Next Article in Journal
Beneficial Role of Exercise in the Modulation of mdx Muscle Plastic Remodeling and Oxidative Stress
Next Article in Special Issue
Diet-Derived Antioxidants and Their Role in Inflammation, Obesity and Gut Microbiota Modulation
Previous Article in Journal
Determination of Suitable Macroporous Resins and Desorbents for Carnosol and Carnosic Acid from Deep Eutectic Solvent Sage (Salvia officinalis) Extract with Assessment of Antiradical and Antibacterial Activity
Previous Article in Special Issue
Mediterranean Diet and Physical Activity Decrease the Initiation of Cardiovascular Drug Use in High Cardiovascular Risk Individuals: A Cohort Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Impact of a Plant-Based Diet on Gestational Diabetes: A Review

by
Antonio Schiattarella
1,
Mauro Lombardo
2,
Maddalena Morlando
1 and
Gianluca Rizzo
3,*
1
Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
2
Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
3
Independent Researcher, Via Venezuela 66, 98121 Messina, Italy
*
Author to whom correspondence should be addressed.
Antioxidants 2021, 10(4), 557; https://doi.org/10.3390/antiox10040557
Submission received: 8 March 2021 / Revised: 30 March 2021 / Accepted: 31 March 2021 / Published: 2 April 2021
(This article belongs to the Special Issue Plant-Based Diets and Their Antioxidant Role in Human Health)

Abstract

:
Gestational diabetes mellitus (GDM) represents a challenging pregnancy complication in which women present a state of glucose intolerance. GDM has been associated with various obstetric complications, such as polyhydramnios, preterm delivery, and increased cesarean delivery rate. Moreover, the fetus could suffer from congenital malformation, macrosomia, neonatal respiratory distress syndrome, and intrauterine death. It has been speculated that inflammatory markers such as tumor necrosis factor-alpha (TNF-α), interleukin (IL) 6, and C-reactive protein (CRP) impact on endothelium dysfunction and insulin resistance and contribute to the pathogenesis of GDM. Nutritional patterns enriched with plant-derived foods, such as a low glycemic or Mediterranean diet, might favorably impact on the incidence of GDM. A high intake of vegetables, fibers, and fruits seems to decrease inflammation by enhancing antioxidant compounds. This aspect contributes to improving insulin efficacy and metabolic control and could provide maternal and neonatal health benefits. Our review aims to deepen the understanding of the impact of a plant-based diet on oxidative stress in GDM.

1. Introduction

Gestational diabetes mellitus (GDM) represents a challenging pregnancy complication in which women present a state of glucose intolerance that is diagnosed for the first time during pregnancy. It has been estimated that 5 to 7% of pregnancies are complicated by diabetes, and almost 80% is GDM [1,2,3]. Diagnosis of GDM is achieved by the 75 g oral glucose tolerance test, although thresholds vary among different health and diabetes associations [4].
GDM has been associated with various obstetric complications, such as polyhydramnios, preterm delivery, shoulder dystocia, and increased rates of cesarean delivery [5]. Moreover, the fetus could suffer from congenital malformations, macrosomia, neonatal respiratory distress syndrome, hypoglycemia, and intrauterine death [5]. GDM presents a challenging diagnosis, and its management can be difficult. Ethnicity is a risk factor for the development of GDM, as the incidence of GDM is increased among Hispanic and African women [6]. Obesity, a family history of type 2 diabetes (T2D), and a prior history of GDM could also increase the risk of GDM.
There is increasing evidence that GDM presents a genetic component like T2D and aggregates within families [7]. Women with a diabetic sibling had an 8.4-fold increased risk of GDM [8]. In addition, specific gene variants of melatonin receptor 1B (MTNR1B), transcription factor 7-like 2 (TCF7L2), and insulin receptor substrate 1 (IRS-1) have been found to be associated with GDM [9,10]. It has been speculated that a high sugar intake and increased weight gain during pregnancy might be responsible for an inflammatory pathway that impacts on the onset of insulin resistance [11,12]. Findings from other studies correlated heme iron levels with GDM but not non-heme iron derived from plant-based foods such as grains, vegetables, and seeds [13,14,15].
The first line therapy for GDM is based on a lifestyle approach with a low glycemic diet and an increase in physical activity. If these measures are not effective in reaching the desired glycemic control, a drug approach with insulin can be started. In view of the above, the role of the diet is crucial during pregnancy. Additionally, other pregnancy-related diseases such as hypertension and fetal growth restriction could be affected by dietary patterns [16].
Several studies have been performed about the effects of different dietary patterns on GDM, but the findings are not conclusive [17,18]. A plant-based diet could represent a suitable option for preventing inflammation through a wide range of antioxidant-rich foods [19]. The literature suggests that a high intake of vegetables, fruits, grains, fish, and legumes, according to the Mediterranean diet (MedDiet), presents a low glycemic pattern and may lower the risk of GDM in a low-risk population [20,21,22].
Zamani et al. revealed that food quality can also impact on GDM. In fact, following unhealthy dietary patterns seems to increase the risk of GDM [23].
The balance between oxidant and antioxidant molecules also represents a pivotal aspect of treating the inflammatory state in GDM [24]. Oxidative stress is characterized by a critical imbalance between antioxidant defenses and reactive oxygen species (ROS) [25]. Hyperglycemia could facilitate ROS production, thus creating an inflammatory state and increasing insulin resistance.
Vascular impairment is another mechanism involved in GDM related to oxidative stress [26]. Chronic exposure to ROS could lead to the increased production of mediators that drive stress-signaling pathways and cause potential tissue damage to key target organs, such as the vasculature and pancreas [26]. Tumor necrosis factor-α (TNF-α) has a pivotal role in insulin resistance as its concentrations are raised in GDM [27].
Despite the few data regarding the role of different dietary patterns in the onset and development of GDM, we aim to assess and deepen the understanding of the impact of a plant-based diet on oxidative stress in GDM.

2. The Impact of a Plant-Based Diet on Gestational Diabetes

The term “plant-based” presents a wide definition as it could either partially include a limited amount of foods derived from animals or include only plant foods such as fruits, vegetables, and legumes [28].
A MedDiet is characterized by food derived from plants without a complete exclusion of animal-source foods. In contrast, both vegetarian and vegan diets exclude meat, chicken, and fish, and a vegan diet additionally excludes dairy and eggs.
A plant-based diet is rich in fibers, magnesium, potassium, and antioxidants but presents a lower intake of saturated fatty acids.
A plant-based diet can exert its role in the prevention of GDM via multiple mechanisms of action. Figure 1 summarizes some beneficial effects of this dietary pattern.
Most dietetic associations agree that a well-planned vegetarian diet presents an adequate amount of nutrients and is helpful for the prevention and treatment of several diseases [29,30]. Although plant-based diets are associated with an increased risk of nutritional deficiencies such as vitamin B12, the available evidence also supports a well-planned vegetarian or vegan diet as a safe option during pregnancy and lactation [31,32]. However, they require strong awareness and monitoring to achieve a balanced intake of all the key nutrients. In a retrospective study of 1419 women, Kesary et al. found that a maternal vegan diet might act as a protective factor from maternal weight gain but that it also increases the occurrence of lower birth weight in the neonate [19]. In a different study, a calorie-restricted vegetarian diet was found to increase insulin sensitivity compared to a conventional diabetic diet over 24 weeks of gestation [33]. Moreover, physical activity along with a low calorie diet had a positive effect on oxidative stress marker levels. A vegetarian diet was also reported to reduce intramyocellular lipid concentrations and visceral fat, favorably impacting on insulin sensitivity and enzymatic oxidative stress markers [33].
Zulyniak et al., in 2017, analyzed 3997 full-term Canadian mothers and found that a plant-based diet was associated with increasing numbers of neonates with a low birth weight in women of Caucasian ethnicity, while, at the same time, the same dietary pattern was associated with increasing numbers of neonates with a higher birth weight in women of Asiatic ethnicity living in Canada [34].
The MedDiet and its role in the prevention of GDM has also been the subject of several investigations. A prospective study by García de la Torre et al. analyzed 1066 normoglycemic women before 12 gestational weeks following a MedDiet with extra-virgin olive oil (EVOO) and pistachio supplementation and found that GDM incidence and maternal–fetal outcomes were lower than in the control group [35]. In a different prospective study including 1076 pregnant women adhering to a MedDiet pattern, better glucose tolerance and a decreased incidence of GDM was highlighted [36].
A case-control study of 299 pregnant women affected by GDM found that a high adherence to the MedDiet before pregnancy was strongly associated with a decreased risk in GDM, suggesting a dose-dependent fashion [37].
Mak et al. performed a prospective cohort study of 1337 Chinese pregnant women and did not find a significant association between the risk of GDM and early pregnancy dietary patterns. However, the authors found that a high protein–low starch diet decreased the risk for GDM among obese women [38].
In a different study, the Dietary Approach to Stop Hypertension (DASH) was found to be effective to prevent GDM in 200 pregnant women [39]. The DASH, which was created to lower blood pressure, emphasizes a lower sodium intake and prefers foods rich in potassium, magnesium, and calcium [39].
Jali et al. analyzed 325 pregnant women undergoing screening for GDM and found out that 52 (16%) presented GDM. Particularly, authors have revealed an increased prevalence of GDM in patients following a non-vegetarian diet compared to a vegetarian diet (65.5% vs. 38.5%) [40].
Another Indian study analyzed 5100 pregnant women and revealed that non-vegetarianism was associated with an increased risk of developing GDM [41].
According to the available evidence, several possible mechanisms could explain the beneficial effects of a plant-based diet on GDM: the presence of fibers and vegetable proteins, a higher intake of antioxidants, a lower intake of saturated fat, and a higher intake of non-heme iron [42,43].
Table 1 summarizes the available studies and the main findings concerning the link between a plant-based diet and GDM.

3. The Role of Insulin Sensitivity in Pregnancy

Pregnancy is characterized by metabolic and immunological changes and by a physiological state of insulin resistance. All these aspects are reversible after delivery [46]. Women who develop GDM usually recover after pregnancy but up to 55% of them will develop T2D during subsequent years [47]. GDM presents similar characteristics to T2D, such as the risk factor of obesity, age, and ethnicity. These findings could suggest that GDM may help in the detection of a genetic susceptibility to develop T2D given the hormonal shift caused by pregnancy.
Several studies have deepened the understanding of the molecular changes involved in insulin resistance in the third trimester of pregnancy, but few data are available about the hormonal interactions during the first and second trimesters [48,49,50].
It seems that during the first trimester of pregnancy insulin secretion increases to promote adipose tissue storage. In the third trimester, there is instead a shift through increased insulin resistance, a rise in free fatty acid (FFA) serum concentrations, and, therefore, a reduction of adipose tissue storage [51,52]. Given the above, insulin exerts a reduced capacity to control lipolysis in the late part of pregnancy, which is more significant in women affected by GDM [51].
Insulin resistance is characterized by a reduction in the ability of the liver, adipose tissue, and muscle to intake adequate glucose. Insulin normally binds the insulin receptor provoking the phosphorylation of the β-subunit receptor and insulin receptor substrate-1 (IRS-1) on at least six tyrosine residues [46].
IRS-1 phosphorylation provokes the binding and phosphorylation of the regulatory subunit p85α of phosphatidylinositol 3-kinase (PI 3-kinase) to IRS-1 [53]. P85α acts as a positive enhancer of the insulin pathway and seems to be relevant for activating glucose transporters (GLUT) in lipid cells [54]. Particularly, levels of the p85α subunit are increased in skeletal muscle and lipid cells in pregnant women with GDM compared to obese non-pregnant women [51].
Pregnant women showed a reduction of IRS-1 expression, and this finding could explain the reduced activity of insulin on its pathway [47].
Friedman et al. found that insulin resistance in pregnancy correlates with a reduced activity of IRS-1 tyrosine phosphorylation [47]. This aspect is related to a reduced expression of IRS-1. Particularly, women affected by GDM present a reduced activity of tyrosine phosphorylation of the β-subunit receptor and an impairment in the glucose transporter [47].
Another mechanism involved in insulin resistance is the increase in IRS-1 serine phosphorylation. This aspect seems to impair the action of insulin on its receptor, the IRS-1 tyrosine phosphorylation, and, therefore, glucose cellular intake [55].
These alterations could represent an initial insulin resistance and also an increased risk to develop T2D during life [47].
Recent studies analyzed the role of apolipoprotein A-1 (ApoA1), the main lipoprotein associated with high density lipoprotein (HDL), in insulin resistance. However, despite the finding of a relationship between HDL and ApoA1 and the activation of pancreatic β-cell function, there were no conclusive findings on this aspect [56,57].
Adipokines, such as adiponectin and leptin, are cytokines secreted by fat cells that are involved in the regulation of insulin secretion as well as in fetal growth [58,59].
Adiponectin improves glucose intake in skeletal muscle cells and decreases glucose secretion in the liver [60]. Moreover, the serum levels of adiponectin decline during pregnancy in both obese and lean women. Given the above, adiponectin could be considered as an insulin-sensitizing agent [60].
Leptin, produced mainly by lipid cells, presents a serum concentration proportional to the adipose tissue percentage [58]. Leptin is also produced by the human placenta, and its receptors were found in both the maternal and fetal surfaces, suggesting an autocrine or paracrine effect on placenta functioning [61].
Thyroid function anomalies could influence glucose homeostasis and insulin resistance in women affected by diabetes [62]. New evidence has suggested that higher concentrations of thyroid-stimulating hormone (TSH) and free triiodothyronine (FT3) as well as an elevated FT3:thyroxine (FT4) ratio could suggest an increased risk of GDM [63].
Other authors have found an association between reduced amylase levels and GDM [64]. This relationship, still defined in patients affected by T2D, could be considered as a predicting factor for GDM [64,65].
So far, the mechanisms by which female hormones such as estrogens and progestins act on insulin resistance during pregnancy is not completely understood.
As it is increased during normal pregnancy, progesterone might play a crucial role in this process. Progesterone inhibits the phosphatidylinositol-3-kinase-mediated pathway by blocking the expression of IRS-1 [43]. Moreover, it suppresses insulin-induced GLUT-4 translocation [43].
Estrogens, particularly 17beta-estradiol, seem to present a complementary role along with progesterone regarding insulin sensitivity. Estrogens are involved in storing and exporting triglycerides from liver, thus impacting on insulin sensitivity [66]. Several studies have analyzed the impact of hormone replacement therapy on improving liver function and the risk of non-alcoholic fatty liver disease in women affected by T2D [67,68].
Human placental lactogen (hPL), secreted by syncytiotrophoblast cells, promotes insulin resistance [69]. It is associated with glucose cell intake, glycogen storage, and oxidation [69]. At the same time, it sustains the storage of energetic supplies for the fetus, thus increasing maternal hyperglycemia.

The Effects of a Plant-Based Diet on Insulin Sensitivity

The impact of diet on glucose homeostasis and therefore on insulin sensitivity has been investigated by several studies in the literature [70,71].
Plant-based diets have spread worldwide in the last few years, according to evidence of such diets preventing T2D, cardiovascular diseases, and cancer [72].
Vegetables and fruits present an important amount of fiber, which is known to reduce gastric emptying and, therefore, lower the glycemic response [73]. Fiber could also positively impact on inflammatory markers and fat storing in the liver [74].
Unsaturated fatty acids, mainly found in olive oil and nuts, improve insulin sensitivity through a reduction of serum lipids and ameliorate the inflammatory response of adipose tissue [75].
Kahleova et al. revealed that a plant-based diet enhances postprandial incretin and insulin secretion [76].
In another study, Bligh et al. found that plant-rich meals significantly increase the serum levels of glucagon-like peptide-1 (GLP-1), a hormone that augments the secretion of insulin, and peptide YY (PYY), while the gastric inhibitory peptide (GIP) level is lowered [77].

4. The Link between Microbiota and Diet

The intestinal microbiota includes trillions of microbic cells and represents a critical aspect of human wellbeing as it seems to modulate the risk of several chronic diseases, such as inflammatory bowel and immune diseases, T2D, cardiovascular disease, and cancer.
The gut microbiota is involved in different activities: protection against pathogenic bacteria and systemic immunomodulation and production of metabolites from foods [78,79].
Moreover, it is fundamental for T-cell and B-cell differentiation in the gut [78].
The microbiota starts to develop immediately after the vaginal passage during birth and many factors contribute to its development, such as age, diet, and genes. Moreover, it has been speculated that the vertical transmission from the mother to the offspring represents another key aspect of this process [80].
The human gut microbiota is characterized by five bacterial groups: Firmicutes, Bacteroides, Proteobacteria, Actinobacteria, and Verrumicrobia. However, the predominant bacteria are Firmicutes and Bacteroides. The microbiota could be considered as an endocrine organ, as it produces metabolites and molecules that impact on several functions [79]. Particularly, through fermentation, gut bacteria produce short-chain free fatty acids that exert immunomodulatory activity [81].
During pregnancy, hormones modulate the composition of gut microbiota: from the first to the third trimester, there is an increase in Proteobacteria and Actinobacteria and an increase in heterogeneity, also called beta diversity [82]. A decreased richness of bacterial populations, or alpha diversity, represents the opposite condition in the last part of the pregnancy [82].
In the third trimester, there is an increase in gut inflammation and hyperglycemia is promoted [82]. This condition has been considered as an adaptative function in order to augment energy storage for fetus development [82].
Another important aspect is the reduction of short-chain-FFA-producing bacteria [82]. This change represents a critical point that permits a rise in the levels of T regulatory cells, a subpopulation of the immune system that preserves tolerance to self-antigens and forestalls autoimmune diseases [82]. In particular, T regulatory cells might prevent maternal rejection of the fetal allograft [83].
The colonization by the uterine microbiota of the placenta and the amniotic fluid is a novel and relevant concept. This condition could explain how a mother’s microbiota colonizes the fetus [84,85]. Moreover, the immune tolerance of pregnancy might disclose the lack of an inflammatory activity against microbial cells in the placenta and the amniotic fluid [84].
The placenta seems to present a reduced microbiota colonization, particularly of non-pathogenic bacteria such as Proteobacteria [86]. This microbiota has been found to be similar to the oral cavity microbiota and could reveal the long-term link between periodontal disease and preterm birth [86]. An impairment of this colonization could also explain the development of intrauterine infections [86].
Other authors have underlined the correlation between bacterial DNA in amniotic fluid and the rise in immune cells [87]. However, other researchers have revealed opposite findings [88].
Given the above, pregnancy is characterized by a change in the gut microbiota that could probably contribute to the development of GDM [89]. Several studies have deepened the connection between the microbiota and dietary patterns, therefore speculating a target therapy.
Prevotella copri and Bacteroides vulgatus represent bacteria that might alter insulin homeostasis, whereas Bacteroides and Staphylococcus aureus are more expressed in obese women than in lean women [85,90]. Other researchers found that in women affected by GDM there was a reduction of Firmicutes species, while the alpha and beta diversity were similar to the control group [91]. Moreover, a different composition in the oral, vaginal, and gut microbiota was found in women affected by GDM compared to healthy pregnancies [92]. Similar findings were revealed by Bassols et al. about the placenta microbiota: in women with GDM, there were less bacteria from Pseudomonadales and Acinetobacter and increased inflammatory expression [93]. In particular, Acinetobacter seems to induce the expression of IL-10—a cytokine with an anti-inflammatory effect and that is involved in the stimulation of T cells and mast cells [93].
Several studies have analyzed the correlation between the microbiota and insulin resistance, but the available evidence is still scarce. Despite this consideration, diet is able to modulate the microbiota composition in a few days and it could represent an intriguing option to prevent the development of GDM.

The Effects of a Plant-Based Diet on Microbiota

A potential beneficial effect of a plant-based diet is the dietary modulation of the gut microbiota. Protein and insoluble fibers impact on the gut microbiota by modulating the immune system and the inflammatory pathways [94]. A diet high in fibers decreases Firmicutes species and increases Bacteroidetes ones, improving short-chain fatty acid production [94]. These changes seem to reduce the incidence of inflammatory diseases and modulate the immune system [94]. Accumulating evidence underlines the impact of the diet on the microbiota. In particular, Barrett et al. revealed that women in early pregnancy and following a vegetarian diet present a different gut microbiota as compared to an omnivorous diet, with a higher abundance of bacteria producing short-chain fatty acids (SCFAs) but without any impact on GDM [44]. At the same time, a diet low in fiber and with a high glycemic intake was proven to increase the risk for GDM in a prospective study of 13,110 patients [45].

5. Oxidative Stress and Insulin Resistance

Oxidative stress remains an important factor in the development of insulin resistance. It represents a normal aspect of a healthy pregnancy, although an abnormal consumption of antioxidants could impair several energetic pathways. Moreover, this aspect is fairly recognized as a critical factor for several pregnancy diseases [95].
Oxidative stress is characterized by a disbalance between antioxidant defenses and oxidant production. The latter category involves ROS radicals, such as superoxide radical (O2−), nitric oxide radical (NO), and hydroxyl radical (˙HO−), and non-radicals, such as hydrogen peroxide (H2O2) and peroxynitrite (ONOO) [25].
Conversely, antioxidant processes involve several enzymes, such as superoxide dismutase (SOD), catalase, thioredoxin, and glutathione peroxidase (GTX) [25]. Non-enzymatic compounds comprise glutathione, ascorbate, and α-tocopherol.
The main causes of oxidative disbalance are the mitochondrial overproduction of ROS, due to high glucose levels and lipid excess, and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase’s increased action through angiotensin II (ANG II) receptors [96].
Several studies have claimed higher levels of oxidative stress and inflammatory markers such as xanthine oxidase, lipid peroxides, malondialdehyde (MDA), 8-isoprostane, TNF-α, and IL-10 in women affected by GDM [97,98] [97,98].
Moreover, systemic insulin resistance, along with oxidative stress, has been correlated not only to an impairment of liver, muscle, and fat tissue but also to central nervous system deterioration [99]. Some authors have revealed that higher levels of insulin modify the blood–brain barrier through a reduced expression of insulin receptors [100]. This mechanism reduces the permeability of insulin through the central nervous system [100].
Particularly, chronic oxidative stress could impact on neuroplasticity and neuronal survival [99]. Insulin plays a critical role in neuronal functioning by participating in neuronal metabolism, as GLUT-4 has been found on neuronal membranes [101]. However, insulin exerts its metabolic effects through the autonomic nervous system on the liver to suppress glucose production or increase triglyceride secretion [101,102].
An Indian research group found that TNF-α is significantly associated with pre-eclampsia in GDM women while increased serum levels of uric acid, IL-8, and TNF-α were associated with maternal–fetal adverse outcomes [103]. In contrast, antioxidants such as serum bilirubin, GTX, and SOD positively correlated with pregnancy outcomes [103].
Jamilian et al. performed a randomized clinical trial (RCT) to analyze the effects of a supplementation of omega-3 fatty acids and vitamin E in 60 women affected by GDM [104]. The outcomes revealed an increase in total antioxidant capacity, MDA, and NO levels and decreased fetal hyperbilirubinemia incidence [104].
Other studies have discovered that in mammalian skeletal muscle cells oxidant stress activates a serine kinase p38 mitogen-activated protein kinase (p38 MAPK) pathway linked to a decrease in insulin sensitivity [96].
Other findings regarding the role of the renin–angiotensin system (RAS) and oxidative stress were presented by Wei et al. They observed that ANG II interferes with the phosphorylation of IRS-1 tyrosine and Protein kinase B (Akt) serine thus impairing insulin-dependent GLUT-4 translocation. This mechanism has been found to be prevented by blocking NADPH oxidase [105]. ANG II can also act through the nuclear factor-κB (NF-κB) pathway to interfere with insulin resistance [106]. Moreover, Akaishi et al. found that the ANG II receptor antagonist reduced ROS concentrations induced by high glucose levels in human renal mesangial cells [107].
Several antioxidants have been studied in insulin resistance. Alpha-lipoic acid (ALA) can reduce the formation in vitro of advanced glycation end products (AGEs), which are markers of oxidative damage and associated with impaired insulin action [108].
Pyridoxamine (PYR), a B6 vitamin family compound, is involved in different metabolic pathways that directly inhibit AGE formation thanks to the ability of trapping reactive carboxyl intermediates, although few and inconclusive data are available on humans [109,110].
Oxidative stress has also been implicated in the development of diabetic complications such as retinopathy, nephropathy, peripheral neuropathy, and heart disease [111]. It alters gene expression and tissue functionality. Increased ROS production by the mitochondria in endothelial cells has a pivotal role in microvasculature damage [96].

The Effects of a Plant-Based Diet on Oxidative Stress

Environment and lifestyle concur to achieve a healthy pregnancy. Moreover, intrauterine development seems to be crucial for preventing several diseases, such as cardiovascular and renal diseases, T2D, and neurological impairment [21]. This particular condition has been studied and defined as “early life programming” [112].
Given the above, the role of the maternal diet, along with sleep quality, physical activity, and avoiding smoking and pollutants, is crucial for the fetus. New evidence has been raised about the benefit of dietary patterns, such as a plant-based diet, on maternal outcomes in pregnancy [113]. Women following a plant-based diet seem to incur in a reduction in pregnancy hypertensive disorders and GDM thanks to the increased intake of fibers [45,114].
Plant-based diets represent a new approach for promoting health. However, the term “plant-based” has a fairly broad definition, as it could either partially include a limited amount of foods derived from animals or include only plant foods such as fruits, nuts, grains, vegetables, and legumes [28].
Several studies have deepened the relationship between this kind of diet and the reduction of the incidence of chronic diseases [115,116,117].
A plant-based diet may increase antioxidant status and nitric oxide bioavailability and decrease ROS, homocysteine serum levels, blood pressure, hyperglycemia, lipids, and even atherosclerosis [118]. Oxidative stress is triggered by an imbalanced redox state caused by mitochondria dysfunction, the overproduction of ROS, or a defective antioxidant system. Several authors have correlated chronic oxidative stress and inflammation with obesity. A healthy dietary pattern is a powerful tool to achieve redox homeostasis. Food and drink with adequate antioxidant compounds, physical activity, and stress management are fundamental aspects for the achievement of a healthy weight and to promote weight loss in obese and overweight people [119]. In view of the above, plant-based diets have spread worldwide in the last few years, according to the knowledge that they might improve the overall quality of life and lead to a healthier status by not only preventing coronary heart disease, cancer, and type 2 diabetes but also ameliorating menopausal complaints in women [73,120,121].
The most widespread plant-based diet is the MedDiet, a dietary pattern associated with a reduced risk of heart disease, metabolic syndrome, diabetes, and cancer [120].
A MedDiet is characterized by food derived from plants such as fruits, vegetables, oils, grains, legumes, beans, and nuts with no absolute exclusion of animal-source foods. In contrast, both vegetarian and vegan diets exclude meat, poultry, and fish, and a vegan diet additionally excludes animal products such as dairy and eggs. A plant-based diet is rich in fibers, dietary nitrates, and cardioprotective micronutrients, such as magnesium and potassium, and antioxidants and low in saturated/trans fats. In contrast, animal foods are typically much lower in nitrates, magnesium, potassium, and antioxidants.
There is observational and interventional evidence that a plant-based diet high in antioxidants, micronutrients, nitrates, and fibers and low in saturated and trans fats may decrease the incidence and severity of several diseases [118,121].

6. Conclusions

Our review highlights that a healthy plant-based diet might favorably impact on the onset of GDM. GDM remains a pregnancy complication influenced both by genetic and inflammatory factors. An imbalanced redox state could act as a trigger to alter insulin sensitivity, and a healthy dietary pattern such as a plant-based one represents a suitable option to improve the intake of antioxidant compounds. However, further studies, especially RCTs, are required to deepen the current knowledge on the interactions between diet and the redox state.

Author Contributions

Conceptualization, A.S. and G.R.; writing—original draft preparation, A.S.; writing—review and editing, G.R., M.L. and M.M.; supervision, M.L. and M.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AGEAdvanced Glycation End Products
AktProtein kinase B
ALAAlpha-lipoic acid
ANG IIAngiotensin II
ApoA1Apolipoprotein A-1
CRPC-Reactive Protein
DASHDietary Approaches To Stop Hypertension
EVOOExtra-Virgin Olive Oil
FFAFree fatty acid
FT3Free Triiodothyronine
FT4Thyroxine
GDMGestational Diabetes Mellitus
GLUTGlucose Transporter
GTXGlutathione Peroxidase
HDLHigh density lipoprotein
HPLHuman Placental Lactogen
I-κBKappa b inhibitor
ILInterleukin
IRS-1Insulin Receptor Substrate 1
MDAMalondialdehyde
MedDietMediterranean Diet
MTNR1BMelatonin Receptor 1B
NAPDHNicotinamide Adenine Dinucleotide Phosphate
NF-κNuclear Factor Kappa
NONitric Oxide
P38 MAPKSerine Kinase P38 Mitogen-activated Protein Kinase
PI 3-kinasePhosphatidylinositol 3-kinase
PYRPyridoxamine
RASRenin–angiotensin system
RCTRandomized Controlled Trial
ROSReactive Oxygen Species
SCFAShort-chain Fatty Acid
SODSuperoxide Dismutase
T2DType 2 Diabetes
TCF7L2Transcription Factor 7-like 2
TNF-αTumor Necrosis Factor-α
TSHThyroid-stimulating hormone

References

  1. Correa, A.; Bardenheier, B.; Elixhauser, A.; Geiss, L.S.; Gregg, E. Trends in Prevalence of Diabetes Among Delivery Hospitalizations, United States, 1993–2009. Matern. Child Health J. 2015, 19, 635–642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Mack, L.R.; Tomich, P.G. Gestational Diabetes. Obstet. Gynecol. Clin. N. Am. 2017, 44, 207–217. [Google Scholar] [CrossRef]
  3. Eades, C.E.; Cameron, D.M.; Evans, J.M. Prevalence of gestational diabetes mellitus in Europe: A meta-analysis. Diabetes Res. Clin. Pr. 2017, 129, 173–181. [Google Scholar] [CrossRef] [PubMed]
  4. Todi, S.; Sagili, H.; Kamalanathan, S.K. Comparison of criteria of International Association of Diabetes and Pregnancy Study Groups (IADPSG) with National Institute for Health and Care Excellence (NICE) for diagnosis of gestational diabetes mellitus. Arch. Gynecol. Obstet. 2020, 302, 47–52. [Google Scholar] [CrossRef]
  5. Committee on Practice Bulletins—Obstetrics. ACOG Practice Bulletin No. 190: Gestational Diabetes Mellitus. Obstet. Gynecol. 2018, 131, e49–e64. [Google Scholar] [CrossRef] [PubMed]
  6. Caughey, A.B.; Cheng, Y.W.; Stotland, N.E.; Washington, A.E.; Escobar, G.J. Maternal and paternal race/ethnicity are both associated with gestational diabetes. Am. J. Obstet. Gynecol. 2010, 202, 616.e1–616.e5. [Google Scholar] [CrossRef] [PubMed]
  7. Robitaille, J.; Grant, A.M. The genetics of gestational diabetes mellitus: Evidence for relationship with type 2 diabetes mellitus. Genet. Med. 2008, 10, 240–250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Williams, M.A.; Qiu, C.; Dempsey, J.C.; Luthy, D.A. Familial aggregation of type 2 diabetes and chronic hypertension in women with gestational diabetes mellitus. J. Reprod. Med. 2003, 48, 955–962. [Google Scholar]
  9. Wu, L.; Cui, L.; Tam, W.H.; Ma, R.C.W.; Wang, C.C. Genetic variants associated with gestational diabetes mellitus: A meta-analysis and subgroup analysis. Sci. Rep. 2016, 6, 1–9. [Google Scholar] [CrossRef]
  10. Xie, K.; Zhang, Y.; Wen, J.; Chen, T.; Kong, J.; Zhang, J.; Wu, X.; Hu, C.; Xu, B.; Ji, C.; et al. Genetic predisposition to gestational glucose metabolism and gestational diabetes mellitus risk in a Chinese population. J. Diabetes 2019, 11, 869–877. [Google Scholar] [CrossRef]
  11. Skórzyńska-Dziduszko, K.E.; Kimber-Trojnar, Ż.; Patro-Małysza, J.; Olszewska, A.; Zaborowski, T.; Małecka-Massalska, T. An Interplay between Obesity and Inflammation in Gestational Diabetes Mellitus. Curr. Pharm. Biotechnol. 2016, 17, 603–613. [Google Scholar] [CrossRef]
  12. Alamolhoda, S.H.; Yazdkhasti, M.; Namdari, M.; Zakariayi, S.J.; Mirabi, P. Association between C-reactive protein and gestational diabetes: A prospective study. J. Obstet. Gynaecol. 2019, 40, 349–353. [Google Scholar] [CrossRef] [PubMed]
  13. Qiu, C.; Zhang, C.; Gelaye, B.; Enquobahrie, D.A.; Frederick, I.O.; Williams, M.A. Gestational Diabetes Mellitus in Relation to Maternal Dietary Heme Iron and Nonheme Iron Intake. Diabetes Care 2011, 34, 1564–1569. [Google Scholar] [CrossRef] [Green Version]
  14. Fu, S.; Li, F.; Zhou, J.; Liu, Z. The Relationship Between Body Iron Status, Iron Intake and Gestational Diabetes. Medicine 2016, 95, e2383. [Google Scholar] [CrossRef] [PubMed]
  15. Rizzo, G.; Garzon, S.; Fichera, M.; Panella, M.M.; Catena, U.; Schiattarella, A.; De Franciscis, P.; Vilos, G.; Tesarik, J.; Török, P.; et al. Vitamin D and Gestational Diabetes Mellitus: Is There a Link? Antioxidants 2019, 8, 511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Donazar-Ezcurra, M.; Burgo, C.L.-D.; Bes-Rastrollo, M. Primary prevention of gestational diabetes mellitus through nutritional factors: A systematic review. BMC Pregnancy Childbirth 2017, 17, 1–5. [Google Scholar] [CrossRef] [Green Version]
  17. Rogozińska, E.; Chamillard, M.; Hitman, G.A.; Khan, K.S.; Thangaratinam, S. Nutritional Manipulation for the Primary Prevention of Gestational Diabetes Mellitus: A Meta-Analysis of Randomised Studies. PLoS ONE 2015, 10, e0115526. [Google Scholar] [CrossRef] [Green Version]
  18. Song, C.; Li, J.; Leng, J.; Ma, R.C.; Yang, X. Lifestyle intervention can reduce the risk of gestational diabetes: A meta-analysis of randomized controlled trials. Obes. Rev. 2016, 17, 960–969. [Google Scholar] [CrossRef]
  19. Kesary, Y.; Avital, K.; Hiersch, L. Maternal plant-based diet during gestation and pregnancy outcomes. Arch. Gynecol. Obstet. 2020, 302, 887–898. [Google Scholar] [CrossRef]
  20. Barger, M.K. Maternal Nutrition and Perinatal Outcomes. J. Midwifery Women’s Health 2010, 55, 502–511. [Google Scholar] [CrossRef]
  21. Pistollato, F.; Cano, S.S.; Elio, I.; Vergara, M.M.; Giampieri, F.; Battino, M. Plant-Based and Plant-Rich Diet Patterns during Gestation: Beneficial Effects and Possible Shortcomings. Adv. Nutr. 2015, 6, 581–591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Assaf-Balut, C.; De La Torre, N.G.; Durán, A.; Fuentes, M.; Bordiú, E.; Del Valle, L.; Familiar, C.; Ortolá, A.; Jiménez, I.; Herraiz, M.A.; et al. A Mediterranean diet with additional extra virgin olive oil and pistachios reduces the incidence of gestational diabetes mellitus (GDM): A randomized controlled trial: The St. Carlos GDM prevention study. PLoS ONE 2017, 12, e0185873. [Google Scholar] [CrossRef] [PubMed]
  23. Zamani, B.; Milajerdi, A.; Tehrani, H.; Bellissimo, N.; Brett, N.R.; Azadbakht, L. Association of a plant-based dietary pattern in relation to gestational diabetes mellitus. Nutr. Diet. 2019, 76, 589–596. [Google Scholar] [CrossRef]
  24. Genc, S.; Kusku-Kiraz, Z.; Dervisoglu, E.; Oztop, N.; Dinccag, N.; Gurdol, F. The Relation of Oxidative Stress Biomarkers with Proinflammatory Cytokines in Gestational Diabetes. Clin. Investig. 2017, 7, 44–48. [Google Scholar] [CrossRef] [Green Version]
  25. Kregel, K.C.; Zhang, H.J. An integrated view of oxidative stress in aging: Basic mechanisms, functional effects, and pathological considerations. Am. J. Physiol. Integr. Comp. Physiol. 2007, 292, R18–R36. [Google Scholar] [CrossRef] [PubMed]
  26. Jensen, L.A.; Chik, C.L.; Ryan, E.A. Review of gestational diabetes mellitus effects on vascular structure and function. Diabetes Vasc. Dis. Res. 2016, 13, 170–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Khambule, L.; George, J.A. The Role of Inflammation in the Development of GDM and the Use of Markers of Inflammation in GDM Screening. Adv. Exp. Med. Biol. 2019, 1134, 217–242. [Google Scholar] [CrossRef]
  28. Shikany, J.M.; Safford, M.M.; Newby, P.K.; Durant, R.W.; Brown, T.M.; Judd, S.E. Southern Dietary Pattern Is Associated with Hazard of Acute Coronary Heart Disease in the Reasons for Geographic and Racial Differences in Stroke (REGARDS) Study. Circulation 2015, 132, 804–814. [Google Scholar] [CrossRef] [PubMed]
  29. Melina, V.; Craig, W.; Levin, S. Position of the Academy of Nutrition and Dietetics: Vegetarian Diets. J. Acad. Nutr. Diet. 2016, 116, 1970–1980. [Google Scholar] [CrossRef]
  30. Craig, W.J.; Mangels, A.R. American Dietetic Association Position of the American Dietetic Association: Vegetarian Diets. J. Am. Diet. Assoc. 2009, 109, 1266–1282. [Google Scholar] [CrossRef]
  31. Sebastiani, G.; Barbero, A.H.; Borràs-Novell, C.; Alsina, M.; Aldecoa-Bilbao, V.; Andreu-Fernández, V.; Tutusaus, M.P.; Martínez, S.F.; Gómez-Roig, M.D.; García-Algar, Ó. The Effects of Vegetarian and Vegan Diet during Pregnancy on the Health of Mothers and Offspring. Nutrients 2019, 11, 557. [Google Scholar] [CrossRef] [Green Version]
  32. Baroni, L.; Rizzo, G.; Goggi, S.; Giampieri, F.; Battino, M. Vegetarian diets during pregnancy: Effects on the mother’s health. A systematic review. Food Funct. 2021, 12, 466–493. [Google Scholar] [CrossRef] [PubMed]
  33. Kahleova, H.; Matoulek, M.; Malinska, H.; Oliyarnik, O.; Kazdova, L.; Neskudla, T.; Skoch, A.; Hajek, M.; Hill, M.; Kahle, M.; et al. Vegetarian diet improves insulin resistance and oxidative stress markers more than conventional diet in subjects with Type 2 diabetes. Diabet. Med. 2011, 28, 549–559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Zulyniak, M.A.; De Souza, R.J.; Shaikh, M.; Desai, D.; Lefebvre, D.L.; Gupta, M.; Wilson, J.; Wahi, G.; Subbarao, P.; Becker, A.B.; et al. Does the impact of a plant-based diet during pregnancy on birth weight differ by ethnicity? A dietary pattern analysis from a prospective Canadian birth cohort alliance. BMJ Open 2017, 7, e017753. [Google Scholar] [CrossRef] [Green Version]
  35. De La Torre, N.G.; Assaf-Balut, C.; Varas, I.J.; Del Valle, L.; Durán, A.; Fuentes, M.; Del Prado, N.; Bordiú, E.; Valerio, J.J.; Herraiz, M.A.; et al. Effectiveness of Following Mediterranean Diet Recommendations in the Real World in the Incidence of Gestational Diabetes Mellitus (GDM) and Adverse Maternal-Foetal Outcomes: A Prospective, Universal, Interventional Study with a Single Group. The St Carlos Study. Nutrition 2019, 11, 1210. [Google Scholar] [CrossRef] [Green Version]
  36. Karamanos, B.; The MGSD-GDM Study Group; Thanopoulou, A.; Anastasiou, E.; Assaad-Khalil, S.; Albache, N.; Bachaoui, M.; Slama, C.B.; El Ghomari, H.; Jotic, A.; et al. Relation of the Mediterranean diet with the incidence of gestational diabetes. Eur. J. Clin. Nutr. 2014, 68, 8–13. [Google Scholar] [CrossRef] [Green Version]
  37. Olmedo-Requena, R.; Gómez-Fernández, J.; Amezcua-Prieto, C.; Mozas-Moreno, J.; Khan, K.S.; Jiménez-Moleón, J.J. Pre-Pregnancy Adherence to the Mediterranean Diet and Gestational Diabetes Mellitus: A Case-Control Study. Nutrition. 2019, 11, 1003. [Google Scholar] [CrossRef] [Green Version]
  38. Mak, J.K.L.; Pham, N.M.; Lee, A.H.; Tang, L.; Pan, X.-F.; Binns, C.W.; Sun, X. Dietary patterns during pregnancy and risk of gestational diabetes: A prospective cohort study in Western China. Nutr. J. 2018, 17, 1–11. [Google Scholar] [CrossRef]
  39. Izadi, V.; Tehrani, H.; Haghighatdoost, F.; Dehghan, A.; Surkan, P.J.; Azadbakht, L. Adherence to the DASH and Mediterranean diets is associated with decreased risk for gestational diabetes mellitus. Nutrition 2016, 32, 1092–1096. [Google Scholar] [CrossRef]
  40. Jali, M.V.; Desai, B.R.; Gowda, S.; Kambar, S.; Jali, S.M. A hospital based study of prevalence of gestational diabetes mellitus in an urban population of India. Eur. Rev. Med. Pharmacol. Sci. 2011, 15, 1306–1310. [Google Scholar] [PubMed]
  41. Arora, G.P.; Thaman, R.G.; Prasad, R.B.; Almgren, P.; Brøns, C.; Groop, L.C.; Vaag, A.A. Prevalence and risk factors of gestational diabetes in Punjab, North India: Results from a population screening program. Eur. J. Endocrinol. 2015, 173, 257–267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. De Filippis, F.; Pellegrini, N.; Vannini, L.; Jeffery, I.B.; La Storia, A.; Laghi, L.; Serrazanetti, D.I.; Di Cagno, R.; Ferrocino, I.; Lazzi, C.; et al. High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut 2016, 65, 1812–1821. [Google Scholar] [CrossRef]
  43. Wada, T.; Hori, S.; Sugiyama, M.; Fujisawa, E.; Nakano, T.; Tsuneki, H.; Nagira, K.; Saito, S.; Sasaoka, T. Progesterone inhibits glucose uptake by affecting diverse steps of insulin signaling in 3T3-L1 adipocytes. Am. J. Physiol. Metab. 2010, 298, E881–E888. [Google Scholar] [CrossRef] [Green Version]
  44. Barrett, H.L.; Gomez-Arango, L.F.; Wilkinson, S.A.; McIntyre, H.D.; Callaway, L.K.; Morrison, M.; Nitert, M.D. A Vegetarian Diet Is a Major Determinant of Gut Microbiota Composition in Early Pregnancy. Nutrition 2018, 10, 890. [Google Scholar] [CrossRef] [Green Version]
  45. Zhang, C.; Liu, S.; Solomon, C.G.; Hu, F.B. Dietary Fiber Intake, Dietary Glycemic Load, and the Risk for Gestational Diabetes Mellitus. Diabetes Care 2006, 29, 2223–2230. [Google Scholar] [CrossRef] [Green Version]
  46. Barbour, L.A.; McCurdy, C.E.; Hernandez, T.L.; Kirwan, J.P.; Catalano, P.M.; Friedman, J.E. Cellular Mechanisms for Insulin Resistance in Normal Pregnancy and Gestational Diabetes. Diabetes Care 2007, 30, S112–S119. [Google Scholar] [CrossRef] [Green Version]
  47. Friedman, J.E.; Ishizuka, T.; Shao, J.; Huston, L.; Highman, T.; Catalano, P. Impaired glucose transport and insulin receptor tyrosine phosphorylation in skeletal muscle from obese women with gestational diabetes. Diabetes 1999, 48, 1807–1814. [Google Scholar] [CrossRef]
  48. Stanley, K.; Fraser, R.; Bruce, C. Physiological changes in insulin resistance in human pregnancy: Longitudinal study with the hyperinsulinaemic euglycaemic clamp technique. Br. J. Obstet. Gynaecol. 1998, 105, 756–759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Mauri, M.; Calmarza, P.; Ibarretxe, D. Dislipemias y embarazo, una puesta al día. Clín. Investig. Arterioscler. 2021, 33, 41–52. [Google Scholar] [CrossRef]
  50. Sonagra, A.D. Normal Pregnancy—A State of Insulin Resistance. J. Clin. Diagn. Res. 2014, 8, CC01. [Google Scholar] [CrossRef] [PubMed]
  51. Catalano, P.M.; Nizielski, S.E.; Shao, J.; Preston, L.; Qiao, L.; Friedman, J.E. Downregulated IRS-1 and PPARγ in obese women with gestational diabetes: Relationship to FFA during pregnancy. Am. J. Physiol. Metab. 2002, 282, E522–E533. [Google Scholar] [CrossRef] [Green Version]
  52. Catalano, P.M.; Huston, L.; Amini, S.B.; Kalhan, S.C. Longitudinal changes in glucose metabolism during pregnancy in obese women with normal glucose tolerance and gestational diabetes mellitus. Am. J. Obstet. Gynecol. 1999, 180, 903–916. [Google Scholar] [CrossRef]
  53. White, M.F.; Kahn, C.R. The insulin signaling system. J. Biol. Chem. 1994, 269, 1–4. [Google Scholar] [CrossRef]
  54. Herbst, J.J.; Andrews, G.C.; Contillo, L.G.; Singleton, D.H.; Genereux, P.E.; Gibbs, E.M.; Lienhard, G.E. Effect of the Activation of Phosphatidylinositol 3-Kinase by a Thiophosphotyrosine Peptide on Glucose Transport in 3T3-L1 Adipocytes. J. Biol. Chem. 1995, 270, 26000–26005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Bandyopadhyay, G.K.; Yu, J.G.; Ofrecio, J.; Olefsky, J.M. Increased p85/55/50 Expression and Decreased Phosphotidylinositol 3-Kinase Activity in Insulin-Resistant Human Skeletal Muscle. Diabetes 2005, 54, 2351–2359. [Google Scholar] [CrossRef] [Green Version]
  56. Retnakaran, R.; Ye, C.; Connelly, P.W.; Hanley, A.J.; Sermer, M.; Zinman, B. Serum apoA1 (Apolipoprotein A-1), Insulin Resistance, and the Risk of Gestational Diabetes Mellitus in Human Pregnancy—Brief Report. Arter. Thromb. Vasc. Biol. 2019, 39, 2192–2197. [Google Scholar] [CrossRef] [PubMed]
  57. Rye, K.-A.; Barter, P.J.; Cochran, B.J. Apolipoprotein A-I interactions with insulin secretion and production. Curr. Opin. Lipidol. 2016, 27, 8–13. [Google Scholar] [CrossRef] [PubMed]
  58. Lea, R.G.; Howe, D.; Hannah, L.T.; Bonneau, O.; Hunter, L.; Hoggard, N. Placental leptin in normal, diabetic and fetal growth-retarded pregnancies. Mol. Hum. Reprod. 2000, 6, 763–769. [Google Scholar] [CrossRef]
  59. Zavalza-Gómez, A.B.; Anaya-Prado, R.; Rincón-Sánchez, A.R.; Mora-Martínez, J.M. Adipokines and insulin resistance during pregnancy. Diabetes Res. Clin. Pract. 2008, 80, 8–15. [Google Scholar] [CrossRef]
  60. Catalano, P.M.; Hoegh, M.; Minium, J.; Huston-Presley, L.; Bernard, S.; Kalhan, S.; Mouzon, S.H.-D. Adiponectin in human pregnancy: Implications for regulation of glucose and lipid metabolism. Diabetologia 2006, 49, 1677–1685. [Google Scholar] [CrossRef] [Green Version]
  61. Akerman, F.; Lei, Z.M.; Rao, C. V Human umbilical cord and fetal membranes co-express leptin and its receptor genes. Gynecol. Endocrinol. 2002, 16, 299–306. [Google Scholar] [CrossRef]
  62. Biondi, B.; Kahaly, G.J.; Robertson, R.P. Thyroid Dysfunction and Diabetes Mellitus: Two Closely Associated Disorders. Endocr. Rev. 2019, 40, 789–824. [Google Scholar] [CrossRef] [Green Version]
  63. Yanachkova, V.E.; Kamenov, Z. The relationship between Thyroid dysfunction during pregnancy and Gestational diabetes mellitus. Endokrynol. Polska 2021. [Google Scholar] [CrossRef]
  64. Zheng, R.; Zhang, J.; Ying, Z.; Zheng, N. Low Serum Amylase is Associated with Gestational Diabetes Mellitus in Chinese Pregnant Women. Clin. Lab. 2015, 59, 64. [Google Scholar] [CrossRef]
  65. Yu, F.; Zhou, W.; Tan, X.; Jiang, Y. Gestational Diabetes Mellitus is Associated with Plasma Amylase in a Chinese Pregnant Women Population. Clin. Lab. 2019, 65, 65. [Google Scholar] [CrossRef]
  66. Palmisano, B.T.; Zhu, L.; Stafford, J.M. Role of Estrogens in the Regulation of Liver Lipid Metabolism. Adv. Exper. Med. Biol. 2017, 1043, 227–256. [Google Scholar] [CrossRef] [Green Version]
  67. McKenzie, J.; Fisher, B.M.; Jaap, A.J.; Stanley, A.; Paterson, K.; Sattar, N. Effects of HRT on liver enzyme levels in women with type 2 diabetes: A randomized placebo-controlled trial. Clin. Endocrinol. 2006, 65, 40–44. [Google Scholar] [CrossRef]
  68. Khoo, C.L.; Perera, M. Diabetes and the menopause. Br. Menopause Soc. J. 2005, 11, 6–11. [Google Scholar] [CrossRef]
  69. Sibiak, R.; Jankowski, M.; Gutaj, P.; Mozdziak, P.; Kempisty, B.; Wender-Ożegowska, E. Placental Lactogen as a Marker of Maternal Obesity, Diabetes, and Fetal Growth Abnormalities: Current Knowledge and Clinical Perspectives. J. Clin. Med. 2020, 9, 1142. [Google Scholar] [CrossRef]
  70. Kahleova, H.; Tura, A.; Hill, M.; Holubkov, R.; Barnard, N.D. A Plant-Based Dietary Intervention Improves Beta-Cell Function and Insulin Resistance in Overweight Adults: A 16-Week Randomized Clinical Trial. Nutrition 2018, 10, 189. [Google Scholar] [CrossRef] [Green Version]
  71. Barnard, N.D.; Scialli, A.R.; Turner-McGrievy, G.; Lanou, A.J.; Glass, J. The effects of a low-fat, plant-based dietary intervention on body weight, metabolism, and insulin sensitivity. Am. J. Med. 2005, 118, 991–997. [Google Scholar] [CrossRef]
  72. Kahleova, H.; Levin, S.; Barnard, N. Cardio-Metabolic Benefits of Plant-Based Diets. Nutrition 2017, 9, 848. [Google Scholar] [CrossRef]
  73. Livesey, G.; Tagami, H. Interventions to lower the glycemic response to carbohydrate foods with a low-viscosity fiber (resistant maltodextrin): Meta-analysis of randomized controlled trials. Am. J. Clin. Nutr. 2008, 89, 114–125. [Google Scholar] [CrossRef] [Green Version]
  74. Wannamethee, S.G.; Whincup, P.H.; Thomas, M.C.; Sattar, N. Associations Between Dietary Fiber and Inflammation, Hepatic Function, and Risk of Type 2 Diabetes in Older Men: Potential mechanisms for the benefits of fiber on diabetes risk. Diabetes Care 2009, 32, 1823–1825. [Google Scholar] [CrossRef] [Green Version]
  75. Mirabelli, M.; Chiefari, E.; Arcidiacono, B.; Corigliano, D.M.; Brunetti, F.S.; Maggisano, V.; Russo, D.; Foti, D.P.; Brunetti, A. Mediterranean Diet Nutrients to Turn the Tide against Insulin Resistance and Related Diseases. Nutrients 2020, 12, 1066. [Google Scholar] [CrossRef] [Green Version]
  76. Kahleova, H.; Tura, A.; Klementova, M.; Thieme, L.; Haluzik, M.; Pavlovicova, R.; Hill, M.; Pelikanova, T. A Plant-Based Meal Stimulates Incretin and Insulin Secretion More Than an Energy- and Macronutrient-Matched Standard Meal in Type 2 Diabetes: A Randomized Crossover Study. Nutrients 2019, 11, 486. [Google Scholar] [CrossRef] [Green Version]
  77. Bligh, H.F.J.; Godsland, I.F.; Frost, G.; Hunter, K.J.; Murray, P.; Macaulay, K.; Hyliands, D.; Talbot, D.C.S.; Casey, J.; Mulder, T.P.J.; et al. Plant-rich mixed meals based on Palaeolithic diet principles have a dramatic impact on incretin, peptide YY and satiety response, but show little effect on glucose and insulin homeostasis: An acute-effects randomised study. Br. J. Nutr. 2015, 113, 574–584. [Google Scholar] [CrossRef]
  78. Taddei, C.R.; Cortez, R.V.; Mattar, R.; Torloni, M.R.; Daher, S. Microbiome in normal and pathological pregnancies: A literature overview. Am. J. Reprod. Immunol. 2018, 80, e12993. [Google Scholar] [CrossRef]
  79. Schiattarella, A.; Riemma, G.; La Verde, M.; Franci, G.; Chianese, A.; Fasulo, D.; Fichera, M.; Gallo, P.; De Franciscis, P. Polycystic Ovary Syndrome and Probiotics: A Natural Approach to an Inflammatory Disease. Curr. Women’s Health Rev. 2021, 17, 14–20. [Google Scholar] [CrossRef]
  80. Blaser, M.J. Who are we? EMBO Rