for concern, as they can result in serious health consequences for both mothers and infants. The pathogenesis of these complications is still not fully understood, although it is known that the pathologic placenta plays a crucial role. Studies have shown that PPARγ, a transcription factor involved in glucose and lipid metabolism, may have a critical role in the etiology of these complications. While PPARγ agonists are FDA-approved drugs for Type 2 Diabetes Mellitus, their safety during pregnancy is not yet established. Nevertheless, there is growing evidence for the therapeutic potential of PPARγ in the treatment of PE using mouse models and in cell cultures. This review aims to summarize the current understanding of the mechanism of PPARγ in placental pathophysiology and to explore the possibility of using PPARγ ligands as a treatment option for pregnancy complications. Overall, this topic is of great significance for improving maternal and fetal health outcomes and warrants further investigation.

1. Introduction

Pregnancy can lead to complications that pose serious risks to both the mother and infant during pregnancy, labor, and postpartum. These complications typically arise from conditions unique to pregnancy. Alarmingly, there has been a 16.4% increase in the incidence of pregnancy complications between 2014 and 2018, with gestational diabetes mellitus (GDM) increasing by 16.6% and preeclampsia (PE) increasing by 19% [1]. Although it is recognized that the pathologic placenta is the root cause of many pregnancy complications, the exact mechanism is not yet fully understood. Recent clinical studies have suggested that genetic analysis, such as Peroxisome proliferator-activated receptor-γ (PPARγ) as a transcription factor, can offer a novel approach to diagnosis and prediction [2]. PPARγ is crucial for metabolism homeostasis, adipocyte differentiation, and the immune system. Research has revealed significant associations between certain PPARγ gene variations and PE, underscoring the importance of PPARγ in the development of this condition. Notably, PE is more prevalent in women with hyperglycemia, a well-known risk factor [3,4,5]. Women with diabetes are at least twice as likely to develop PE, with around 50% of diabetic pregnancies experiencing hypertensive disorders of pregnancy (HDP), particularly those with pre-existing diabetes and poor glycemic control [6,7,8,9]. Considering the therapeutic potential of PPARγ agonists, which are FDA-approved for Type 2 Diabetes Mellitus, it becomes evident that these agents hold promise for preeclampsia treatment, particularly in patients with risk factors such as hyperglycemia. Therefore, this review aims to provide a comprehensive overview of the current research on the mechanisms of PPARγ and the effects of PPARγ agonists on placenta pathophysiology. Such understanding paves the way for precision medicine strategies to prevent or mitigate the risk of PE, taking into account individual factors such as race, genetics, and maternal risk factors.
To examine the role of PPARγ in GDM and PE, with a particular focus on placental pathophysiology, this review conducted a comprehensive search using the PubMed database. Only original research and scientific abstracts published between 2005 and 2022 that investigated the role of PPARγ in GDM and PE were included. The search terms used were “peroxisome proliferator-activated receptor-gamma”, “PPARγ”, “PPAR gamma”, “gestational diabetes mellitus” and “preeclampsia”. Studies involving other PPARs and other pregnancy complications such as infertility, hypertensive pregnancy, and polycystic ovarian syndrome were excluded (Figure 1).

2. Peroxisome Proliferator-Activated Receptor-γ

The Peroxisome Proliferator-Activated Receptor-γ (PPARγ) is a PPAR subfamily member consisting of two isoforms. PPARγ1 is encoded by mRNA PPARγ1, PPARγ3, and PPARγ4, while PPARγ2 is translated from mRNA PPARγ2 [10]. PPARγ1 is broadly expressed in various tissues including adipose tissue, the liver, colon, heart, epithelial cells, and skeletal muscle, and is also found in immune cells such as monocytes/macrophages, dendritic cells, and T lymphocytes [11]. On the other hand, PPARγ2 contains 28 additional amino acids and is primarily found in adipose tissue. Both isoforms are highly expressed in reproductive organs such as the placenta, testis, and ovary [12].
PPARγ is a ligand-dependent transcription factor, meaning it can be regulated by agonists and antagonists. It acts as a sensor for different fatty acid types, also known as a lipid sensor. In addition to endogenous ligands, synthetic ligands are widely used in clinical practice and in vitro studies to modulate PPARγ [13,14]. A summary of reported PPARγ ligands is provided in Table 1.
PPARγ, in addition to its well-established roles in lipid metabolism and adipocyte differentiation, has also been shown to be essential in regulating insulin resistance, glucose metabolism, immunity, as well as cell biology, including cell differentiation [45,46,47]. The TZD family comprises FDA-approved drugs used for treating Type 2 Diabetes Mellitus [13]. Beyond its function in immunology and maintaining energy homeostasis, PPARγ is also indispensable for the early development of the conceptus as early as E10. Its critical role in development seems to be particularly important in the placenta [48]. This review primarily focuses on the function of PPARγ in trophoblast differentiation and invasion, as well as its relationship with pregnancy complications, including GDM and PE.

3. PPARγ Functions in the Placenta and Trophoblasts

PPARγ is highly expressed in human placentas, particularly in syncytiotrophoblasts, cytotrophoblasts, and extravillous trophoblasts (EVTs) [49,50]. Its expression in the placenta is associated with infant birth weight. Placentas from small-for-gestational-age (SGA) infants were found to have lower expression of PPARγ, whereas placentas from average-for-gestational-age and large-for-gestational-age infants showed a nearly 2-fold higher expression of PPARγ compared with that from SGA infants [51]. These findings suggest that PPARγ may play a role in regulating fetal growth and development in the placenta.
Recent in vitro studies have shown that PPARγ is associated with trophoblast migration and invasion, although its exact role in these processes appears to be paradoxical. Some studies have reported that PPARγ inhibits trophoblast invasion in human primary cultures of EVTs [52,53,54]. One proposed mechanism is through the repression of pregnancy-associated plasma protein A, which reduces insulin-like growth factor (IGF) availability and limits trophoblast invasion [55,56]. Additionally, heme oxygenase-1 (HO-1) has been reported to negatively regulate trophoblast motility through the up-regulation of PPARγ [57]. Furthermore, PPARγ has been shown to inhibit trophoblast migration through its interaction with endocrine gland-derived vascular endothelial growth factor (EG-VEGF), a placental angiogenic factor [58]. Some studies have also reported that rosiglitazone, a PPARγ agonist, blocked lipopolysaccharide (LPS)-induced invasion in human first-trimester trophoblast cell lines [59].
However, more recent studies have suggested that PPARγ may promote trophoblast migration. Activated PPARγ/RXRα heterodimer by IL-17 was found to promote proliferation, migration, and invasion in HTR8/SVneo, a trophoblast cell line [60]. Furthermore, pioglitazone, which increases PPARγ expression, was shown to stimulate EVT migration by promoting IGF signaling [56]. In addition, mutations on the ligand-binding domain of PPARγ have been found to significantly suppress migration in the primary villous cytotrophoblasts [61]. These findings suggest that the role of PPARγ in trophoblast migration and invasion may be complex, and further research is needed to fully understand its mechanisms and effects in these processes.
PPARγ has also been identified as a regulator of trophoblast differentiation. In the BeWo cell model, blocking PPARγ activity has been shown to induce cell proliferation but suppress the differentiation [62]. In human placenta explants, PPARγ/RXRα heterodimers have been found to promote cytotrophoblast differentiation into syncytiotrophoblasts [63], which is a key event in placental development. In PPARγ-deficient mouse placentas, diminished expression of several trophoblast differentiation markers, such as Tpbpα and Mash2, as well as the abnormal spatial expression of glial cell missing 1 (GCM1), a transcription factor important for syncytiotrophoblast differentiation, were observed [64]. In addition, oral administration of troglitazone, a PPARγ agonist, was found to enhance cytotrophoblast differentiation into syncytiotrophoblasts [65]. PPARγ also promotes the differentiation of syncytiotrophoblasts, but not trophoblast giant cells (TGCs) in the mouse labyrinth, which is the region of the placenta where nutrient exchange occurs [66]. On the other hand, rosiglitazone, another PPARγ agonist, has been reported to reduce TGC differentiation while inducing GCM1 expression [67], suggesting that the role of PPARγ in trophoblast differentiation may be complex and dependent on the specific cell type. Further research is needed to fully elucidate the mechanisms and effects of PPARγ in trophoblast differentiation.

4. Genome-Wide Association Studies (GWAS) Suggested That PPARγ Is Associated with Preeclampsia and Gestational Diabetes Mellitus

PPARγ single nucleotide polymorphisms (SNPs) are associated with increased susceptibility to pregnancy-related diseases, including GDM and PE. The rs201018 and C1431T variants of PPARγ have been reported to be significantly associated with susceptibility to PE in different populations [54,55], with the rs201018 polymorphism showing a correlation with the incidence of PE in the Chinese population [55], and the C1431T polymorphism is associated with PE occurrence in the French population [54].
In addition to PE, PPARγ SNPs have also been associated with the incidence of GDM. The Pro12Ala polymorphism of PPARγ is one of the dominant variants associated with GDM susceptibility, as reported in several meta-analyses [56,57,58]. However, there are conflicting findings regarding the role of Pro12Ala in GDM, with some studies suggesting a protective role against GDM in certain populations, such as the Filipino population, while others suggest that it may exacerbate insulin resistance by elevating serum resistin levels [59]. Besides Pro12Ala, the rs1801282 variant of PPARγ is associated with increased GDM incidence in Russian [60] and Asian [61] populations, but not in the Brazilian population [62]. Recent studies have also suggested that the PPARΓ (rs1801282) variant may be a significant risk factor for the development of PE in women with GDM in the Russian population [63]. These findings highlight the potential role of PPARγ SNPs in modulating the risk of pregnancy-related diseases, although further research is needed to fully understand the underlying mechanisms and implications of these genetic variants. Large epidemiological studies of the population considering different demographic information will be essential to establish a comprehensive understanding of PPARγ SNPs in modulating the risk of pregnancy-related diseases.

5. The Role of PPARγ in Preeclampsia

PE is a serious pregnancy complication that affects 5–7% of pregnancies worldwide [68] and is responsible for over 500,000 maternal and fetal deaths each year [69]. PE is considered the leading cause of maternal morbidity and mortality in the United States, accounting for 16% of maternal deaths [70]. Women diagnosed with PE are at increased risk of adverse outcomes not only for themselves, but also for their babies in the future.
PE can have serious consequences for both the mother and the baby. If the placenta is not implanted correctly, it can result in inadequate blood flow to the placenta, leading to fetal growth restriction or intrauterine growth restriction (IUGR) [71]. This can result in babies being born with low birth weight and other health complications. Additionally, women who have experienced PE have an increased risk of developing cardiovascular disease later in life, as well as their children [72].
Diagnosis of PE typically involves measuring blood pressure, with systolic blood pressure greater than 140 mmHg and diastolic blood pressure greater than 90 mmHg, along with proteinuria [69]. PE is usually diagnosed after 20 weeks of pregnancy, but it can occur in both early and late pregnancy. The development of PE is believed to involve two interconnected stages. The first stage is usually asymptomatic and is initiated by inadequate placental circulation due to abnormal placental implantation and/or reduced blood flow to the placenta. This can result in placental ischemia, or reduced blood flow to the placenta, due to aberrant failed remodeling of spiral arteries and impaired vascularization [73]. If placental perfusion remains compromised, it can progress to the second stage of PE, which is characterized by clinical manifestations such as high blood pressure and other symptoms [74]. Understanding the pathogenesis of PE is complex, and ongoing research is needed to further elucidate the underlying mechanisms and develop effective prevention and treatment strategies.

5.1. PAPRγ in the Pathogenesis of PE

The role of PPARγ in the pathogenesis of PE is still not fully understood, but studies have shown conflicting results regarding its expression levels in PE placentas. While some studies have reported increased expression of PPARγ in PE placentas [75,76], most studies have found decreased expression of PPARγ in PE placentas [76,77,78,79], while the expression of PPARγ increased in the blood serum of PE patients [80].
One mechanism that has been explored is the modulation of 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2), a gene that helps in the maintenance of cortisol levels in the placenta. PPARγ was found to positively correlate with 11β-HSD2 expression in PE placentas, and treatment with rosiglitazone, a PPARγ agonist, increased the expression of 11β-HSD2 in placental explants, while treatment with GW9662, a PPARγ antagonist, decreased the expression of 11β-HSD2. Interestingly, the effect of PPARγ ligands was blocked when specificity protein 1 (Sp-1) was knocked out [79]. Another transcription factor that has been implicated in the pathogenesis of PE is GCM1, which regulates trophoblast differentiation [81]. GCM1 expression is significantly downregulated in PE placentas, and depletion of GCM1 in normal placental explants elevated the secretion of soluble Fms-like tyrosine kinase 1 (sFlt-1), a marker for PE. Treatment with rosiglitazone increased GCM1 expression, while treatment with T0070907, a PPARγ antagonist, reduced GCM1 expression [78].
PPARγ has also been shown to participate in regulating sFlt-1 secretion through nuclear factor erythroid 2-related factor 2 (Nrf2) [82]. Procyanidin B2, a natural compound, reduced sFlt-1 secretion and restored the migration capacity of trophoblasts in placental explants from PE pregnancies by activating Nrf2, which bound to the promoter region of PPARγ and enhanced its transcriptional activity.
In addition, studies have shown that PPARγ and angiopoietin-like protein 4 (ANGPTL4), which is associated with fat metabolism and vessel formation, are reduced in PE placentas compared with control placentas [83]. Treatment with rosiglitazone upregulated the expression and secretion of ANGPTL4 in placental explants [83], suggesting a potential interaction between ANGPTL4 and PPARγ in the pathogenesis of PE.
PPARγ has been implicated in regulating epigenetic modifications in the placenta, specifically histone methylation and acetylation. Epigenetic modifications play a crucial role in regulating gene expression and can impact various cellular processes, including trophoblast invasion and migration [84]. The co-expression of PPARγ with histone markers such as H3K4me3 and H3K9ac is upregulated in preeclamptic placenta [85], suggesting a potential involvement of PPARγ in placental epigenetic regulation in the context of PE. Treatment with ciglitazone, a PPARγ agonist, has been shown to restore the levels of these histone modifications, while treatment with T0070907, a PPARγ antagonist, further induced an increased level of H3K4me3 and H3K9ac [85]. This suggests that PPARγ may play a role in modulating placental epigenetic modifications in PE, although the exact mechanisms and implications of these epigenetic changes are still not fully understood and require further research. One of the potential reasons is the variation in cell populations and epigenetic profiles of placentas among individuals [86,87]. This requires large sample sizes to comprehensively determine the effect of epigenetic-modified PPARγ.
Overall, the role of PPARγ in the pathogenesis of PE is complex and still not fully understood, with conflicting findings in different studies. Further research is needed to elucidate the exact mechanisms and potential therapeutic implications of PPARγ in PE.

5.2. Effect of PAPRγ Ligands on PE in Rodent Models

Although human placental tissue and explants can provide valuable information about the function of PPARγ in PE, ethical concerns make it impossible to determine the effect of its ligands in human studies. Therefore, rodent models, such as mice and rats, have been widely used to study the molecular mechanisms of PE. One commonly used rodent model is the reduced uterine perfusion pressure (RUPP) model in pregnant rats, which involves creating an abdominal incision on E14.5 of gestation to mimic abnormal uteroplacental blood flow [88]. RUPP rats exhibit characteristics of PE, including high blood pressure, impaired vasorelaxation, and elevated ACR (albumin-to-creatinine ratio). Treatment of RUPP rats with rosiglitazone, a PPARγ agonist, has been shown to ameliorate hypertension, improve vasorelaxation, and reduce ACR [89]. Interestingly, this beneficial effect is blocked by an HO-1 (heme oxygenase-1) inhibitor, SnPP [89], suggesting that the regulatory role of PPARγ may be dependent on the HO-1 pathway.
On the other hand, treatment with T0070907, an antagonist of PPARγ, in mice has been shown to induce hallmark symptoms of PE, including hypertension, proteinuria, and fetal growth restriction [90,91]. These mice also displayed increased total placental sFlt-1, increased HO-1, and decreased VEGF, as well as decreased overall labyrinth trophoblast differentiation, which are parameters associated with PE [77].
Both animal models and human tissue studies have shown a decrease in PPARγ expression in the pathogenesis of induced or diagnosed PE, respectively. This suggests that PPARγ could be targeted by pharmacological interventions to potentially reduce the severity of PE in pregnant women diagnosed with the disease.

5.3. Potential Treatments of Preeclampsia Targeting PPARγ

Potential treatments for PE can target PPARγ or genes associated with PPARγ, leading to significant changes in PPARγ expression that positively impact women or result in mice displaying hallmark signs of the disease. Treatment with the PPARγ antagonist T0070907 induced PE-like symptoms in mice [91]. However, administration of aspirin reversed T0070907-induced changes in VEGF, sFlt, and MMP2 in both maternal blood and placental tissue, and increased the expression of PPARγ by inhibiting the cyclooxygenase (COX) pathway [90]. Interestingly, different doses of aspirin showed varying impacts on modulating PE, with a higher dose (20 mg/kg) exhibiting a more significant improvement in maternal blood pressure compared with a lower dose (10 mg/kg) [92]. Angiotensin, a vasodilator hypothesized to inhibit the COX-2 pathway [93], was also studied for its effects on PE. In a study using a rat model of PE (RUPP rat), treatment with angiotensin 1–7 elevated the expression of PPARγ, leading to a significant decrease in systolic blood pressure and other PE symptoms. The researchers speculated that angiotensin 1–7 may increase PPARγ expression by enhancing the actions of the endothelial nitric oxide synthase (eNOS) [94]. Overall, PPARγ appears to be a promising candidate for early diagnostic biomarkers and treatment targets for PE, but further studies are needed to elucidate the underlying mechanisms.

6. PPARγ Functions in Placentas from GDM

Gestational diabetes mellitus (GDM), a form of glucose intolerance that arises during pregnancy, affects approximately 7.6% of pregnancies in the US and is one of the most common obstetric complications [95]. According to a 2020 report by BlueCross BlueShield, the incidence of GDM has increased by 16.6% from 2014 to 2018 [1]. GDM can result in short-term and long-term complications for both the mother and the fetus. Short-term effects include fetal macrosomia (large birth weight), hypoglycemia, respiratory distress syndrome, and preterm birth. Later in life, both the mother and the child are at increased risk of developing Type 2 Diabetes Mellitus (T2DM) [96]. Additionally, GDM pregnancies can also lead to high-risk pregnancy complications such as PE and miscarriage [97,98]. Alarmingly, at least 20% of GDM pregnancies were reported to develop pregnancy-induced hypertension [8]. However, the diagnosis of GDM is challenging due to discrepant diagnosis criteria and a lack of noticeable symptoms [99].
As the intermediate transportation site between mother and fetus, placentas from GDM also displayed a pathophysiological change in the spiral artery and vasculature [100]. While the placenta does not require insulin as a glucose regulator since glucose is the primary energy source for both the placenta and the fetus, the placenta still expresses insulin receptors, making it sensitive to maternal hyperglycemia. This sensitivity positively correlates with fetal growth and macrosomia [101]. Currently, insulin, metformin, and insulin detemir are the only FDA-approved drugs for the treatment of GDM [102]. Interestingly, most gene expression alterations in GDM occur in the lipid pathway rather than the glucose pathway, and are mostly associated with dyslipidemia and insulin resistance [15]. Dysregulation of PPARγ, which is involved in fatty acid storage, glucose storage, and insulin sensitivity, has been implicated in GDM. Omega-3 supplements for women with GDM have been shown to decrease PPARγ expression and fasting blood glucose levels while increasing PPARγ expression in peripheral blood mononuclear cells (PBMCs) [103].
PPARγ agonists, such as rosiglitazone, are commonly used drugs to treat T2DM [14]. However, due to the inability of rosiglitazone to cross the placenta during early gestation [104] but potential transfer and metabolism by embryos during late gestation [105,106], studies have been conducted to assess its safety during pregnancy. Although limited data support its safety, several studies have shown that low doses of rosiglitazone do not have adverse effects on fetal development [107,108,109,110]. However, some studies have demonstrated that activation of PPARγ by rosiglitazone disrupts the vascularity and morphology of murine placenta [111,112]. Meanwhile, only limited research demonstrated its benefit in fetal development. One study showed that rosiglitazone ameliorated the adverse effects caused by nicotine exposure including abnormal cell death and suppressed angiogenesis in murine conceptus [113]. Therefore, it is crucial to further explore and understand the impact of PPARγ and its ligands in GDM, particularly in the placenta.

PPARγ Function in the Placentas of GDM Patients

The expression changes of PPARγ in the placentas of patients with GDM have yielded inconsistent results across various studies. While some studies have reported downregulation of placental PPARγ, This can result in placental ischemia, or reduced blood flow to the placenta, due to failed remodeling of spiral arteries and impaired vascularization expression in GDM patients compared with healthy control groups [76,114,115], particularly in syncytiotrophoblasts and EVT [116]; other studies have found increased gene expression of PPARγ in the placentas of GDM patients, including in Australian women with GDM [117] and in the trophoblast choriocarcinoma BeWo cell line under hyperglycemic conditions [118], which coincided with suppressed cell proliferation. Interestingly, PPARγ was also found to be upregulated in PBMCs of women with GDM [103], and leukocyte PPARγ mRNA levels were significantly higher in GDM patients compared with those of healthy patients [119].
In vitro studies have demonstrated that hyperglycemic conditions can impair placental vascularity through the repression of migration and viability [120]. PPARγ has been studied as a pharmaceutical target for T2DM prevention or treatment in GDM, and several pathways have been implicated in the PPARγ-mediated regulation of GDM from various aspects. For example, exogenous activation of PPARγ by 15dPGJ2 has been shown to prevent nitric oxide overproduction in the placenta of pre-gestational diabetic women [121]. Another study found that C1q/tumor necrosis factor-related protein 6 (CTRP6) interacts with PPARγ to regulate trophoblast function, with both CTRP6 and PPARγ being upregulated in high glucose-induced HTR-8/SVneo cells [122]. Depletion of CTRP6 rescued viability, invasion, and migration in HTR8/SVneo cells, while PPARγ overexpression blocked the protective effects of CTRP6 downregulation [122]. Additionally, a novel adipokine regulated by PPARγ in trophoblasts was discovered, with the administration of PPARγ agonists rosiglitazone and GW1929 elevating the expression of Chemerin and activation of the AKT/PI3K pathway [123]. Depletion of the chemerin receptor chemokine-like receptor 1 restrained this effect. Disulfide-bond A oxidoreductase-like protein (DsbA-L), an enzyme that regulates fat deposition, was also identified as a downstream target of PPARγ [124]. Rosiglitazone was found to improve insulin sensitivity through interaction with DsbA-L in the HTR-8/Svneo cell line, with upregulation of DsbA-L being crucial for the function of rosiglitazone in the PI3K-PKB/AKT pathway [125].

7. Future Directions and Conclusions

Currently, there is substantial evidence supporting the critical role of PPARγ in placental biology. PPARγ is highly expressed in trophoblasts, and its expression is influenced by maternal factors. While numerous in vitro studies have demonstrated that PPARγ regulates trophoblast differentiation and migration, there is still limited in vivo information that provides a comprehensive understanding of placental physiology and the complex pathophysiology of pregnancy. Several genes and chemicals have been identified to interact with PPARγ in trophoblasts, including COX, 11β-HSD2, angiotensin 1–7, HO-1, GCM1, Nrf2, ANGPTL4, CTRP6, DsbA-L, and aspirin. These interactions offer potential mechanistic insights into the function of PPARγ in the placenta. However, the current conclusion of PPARγ in placentas affected by GDM remains inconsistent. Determining whether PPARγ plays a protective role in the progression of GDM requires further in vivo studies, as systematic hyperglycemia may differ from that observed in in vitro models.
Furthermore, the ability of PPARγ ligands to modulate trophoblast function underscores the promising potential of PPARγ as a therapeutic target in the treatment of conditions. However, as depicted in Figure 2, apart from the extensively studied rosiglitazone, the effects of other ligands on preeclampsia require additional information, as each ligand may have distinct actions. Studies regarding the safety and effect of PPARγ ligands on treating GDM still need future attention. Continued research in this field holds great promise for advancing our understanding of PPARγ function in the placenta and developing innovative therapeutic strategies for managing complicated pregnancies.

Author Contributions

Writing—original draft preparation, Y.Q., D.B. and M.A.; writing—review and editing, L.X. and K.Z.; supervision, L.X. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded, in part, by a grant from the National Institute of Environmental Health Sciences (P30 ES029067).

Informed Consent Statement

Not applicable.

Data Availability Statement

Data available in a publicly accessible repository.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

PPARγPeroxisome Proliferator-Activated Receptor-γ
GDMGestational Diabetes Mellitus
PEPreeclampsia
EVTsExtravillous trophoblasts
GCM1Glial cell missing 1
T2DMType 2 Diabetes Mellitus
PBMCPeripheral blood mononuclear cells
TGCTrophoblast giant cells
COXCyclooxygenase
Nrf2Nuclear factor erythroid 2-related factor 2
VEGFVascular endothelial growth factor
sFltsoluble Fms-like tyrosine kinase
RUPPReduced uterine perfusion pressure
HO-1Heme oxygenase 1
TZDThiazolidinedione
LDLLow-density lipoproteins
BADGEBisphenol A diglycidyl ether
NFκBNuclear factor kappa-light-chain-enhancer
EETsEpoxyeicosatrienoic acids
15d-PGJ215-deoxy-Δ-12,14 prostaglandin J2
15-HETE15-Hydroxyeicosatetraenoic Acid
13-HODE13- hydroxyoctadecadienoic acid

References

  1. Trends in Pregnancy and Childbirth Complications in the U.S.; Blue cross Blue Shield: Chicago, IL, USA, 2020.
  2. Psilopatis, I.; Vrettou, K.; Fleckenstein, F.N.; Theocharis, S. The Role of Peroxisome Proliferator-Activated Receptors in Preeclampsia. Cells 2023, 12, 647. [Google Scholar] [CrossRef] [PubMed]
  3. McElwain, C.J.; McCarthy, F.P.; McCarthy, C.M. Gestational Diabetes Mellitus and Maternal Immune Dysregulation: What We Know So Far. Int. J. Mol. Sci. 2021, 22, 4261. [Google Scholar] [CrossRef] [PubMed]
  4. Plows, J.F.; Stanley, J.L.; Baker, P.N.; Reynolds, C.M.; Vickers, M.H. The Pathophysiology of Gestational Diabetes Mellitus. Int. J. Mol. Sci. 2018, 19, 3342. [Google Scholar] [CrossRef]
  5. McElwain, C.J.; Tuboly, E.; McCarthy, F.P.; McCarthy, C.M. Mechanisms of Endothelial Dysfunction in Pre-eclampsia and Gestational Diabetes Mellitus: Windows Into Future Cardiometabolic Health? Front. Endocrinol. 2020, 11, 655. [Google Scholar] [CrossRef]
  6. Longhitano, E.; Siligato, R.; Torreggiani, M.; Attini, R.; Masturzo, B.; Casula, V.; Matarazzo, I.; Cabiddu, G.; Santoro, D.; Versino, E.; et al. The Hypertensive Disorders of Pregnancy: A Focus on Definitions for Clinical Nephrologists. J. Clin. Med. 2022, 11, 3420. [Google Scholar] [CrossRef] [PubMed]
  7. Brown, M.A.; Magee, L.A.; Kenny, L.C.; Karumanchi, S.A.; McCarthy, F.P.; Saito, S.; Hall, D.R.; Warren, C.E.; Adoyi, G.; Ishaku, S.; et al. Hypertensive Disorders of Pregnancy: ISSHP Classification, Diagnosis, and Management Recommendations for International Practice. Hypertension 2018, 72, 24–43. [Google Scholar] [CrossRef]
  8. Sullivan, S.D.; Umans, J.G.; Ratner, R. Hypertension complicating diabetic pregnancies: Pathophysiology, management, and controversies. J. Clin. Hypertens. 2011, 13, 275–284. [Google Scholar] [CrossRef]
  9. Giorgione, V.; Jansen, G.; Kitt, J.; Ghossein-Doha, C.; Leeson, P.; Thilaganathan, B. Peripartum and Long-Term Maternal Cardiovascular Health After Preeclampsia. Hypertension 2023, 80, 231–241. [Google Scholar] [CrossRef]
  10. Aprile, M.; Ambrosio, M.R.; D’Esposito, V.; Beguinot, F.; Formisano, P.; Costa, V.; Ciccodicola, A. PPARG in Human Adipogenesis: Differential Contribution of Canonical Transcripts and Dominant Negative Isoforms. PPAR Res. 2014, 2014, 537865. [Google Scholar] [CrossRef]
  11. Fajas, L.; Auboeuf, D.; Raspe, E.; Schoonjans, K.; Lefebvre, A.M.; Saladin, R.; Najib, J.; Laville, M.; Fruchart, J.C.; Deeb, S.; et al. The organization, promoter analysis, and expression of the human PPARgamma gene. J. Biol. Chem. 1997, 272, 18779–18789. [Google Scholar] [CrossRef]
  12. Froment, P.; Gizard, F.; Defever, D.; Staels, B.; Dupont, J.; Monget, P. Peroxisome proliferator-activated receptors in reproductive tissues: From gametogenesis to parturition. J. Endocrinol. 2006, 189, 199–209. [Google Scholar] [CrossRef] [PubMed]
  13. Grygiel-Gorniak, B. Peroxisome proliferator-activated receptors and their ligands: Nutritional and clinical implications—A review. Nutr. J. 2014, 13, 17. [Google Scholar] [CrossRef] [PubMed]
  14. Villacorta, L.; Schopfer, F.J.; Zhang, J.; Freeman, B.A.; Chen, Y.E. PPARgamma and its ligands: Therapeutic implications in cardiovascular disease. Clin. Sci. 2009, 116, 205–218. [Google Scholar] [CrossRef] [PubMed]
  15. Lehrke, M.; Lazar, M.A. The many faces of PPARgamma. Cell 2005, 123, 993–999. [Google Scholar] [CrossRef] [PubMed]
  16. Makela, J.; Tselykh, T.V.; Kukkonen, J.P.; Eriksson, O.; Korhonen, L.T.; Lindholm, D. Peroxisome proliferator-activated receptor-gamma (PPARgamma) agonist is neuroprotective and stimulates PGC-1alpha expression and CREB phosphorylation in human dopaminergic neurons. Neuropharmacology 2016, 102, 266–275. [Google Scholar] [CrossRef] [PubMed]
  17. Brusotti, G.; Montanari, R.; Capelli, D.; Cattaneo, G.; Laghezza, A.; Tortorella, P.; Loiodice, F.; Peiretti, F.; Bonardo, B.; Paiardini, A.; et al. Betulinic acid is a PPARgamma antagonist that improves glucose uptake, promotes osteogenesis and inhibits adipogenesis. Sci. Rep. 2017, 7, 5777. [Google Scholar] [CrossRef] [PubMed]
  18. Rieusset, J.; Touri, F.; Michalik, L.; Escher, P.; Desvergne, B.; Niesor, E.; Wahli, W. A new selective peroxisome proliferator-activated receptor gamma antagonist with antiobesity and antidiabetic activity. Mol. Endocrinol. 2002, 16, 2628–2644. [Google Scholar] [CrossRef]
  19. Giaginis, C.; Spanopoulou, E.; Theocharis, S. PPAR-gamma signaling pathway in placental development and function: A potential therapeutic target in the treatment of gestational diseases. Expert. Opin. Ther. Targets 2008, 12, 1049–1063. [Google Scholar] [CrossRef]
  20. van Andel, M.; Heijboer, A.C.; Drent, M.L. Adiponectin and Its Isoforms in Pathophysiology. Adv. Clin. Chem. 2018, 85, 115–147. [Google Scholar] [CrossRef]
  21. Wang, Y.; Pan, Z.; Chen, F. Inhibition of PPARgamma by bisphenol A diglycidyl ether ameliorates dexamethasone-induced osteoporosis in a mouse model. J. Int. Med. Res. 2019, 47, 6268–6277. [Google Scholar] [CrossRef]
  22. Liu, Y.; Zhang, Y.; Schmelzer, K.; Lee, T.S.; Fang, X.; Zhu, Y.; Spector, A.A.; Gill, S.; Morisseau, C.; Hammock, B.D.; et al. The antiinflammatory effect of laminar flow: The role of PPARgamma, epoxyeicosatrienoic acids, and soluble epoxide hydrolase. Proc. Natl. Acad. Sci. USA 2005, 102, 16747–16752. [Google Scholar] [CrossRef] [PubMed]
  23. Rocchi, S.; Picard, F.; Vamecq, J.; Gelman, L.; Potier, N.; Zeyer, D.; Dubuquoy, L.; Bac, P.; Champy, M.F.; Plunket, K.D.; et al. A unique PPARgamma ligand with potent insulin-sensitizing yet weak adipogenic activity. Mol. Cell 2001, 8, 737–747. [Google Scholar] [CrossRef] [PubMed]
  24. Mukherjee, R.; Hoener, P.A.; Jow, L.; Bilakovics, J.; Klausing, K.; Mais, D.E.; Faulkner, A.; Croston, G.E.; Paterniti, J.R., Jr. A selective peroxisome proliferator-activated receptor-gamma (PPARgamma) modulator blocks adipocyte differentiation but stimulates glucose uptake in 3T3-L1 adipocytes. Mol. Endocrinol. 2000, 14, 1425–1433. [Google Scholar] [CrossRef]
  25. Li, J.; Guo, C.; Wu, J. 15-Deoxy-∆-(12,14)-Prostaglandin J2 (15d-PGJ2), an Endogenous Ligand of PPAR-gamma: Function and Mechanism. PPAR Res. 2019, 2019, 7242030. [Google Scholar] [CrossRef] [PubMed]
  26. Higgins, L.S.; Mantzoros, C.S. The Development of INT131 as a Selective PPARgamma Modulator: Approach to a Safer Insulin Sensitizer. PPAR Res. 2008, 2008, 936906. [Google Scholar] [CrossRef] [PubMed]
  27. Camp, H.S.; Chaudhry, A.; Leff, T. A novel potent antagonist of peroxisome proliferator-activated receptor gamma blocks adipocyte differentiation but does not revert the phenotype of terminally differentiated adipocytes. Endocrinology 2001, 142, 3207–3213. [Google Scholar] [CrossRef]
  28. Feng, L.; Liu, W.; Yang, J.; Wang, Q.; Wen, S. Effect of Hexadecyl Azelaoyl Phosphatidylcholine on Cardiomyocyte Apoptosis in Myocardial Ischemia-Reperfusion Injury: A Hypothesis. Med. Sci. Monit. 2018, 24, 2661–2667. [Google Scholar] [CrossRef]
  29. O’Connor-Semmes, R.O.B.I.N.; Mydlow, P.; Walker, A.; Clark, R.V. GI262570, A PPAR [Gamma] Agonist, Maintains Metabolic Improvements throughout 24 Hour Profiles In Type 2 Diabetic Patients. Diabetes 2000, 49, A119. [Google Scholar]
  30. Lee, G.; Elwood, F.; McNally, J.; Weiszmann, J.; Lindstrom, M.; Amaral, K.; Nakamura, M.; Miao, S.; Cao, P.; Learned, R.M.; et al. T0070907, a selective ligand for peroxisome proliferator-activated receptor gamma, functions as an antagonist of biochemical and cellular activities. J. Biol. Chem. 2002, 277, 19649–19657. [Google Scholar] [CrossRef]
  31. Saether, T.; Paulsen, S.M.; Tungen, J.E.; Vik, A.; Aursnes, M.; Holen, T.; Hansen, T.V.; Nebb, H.I. Synthesis and biological evaluations of marine oxohexadecenoic acids: PPARalpha/gamma dual agonism and anti-diabetic target gene effects. Eur. J. Med. Chem. 2018, 155, 736–753. [Google Scholar] [CrossRef]
  32. Carmona, M.C.; Louche, K.; Lefebvre, B.; Pilon, A.; Hennuyer, N.; Audinot-Bouchez, V.; Fievet, C.; Torpier, G.; Formstecher, P.; Renard, P.; et al. S 26948: A new specific peroxisome proliferator activated receptor gamma modulator with potent antidiabetes and antiatherogenic effects. Diabetes 2007, 56, 2797–2808. [Google Scholar] [CrossRef] [PubMed]
  33. Seargent, J.M.; Yates, E.A.; Gill, J.H. GW9662, a potent antagonist of PPARgamma, inhibits growth of breast tumour cells and promotes the anticancer effects of the PPARgamma agonist rosiglitazone, independently of PPARgamma activation. Br. J. Pharmacol. 2004, 143, 933–937. [Google Scholar] [CrossRef] [PubMed]
  34. Altmann, R.; Hausmann, M.; Spottl, T.; Gruber, M.; Bull, A.W.; Menzel, K.; Vogl, D.; Herfarth, H.; Scholmerich, J.; Falk, W.; et al. 13-Oxo-ODE is an endogenous ligand for PPARgamma in human colonic epithelial cells. Biochem. Pharmacol. 2007, 74, 612–622. [Google Scholar] [CrossRef] [PubMed]
  35. Ljung, B.; Bamberg, K.; Dahllof, B.; Kjellstedt, A.; Oakes, N.D.; Ostling, J.; Svensson, L.; Camejo, G. AZ 242, a novel PPARalpha/gamma agonist with beneficial effects on insulin resistance and carbohydrate and lipid metabolism in ob/ob mice and obese Zucker rats. J. Lipid Res. 2002, 43, 1855–1863. [Google Scholar] [CrossRef]
  36. Leghmar, K.; Cenac, N.; Rolland, M.; Martin, H.; Rauwel, B.; Bertrand-Michel, J.; Le Faouder, P.; Benard, M.; Casper, C.; Davrinche, C.; et al. Cytomegalovirus Infection Triggers the Secretion of the PPARgamma Agonists 15-Hydroxyeicosatetraenoic Acid (15-HETE) and 13-Hydroxyoctadecadienoic Acid (13-HODE) in Human Cytotrophoblasts and Placental Cultures. PLoS ONE 2015, 10, e0132627. [Google Scholar] [CrossRef]
  37. Cesario, R.M.; Klausing, K.; Razzaghi, H.; Crombie, D.; Rungta, D.; Heyman, R.A.; Lala, D.S. The rexinoid LG100754 is a novel RXR:PPARgamma agonist and decreases glucose levels in vivo. Mol. Endocrinol. 2001, 15, 1360–1369. [Google Scholar] [CrossRef]
  38. Kurtz, T.W. Treating the metabolic syndrome: Telmisartan as a peroxisome proliferator-activated receptor-gamma activator. Acta Diabetol. 2005, 42 (Suppl. 1), S9–S16. [Google Scholar] [CrossRef]
  39. Zhang, Z.Z.; Shang, Q.H.; Jin, H.Y.; Song, B.; Oudit, G.Y.; Lu, L.; Zhou, T.; Xu, Y.L.; Gao, P.J.; Zhu, D.L.; et al. Cardiac protective effects of irbesartan via the PPAR-gamma signaling pathway in angiotensin-converting enzyme 2-deficient mice. J. Transl. Med. 2013, 11, 229. [Google Scholar] [CrossRef]
  40. Tenenbaum, A.; Motro, M.; Fisman, E.Z.; Schwammenthal, E.; Adler, Y.; Goldenberg, I.; Leor, J.; Boyko, V.; Mandelzweig, L.; Behar, S. Peroxisome proliferator-activated receptor ligand bezafibrate for prevention of type 2 diabetes mellitus in patients with coronary artery disease. Circulation 2004, 109, 2197–2202. [Google Scholar] [CrossRef]
  41. Schug, T.T.; Berry, D.C.; Shaw, N.S.; Travis, S.N.; Noy, N. Opposing effects of retinoic acid on cell growth result from alternate activation of two different nuclear receptors. Cell 2007, 129, 723–733. [Google Scholar] [CrossRef]
  42. Kumagai, T.; Ikezoe, T.; Gui, D.; O’Kelly, J.; Tong, X.J.; Cohen, F.J.; Said, J.W.; Koeffler, H.P. RWJ-241947 (MCC-555), a unique peroxisome proliferator-activated receptor-gamma ligand with antitumor activity against human prostate cancer in vitro and in beige/nude/ X-linked immunodeficient mice and enhancement of apoptosis in myeloma cells induced by arsenic trioxide. Clin. Cancer Res. 2004, 10, 1508–1520. [Google Scholar] [CrossRef] [PubMed]
  43. Fukui, Y.; Masui, S.; Osada, S.; Umesono, K.; Motojima, K. A new thiazolidinedione, NC-2100, which is a weak PPAR-gamma activator, exhibits potent antidiabetic effects and induces uncoupling protein 1 in white adipose tissue of KKAy obese mice. Diabetes 2000, 49, 759–767. [Google Scholar] [CrossRef] [PubMed]
  44. Murakami, K.; Tsunoda, M.; Ide, T.; Ohashi, M.; Mochizuki, T. Amelioration by KRP-297, a new thiazolidinedione, of impaired glucose uptake in skeletal muscle from obese insulin-resistant animals. Metabolism 1999, 48, 1450–1454. [Google Scholar] [CrossRef]
  45. Christofides, A.; Konstantinidou, E.; Jani, C.; Boussiotis, V.A. The role of peroxisome proliferator-activated receptors (PPAR) in immune responses. Metabolism 2021, 114, 154338. [Google Scholar] [CrossRef] [PubMed]
  46. Giaginis, C.; Tsantili-Kakoulidou, A.; Theocharis, S. Peroxisome Proliferator-Activated Receptor-gamma Ligands: Potential Pharmacological Agents for Targeting the Angiogenesis Signaling Cascade in Cancer. PPAR Res. 2008, 2008, 431763. [Google Scholar] [CrossRef] [PubMed]
  47. Peng, L.; Ye, Y.; Mullikin, H.; Lin, L.; Kuhn, C.; Rahmeh, M.; Mahner, S.; Jeschke, U.; von Schonfeldt, V. Expression of trophoblast derived prostaglandin E2 receptor 2 (EP2) is reduced in patients with recurrent miscarriage and EP2 regulates cell proliferation and expression of inflammatory cytokines. J. Reprod. Immunol. 2020, 142, 103210. [Google Scholar] [CrossRef] [PubMed]
  48. Barak, Y.; Nelson, M.C.; Ong, E.S.; Jones, Y.Z.; Ruiz-Lozano, P.; Chien, K.R.; Koder, A.; Evans, R.M. PPAR gamma is required for placental, cardiac, and adipose tissue development. Mol. Cell 1999, 4, 585–595. [Google Scholar] [CrossRef] [PubMed]
  49. Wang, Q.; Fujii, H.; Knipp, G.T. Expression of PPAR and RXR isoforms in the developing rat and human term placentas. Placenta 2002, 23, 661–671. [Google Scholar] [CrossRef]
  50. Peng, L.; Yang, H.; Ye, Y.; Ma, Z.; Kuhn, C.; Rahmeh, M.; Mahner, S.; Makrigiannakis, A.; Jeschke, U.; von Schonfeldt, V. Role of Peroxisome Proliferator-Activated Receptors (PPARs) in Trophoblast Functions. Int. J. Mol. Sci. 2021, 22, 433. [Google Scholar] [CrossRef]
  51. Diaz, M.; Bassols, J.; Lopez-Bermejo, A.; Gomez-Roig, M.D.; de Zegher, F.; Ibanez, L. Placental expression of peroxisome proliferator-activated receptor gamma (PPARgamma): Relation to placental and fetal growth. J. Clin. Endocrinol. Metab. 2012, 97, E1468–E1472. [Google Scholar] [CrossRef]
  52. Christians, J.K.; Beristain, A.G. ADAM12 and PAPP-A: Candidate regulators of trophoblast invasion and first trimester markers of healthy trophoblasts. Cell. Adh Migr. 2016, 10, 147–153. [Google Scholar] [CrossRef] [PubMed]
  53. Fournier, T.; Handschuh, K.; Tsatsaris, V.; Evain-Brion, D. Involvement of PPARgamma in human trophoblast invasion. Placenta 2007, 28 (Suppl. A), S76–S81. [Google Scholar] [CrossRef] [PubMed]
  54. Tarrade, A.; Schoonjans, K.; Pavan, L.; Auwerx, J.; Rochette-Egly, C.; Evain-Brion, D.; Fournier, T. PPARgamma/RXRalpha heterodimers control human trophoblast invasion. J. Clin. Endocrinol. Metab. 2001, 86, 5017–5024. [Google Scholar] [CrossRef] [PubMed]
  55. Handschuh, K.; Guibourdenche, J.; Guesnon, M.; Laurendeau, I.; Evain-Brion, D.; Fournier, T. Modulation of PAPP-A expression by PPARgamma in human first trimester trophoblast. Placenta 2006, 27 (Suppl. A), S127–S134. [Google Scholar] [CrossRef]
  56. Mayama, R.; Izawa, T.; Sakai, K.; Suciu, N.; Iwashita, M. Improvement of insulin sensitivity promotes extravillous trophoblast cell migration stimulated by insulin-like growth factor-I. Endocr. J. 2013, 60, 359–368. [Google Scholar] [CrossRef]
  57. Bilban, M.; Haslinger, P.; Prast, J.; Klinglmuller, F.; Woelfel, T.; Haider, S.; Sachs, A.; Otterbein, L.E.; Desoye, G.; Hiden, U.; et al. Identification of novel trophoblast invasion-related genes: Heme oxygenase-1 controls motility via peroxisome proliferator-activated receptor gamma. Endocrinology 2009, 150, 1000–1013. [Google Scholar] [CrossRef]
  58. Garnier, V.; Traboulsi, W.; Salomon, A.; Brouillet, S.; Fournier, T.; Winkler, C.; Desvergne, B.; Hoffmann, P.; Zhou, Q.Y.; Congiu, C.; et al. PPARgamma controls pregnancy outcome through activation of EG-VEGF: New insights into the mechanism of placental development. Am. J. Physiol. Endocrinol. Metab. 2015, 309, E357–E369. [Google Scholar] [CrossRef]
  59. Kadam, L.; Kilburn, B.; Baczyk, D.; Kohan-Ghadr, H.R.; Kingdom, J.; Drewlo, S. Rosiglitazone blocks first trimester in-vitro placental injury caused by NF-kappaB-mediated inflammation. Sci. Rep. 2019, 9, 2018. [Google Scholar] [CrossRef]
  60. Zhang, Z.; Yang, Y.; Lv, X.; Liu, H. Interleukin-17 promotes proliferation, migration, and invasion of trophoblasts via regulating PPAR-gamma/RXR-alpha/Wnt signaling. Bioengineered 2022, 13, 1224–1234. [Google Scholar] [CrossRef]
  61. Shoaito, H.; Chauveau, S.; Gosseaume, C.; Bourguet, W.; Vigouroux, C.; Vatier, C.; Pienkowski, C.; Fournier, T.; Degrelle, S.A. Peroxisome proliferator-activated receptor gamma-ligand-binding domain mutations associated with familial partial lipodystrophy type 3 disrupt human trophoblast fusion and fibroblast migration. J. Cell. Mol. Med. 2020, 24, 7660–7669. [Google Scholar] [CrossRef]
  62. Levytska, K.; Drewlo, S.; Baczyk, D.; Kingdom, J. PPAR-gamma Regulates Trophoblast Differentiation in the BeWo Cell Model. PPAR Res. 2014, 2014, 637251. [Google Scholar] [CrossRef] [PubMed]
  63. Tarrade, A.; Schoonjans, K.; Guibourdenche, J.; Bidart, J.M.; Vidaud, M.; Auwerx, J.; Rochette-Egly, C.; Evain-Brion, D. PPAR gamma/RXR alpha heterodimers are involved in human CG beta synthesis and human trophoblast differentiation. Endocrinology 2001, 142, 4504–4514. [Google Scholar] [CrossRef] [PubMed]
  64. Milstone, D.S.; Pierre, M.A.; Mana, P.M.; O’Donnell, P.E.; Davis, V.M.; Cross, J.C.; Mortensen, R.M.; Stavrakis, G. PPAR gamma is expressed and regulates placental development and trophoblast differentiation in both humans and mice. FASEB 2006, 20, A1077. [Google Scholar] [CrossRef]
  65. Elchalal, U.; Humphrey, R.G.; Smith, S.D.; Hu, C.; Sadovsky, Y.; Nelson, D.M. Troglitazone attenuates hypoxia-induced injury in cultured term human trophoblasts. Am. J. Obstet. Gynecol. 2004, 191, 2154–2159. [Google Scholar] [CrossRef]
  66. Tache, V.; Ciric, A.; Moretto-Zita, M.; Li, Y.; Peng, J.; Maltepe, E.; Milstone, D.S.; Parast, M.M. Hypoxia and trophoblast differentiation: A key role for PPARgamma. Stem Cells Dev. 2013, 22, 2815–2824. [Google Scholar] [CrossRef] [PubMed]
  67. Parast, M.M.; Yu, H.; Ciric, A.; Salata, M.W.; Davis, V.; Milstone, D.S. PPARgamma regulates trophoblast proliferation and promotes labyrinthine trilineage differentiation. PLoS ONE 2009, 4, e8055. [Google Scholar] [CrossRef]
  68. Fox, R.; Kitt, J.; Leeson, P.; Aye, C.Y.L.; Lewandowski, A.J. Preeclampsia: Risk Factors, Diagnosis, Management, and the Cardiovascular Impact on the Offspring. J. Clin. Med. 2019, 8, 1625. [Google Scholar] [CrossRef]
  69. Rana, S.; Lemoine, E.; Granger, J.P.; Karumanchi, S.A. Preeclampsia: Pathophysiology, Challenges, and Perspectives. Circ. Res. 2019, 124, 1094–1112. [Google Scholar] [CrossRef]
  70. Backes, C.H.; Markham, K.; Moorehead, P.; Cordero, L.; Nankervis, C.A.; Giannone, P.J. Maternal preeclampsia and neonatal outcomes. J. Pregnancy 2011, 2011, 214365. [Google Scholar] [CrossRef]
  71. Huppertz, B. Placental origins of preeclampsia: Challenging the current hypothesis. Hypertension 2008, 51, 970–975. [Google Scholar] [CrossRef]
  72. Staff, A.C. The two-stage placental model of preeclampsia: An update. J. Reprod. Immunol. 2019, 134–135, 1–10. [Google Scholar] [CrossRef] [PubMed]
  73. Roberts, J.M.; Hubel, C.A. The two stage model of preeclampsia: Variations on the theme. Placenta 2009, 30 (Suppl. A), S32–S37. [Google Scholar] [CrossRef] [PubMed]
  74. Roberts, J.M. Pathophysiology of ischemic placental disease. Semin. Perinatol. 2014, 38, 139–145. [Google Scholar] [CrossRef] [PubMed]
  75. Jia, J.; Wu, J.; Hu, J. Correlations of MMP-9 and PPARγ gene polymorphisms with occurrence of preeclampsia. Eur. Rev. Med. Pharmacol. Sci. 2022, 26, 771–778. [Google Scholar]
  76. Holdsworth-Carson, S.; Lim, R.; Mitton, A.; Whitehead, C.; Rice, G.E.; Permezel, M.; Lappas, M. Peroxisome proliferator-activated receptors are altered in pathologies of the human placenta: Gestational diabetes mellitus, intrauterine growth restriction and preeclampsia. Placenta 2010, 31, 222–229. [Google Scholar] [CrossRef]
  77. Mahendra, J.; Parthiban, P.S.; Mahendra, L.; Balakrishnan, A.; Shanmugam, S.; Junaid, M.; Romanos, G.E. Evidence linking the role of placental expressions of Peroxisome Proliferator-Activated Receptor-γ and Nuclear Factor-Kappa B in the pathogenesis of preeclampsia associated with periodontitis. J. Periodontol. 2016, 87, 962–970. [Google Scholar] [CrossRef]
  78. Armistead, B.; Kadam, L.; Siegwald, E.; McCarthy, F.P.; Kingdom, J.C.; Kohan-Ghadr, H.R.; Drewlo, S. Induction of the PPARγ (Peroxisome Proliferator-Activated Receptor γ)-GCM1 (Glial Cell Missing 1) Syncytialization Axis Reduces sFLT1 (Soluble fms-Like Tyrosine Kinase 1) in the Preeclamptic Placenta. Hypertension 2021, 78, 230–240. [Google Scholar] [CrossRef]
  79. He, P.; Chen, Z.; Sun, Q.; Li, Y.; Gu, H.; Ni, X. Reduced expression of 11β-hydroxysteroid dehydrogenase type 2 in preeclamptic placentas is associated with decreased PPARγ but increased PPARα expression. Endocrinology 2014, 155, 299–309. [Google Scholar] [CrossRef]
  80. Permadi, W.; Mantilidewi, K.I.; Khairani, A.F.; Lantika, U.A.; Ronosulistyo, A.R.; Bayuaji, H. Differences in expression of Peroxisome Proliferator-activated Receptor-gamma in early-onset preeclampsia and late-onset preeclampsia. BMC Res. Notes 2020, 13, 181. [Google Scholar] [CrossRef]
  81. Zeisler, H.; Llurba, E.; Chantraine, F.; Vatish, M.; Staff, A.C.; Sennstrom, M.; Olovsson, M.; Brennecke, S.P.; Stepan, H.; Allegranza, D.; et al. Predictive Value of the sFlt-1:PlGF Ratio in Women with Suspected Preeclampsia. N. Engl. J. Med. 2016, 374, 13–22. [Google Scholar] [CrossRef]
  82. Liu, L.; Wang, R.; Xu, R.; Chu, Y.; Gu, W. Procyanidin B2 ameliorates endothelial dysfunction and impaired angiogenesis via the Nrf2/PPARgamma/sFlt-1 axis in preeclampsia. Pharmacol. Res. 2022, 177, 106127. [Google Scholar] [CrossRef]
  83. Liu, L.; Zhuang, X.; Jiang, M.; Guan, F.; Fu, Q.; Lin, J. ANGPTL4 mediates the protective role of PPARγ activators in the pathogenesis of preeclampsia. Cell. Death Dis. 2017, 8, e3054. [Google Scholar] [CrossRef] [PubMed]
  84. Kamrani, A.; Alipourfard, I.; Ahmadi-Khiavi, H.; Yousefi, M.; Rostamzadeh, D.; Izadi, M.; Ahmadi, M. The role of epigenetic changes in preeclampsia. Biofactors 2019, 45, 712–724. [Google Scholar] [CrossRef] [PubMed]
  85. Meister, S.; Hahn, L.; Beyer, S.; Paul, C.; Mitter, S.; Kuhn, C.; von Schönfeldt, V.; Corradini, S.; Sudan, K.; Schulz, C.; et al. Regulation of Epigenetic Modifications in the Placenta during Preeclampsia: PPARγ Influences H3K4me3 and H3K9ac in Extravillous Trophoblast Cells. Int. J. Mol. Sci. 2021, 22, 12469. [Google Scholar] [CrossRef]
  86. Bianco-Miotto, T.; Mayne, B.T.; Buckberry, S.; Breen, J.; Rodriguez Lopez, C.M.; Roberts, C.T. Recent progress towards understanding the role of DNA methylation in human placental development. Reproduction 2016, 152, R23–R30. [Google Scholar] [CrossRef] [PubMed]
  87. Nelissen, E.C.; van Montfoort, A.P.; Dumoulin, J.C.; Evers, J.L. Epigenetics and the placenta. Hum. Reprod. Update 2011, 17, 397–417. [Google Scholar] [CrossRef]
  88. Li, J.; LaMarca, B.; Reckelhoff, J.F. A model of preeclampsia in rats: The reduced uterine perfusion pressure (RUPP) model. Am. J. Physiol. Heart Circ. Physiol. 2012, 303, H1–H8. [Google Scholar] [CrossRef]
  89. McCarthy, F.P.; Drewlo, S.; Kingdom, J.; Johns, E.J.; Walsh, S.K.; Kenny, L.C. Peroxisome proliferator-activated receptor-gamma as a potential therapeutic target in the treatment of preeclampsia. Hypertension 2011, 58, 280–286. [Google Scholar] [CrossRef]
  90. Zhang, C.; Zhu, Y.; Shen, Y.; Zuo, C. Aspirin ameliorates preeclampsia induced by a peroxisome proliferator-activated receptor antagonist. Reprod. Sci. 2018, 25, 1655–1662. [Google Scholar] [CrossRef]
  91. McCarthy, F.P.; Drewlo, S.; English, F.A.; Kingdom, J.; Johns, E.J.; Kenny, L.C.; Walsh, S.K. Evidence implicating peroxisome proliferator-activated receptor-γ in the pathogenesis of preeclampsia. Hypertension 2011, 58, 882–887. [Google Scholar] [CrossRef]
  92. Guo, Y.; Zhu, Y.; Sun, Y.; Yang, H. The preventive effect of low-dose aspirin in a PPAR-γ antagonist treated mouse model of preeclampsia. BMC Pregnancy Childbirth 2022, 22, 606. [Google Scholar] [CrossRef] [PubMed]
  93. National Center for Biotechnology Information. Angiotensin (1-7). Available online: https://pubchem.ncbi.nlm.nih.gov/compound/Angiotensin-_1-7 (accessed on 11 May 2023).
  94. El-Saka, M.H.; Madi, N.M.; Ibrahim, R.R.; Alghazaly, G.M.; Elshwaikh, S.; El-Bermawy, M. The ameliorative effect of angiotensin 1-7 on experimentally induced-preeclampsia in rats: Targeting the role of peroxisome proliferator-activated receptors gamma expression & asymmetric dimethylarginine. Arch. Biochem. Biophys. 2019, 671, 123–129. [Google Scholar] [CrossRef] [PubMed]
  95. Buchanan, T.A.; Xiang, A.H.; Page, K.A. Gestational diabetes mellitus: Risks and management during and after pregnancy. Nat. Rev. Endocrinol. 2012, 8, 639–649. [Google Scholar] [CrossRef] [PubMed]
  96. Damm, P. Future risk of diabetes in mother and child after gestational diabetes mellitus. Int. J. Gynaecol. Obstet. 2009, 104 (Suppl. 1), S25–S26. [Google Scholar] [CrossRef] [PubMed]
  97. Weissgerber, T.L.; Mudd, L.M. Preeclampsia and diabetes. Curr. Diab Rep. 2015, 15, 9. [Google Scholar] [CrossRef]
  98. Yang, Y.; Wu, N. Gestational Diabetes Mellitus and Preeclampsia: Correlation and Influencing Factors. Front. Cardiovasc. Med. 2022, 9, 831297. [Google Scholar] [CrossRef]
  99. Karagiannis, T.; Bekiari, E.; Manolopoulos, K.; Paletas, K.; Tsapas, A. Gestational diabetes mellitus: Why screen and how to diagnose. Hippokratia 2010, 14, 151–154. [Google Scholar]
  100. Qin, Y.; McCauley, N.; Ding, Z.; Lawless, L.; Liu, Z.; Zhang, K.; Xie, L. Hyperglycemia results in significant pathophysiological changes of placental spiral artery remodeling and angiogenesis, further contributing to congenital defects. Front. Biosci. 2021, 26, 965–976. [Google Scholar] [CrossRef]
  101. Hay, W.W., Jr. Placental-fetal glucose exchange and fetal glucose metabolism. Trans. Am. Clin. Climatol. Assoc. 2006, 117, 321–339; discussion 339–340. [Google Scholar]
  102. Balsells, M.; Garcia-Patterson, A.; Sola, I.; Roque, M.; Gich, I.; Corcoy, R. Glibenclamide, metformin, and insulin for the treatment of gestational diabetes: A systematic review and meta-analysis. BMJ 2015, 350, h102. [Google Scholar] [CrossRef]
  103. Jamilian, M.; Samimi, M.; Mirhosseini, N.; Afshar Ebrahimi, F.; Aghadavod, E.; Taghizadeh, M.; Asemi, Z. A Randomized Double-Blinded, Placebo-Controlled Trial Investigating the Effect of Fish Oil Supplementation on Gene Expression Related to Insulin Action, Blood Lipids, and Inflammation in Gestational Diabetes Mellitus-Fish Oil Supplementation and Gestational Diabetes. Nutrients 2018, 10, 163. [Google Scholar] [CrossRef] [PubMed]
  104. Holmes, H.J.; Casey, B.M.; Bawdon, R.E. Placental transfer of rosiglitazone in the ex vivo human perfusion model. Am. J. Obstet. Gynecol. 2006, 195, 1715–1719. [Google Scholar] [CrossRef] [PubMed]
  105. Chan, L.Y.; Yeung, J.H.; Lau, T.K. Placental transfer of rosiglitazone in the first trimester of human pregnancy. Fertil. Steril. 2005, 83, 955–958. [Google Scholar] [CrossRef] [PubMed]
  106. Nanovskaya, T.N.; Patrikeeva, S.; Hemauer, S.; Fokina, V.; Mattison, D.; Hankins, G.D.; Ahmed, M.S.; Network, O. Effect of albumin on transplacental transfer and distribution of rosiglitazone and glyburide. J. Matern.-Fetal Neonatal Med. 2008, 21, 197–207. [Google Scholar] [CrossRef] [PubMed]
  107. Klinkner, D.B.; Lim, H.J.; Strawn, E.Y., Jr.; Oldham, K.T.; Sander, T.L. An in vivo murine model of rosiglitazone use in pregnancy. Fertil. Steril. 2006, 86, 1074–1079. [Google Scholar] [CrossRef] [PubMed]
  108. Kalyoncu, N.I.; Yaris, F.; Ulku, C.; Kadioglu, M.; Kesim, M.; Unsal, M.; Dikici, M.; Yaris, E. A case of rosiglitazone exposure in the second trimester of pregnancy. Reprod. Toxicol. 2005, 19, 563–564. [Google Scholar] [CrossRef]
  109. Sağır, D.; Eren, B.; Yılmaz, B.; Eren, Z.; Keleş, O.; Gökçe, A. Effects of prenatal PPAR-γ agonist rosiglitazone exposure on rat hippocampus development in a time-dependent manner: A stereological and histopathological study. Human. Exp. Toxicol. 2018, 37, 827–835. [Google Scholar] [CrossRef]
  110. Chan, L.Y.; Lau, T.K. Effect of rosiglitazone on embryonic growth and morphology: A study using a whole rat embryo culture model. Fertil. Steril. 2006, 86, 490–492. [Google Scholar] [CrossRef]
  111. Schaiff, W.T.; Knapp, F.F., Jr.; Barak, Y.; Biron-Shental, T.; Nelson, D.M.; Sadovsky, Y. Ligand-activated peroxisome proliferator activated receptor gamma alters placental morphology and placental fatty acid uptake in mice. Endocrinology 2007, 148, 3625–3634. [Google Scholar] [CrossRef]
  112. Nadra, K.; Quignodon, L.; Sardella, C.; Joye, E.; Mucciolo, A.; Chrast, R.; Desvergne, B. PPARgamma in placental angiogenesis. Endocrinology 2010, 151, 4969–4981. [Google Scholar] [CrossRef]
  113. Petrik, J.J.; Gerstein, H.C.; Cesta, C.E.; Kellenberger, L.D.; Alfaidy, N.; Holloway, A.C. Effects of rosiglitazone on ovarian function and fertility in animals with reduced fertility following fetal and neonatal exposure to nicotine. Endocrine 2009, 36, 281–290. [Google Scholar] [CrossRef] [PubMed]
  114. Gao, Y.; She, R.; Sha, W. Gestational diabetes mellitus is associated with decreased adipose and placenta peroxisome proliferator-activator receptor γ expression in a Chinese population. Oncotarget 2017, 8, 113928. [Google Scholar] [CrossRef] [PubMed]
  115. Zhao, Q.; Yang, D.; Gao, L.; Zhao, M.; He, X.; Zhu, M.; Tian, C.; Liu, G.; Li, L.; Hu, C. Downregulation of peroxisome proliferator-activated receptor gamma in the placenta correlates to hyperglycemia in offspring at young adulthood after exposure to gestational diabetes mellitus. J. Diabetes Investig. 2019, 10, 499–512. [Google Scholar] [CrossRef] [PubMed]
  116. Knabl, J.; Huttenbrenner, R.; Hutter, S.; Gunthner-Biller, M.; Vrekoussis, T.; Karl, K.; Friese, K.; Kainer, F.; Jeschke, U. Peroxisome proliferator-activated receptor-gamma (PPARgamma) is down regulated in trophoblast cells of gestational diabetes mellitus (GDM) and in trophoblast tumour cells BeWo in vitro after stimulation with PPARgamma agonists. J. Perinat. Med. 2014, 42, 179–187. [Google Scholar] [CrossRef] [PubMed]
  117. Fisher, J.J.; Vanderpeet, C.L.; Bartho, L.A.; McKeating, D.R.; Cuffe, J.S.M.; Holland, O.J.; Perkins, A.V. Mitochondrial dysfunction in placental trophoblast cells experiencing gestational diabetes mellitus. J. Physiol. 2021, 599, 1291–1305. [Google Scholar] [CrossRef] [PubMed]
  118. Suwaki, N.; Masuyama, H.; Masumoto, A.; Takamoto, N.; Hiramatsu, Y. Expression and potential role of peroxisome proliferator-activated receptor gamma in the placenta of diabetic pregnancy. Placenta 2007, 28, 315–323. [Google Scholar] [CrossRef]
  119. Wójcik, M.; Mac-Marcjanek, K.; Nadel, I.; Woźniak, L.; Cypryk, K. Gestational diabetes mellitus is associated with increased leukocyte peroxisome proliferator-activated receptor γ expression. Arch. Med. Sci. 2015, 11, 779–787. [Google Scholar] [CrossRef]
  120. Han, C.S.; Herrin, M.A.; Pitruzzello, M.C.; Mulla, M.J.; Werner, E.F.; Pettker, C.M.; Flannery, C.A.; Abrahams, V.M. Glucose and metformin modulate human first trimester trophoblast function: A model and potential therapy for diabetes-associated uteroplacental insufficiency. Am. J. Reprod. Immunol. 2015, 73, 362–371. [Google Scholar] [CrossRef]
  121. Jawerbaum, A.; Capobianco, E.; Pustovrh, C.; White, V.; Baier, M.; Salzberg, S.; Pesaresi, M.; Gonzalez, E. Influence of peroxisome proliferator-activated receptor gamma activation by its endogenous ligand 15-deoxy Delta12,14 prostaglandin J2 on nitric oxide production in term placental tissues from diabetic women. Mol. Hum. Reprod. 2004, 10, 671–676. [Google Scholar] [CrossRef]
  122. Zhang, J.; Bai, W.P. C1q/tumor necrosis factor related protein 6 (CTRP6) regulates the phenotypes of high glucose-induced gestational trophoblast cells via peroxisome proliferator-activated receptor gamma (PPARgamma) signaling. Bioengineered 2022, 13, 206–216. [Google Scholar] [CrossRef]
  123. Zhou, X.; Wei, L.-J.; Li, J.-Q.; Zhang, J.-Y.; Zhu, S.-L.; Zhang, H.-T.; Jia, J.; Yu, J.; Wang, S.-S.; Feng, L.; et al. The Activation of Peroxisome Proliferator-activated Receptor γ Enhances Insulin Signaling Pathways via Up-regulating Chemerin Expression in High Glucose Treated HTR-8/SVneo Cells. Matern.-Fetal Med. 2020, 2, 131–140. [Google Scholar] [CrossRef]
  124. Jin, D.; Sun, J.; Huang, J.; Yu, X.; Yu, A.; He, Y.; Li, Q.; Yang, Z. Peroxisome proliferator-activated receptor gamma enhances adiponectin secretion via up-regulating DsbA-L expression. Mol. Cell. Endocrinol. 2015, 411, 97–104. [Google Scholar] [CrossRef] [PubMed]
  125. Zhou, X.; Li, J.Q.; Wei, L.J.; He, M.Z.; Jia, J.; Zhang, J.Y.; Wang, S.S.; Feng, L. Silencing of DsbA-L gene impairs the PPARgamma agonist function of improving insulin resistance in a high-glucose cell model. J. Zhejiang Univ. Sci. B 2020, 21, 990–998. [Google Scholar] [CrossRef] [PubMed]
Figure 1. PRISMA Flowchart of article selection process.
Figure 1. PRISMA Flowchart of article selection process.
Nutrients 15 02459 g001
Figure 2. Illustrative representation of ligands and genes that interact with PPARγ in regulating placenta function related to preeclampsia. PPARγ activated by agonists or upstream regulation by Angiotensin 1-7 or Procyanidin B2 ameliorate preeclamptic phenotypes. Decreased PPARγ activity triggered by antagonists exacerbates the preeclamptic condition. AT1-7: Angiotensin 1-7; PCB2: Procyanidin B2; Rosi: Rosiglitazone: Ci: Ciglitazone.
Figure 2. Illustrative representation of ligands and genes that interact with PPARγ in regulating placenta function related to preeclampsia. PPARγ activated by agonists or upstream regulation by Angiotensin 1-7 or Procyanidin B2 ameliorate preeclamptic phenotypes. Decreased PPARγ activity triggered by antagonists exacerbates the preeclamptic condition. AT1-7: Angiotensin 1-7; PCB2: Procyanidin B2; Rosi: Rosiglitazone: Ci: Ciglitazone.
Nutrients 15 02459 g002
Table 1. Natural and synthetic ligands of PPARγ *.
Table 1. Natural and synthetic ligands of PPARγ *.
Agonists Antagonists
Natural LigandSynthetic LigandNatural LigandSynthetic Ligand
Unsaturated fatty acid ** [15]GW1929 [16]Betulinic acid [17]SR-202 [18]
Oxidized LDL [19]TZD *** NFκB [20]BADGE [21]
EETs [22]FMOC-L-Leucine [23]Fetuin A [20]LG100641 [24]
15d-PGJ2 [25]INT131 [26] PD068235 [27]
Azelaoyl phosphatidylcholine [28]Farglitazar (GI262570) [29] T0070907 [30]
9-oxoODE [31]S26948 [32] GW9662 [33]
13-oxoODE [34]AZ 242 [35]
15-HETE [36]LG100754 [37]
13-HODE [36]
* Partial ligands of PPARγ such as telmisartan [38], Irbesartan [39], metaglidasen [40], and non-TZD partial agonist (nTZDpa) [41] are not included. ** poly-unsaturated FAs γ-linolenic (18:3), eicosatrienoic acid (C20:3), dihomo-γ-linolenic (20:3), arachidonic acid (C20:4), and eicosapentaenoic acid (C20:5). *** rosiglitazone, pioglitazone, troglitazone, ciglitazone [21], RWJ-241947 [42], NC-2100 [43], and KRP-297 [44].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Qin, Y.; Bily, D.; Aguirre, M.; Zhang, K.; Xie, L. Understanding PPARγ and Its Agonists on Trophoblast Differentiation and Invasion: Potential Therapeutic Targets for Gestational Diabetes Mellitus and Preeclampsia. Nutrients 2023, 15, 2459. https://doi.org/10.3390/nu15112459

AMA Style

Qin Y, Bily D, Aguirre M, Zhang K, Xie L. Understanding PPARγ and Its Agonists on Trophoblast Differentiation and Invasion: Potential Therapeutic Targets for Gestational Diabetes Mellitus and Preeclampsia. Nutrients. 2023; 15(11):2459. https://doi.org/10.3390/nu15112459

Chicago/Turabian Style

Qin, Yushu, Donalyn Bily, Makayla Aguirre, Ke Zhang, and Linglin Xie. 2023. "Understanding PPARγ and Its Agonists on Trophoblast Differentiation and Invasion: Potential Therapeutic Targets for Gestational Diabetes Mellitus and Preeclampsia" Nutrients 15, no. 11: 2459. https://doi.org/10.3390/nu15112459

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop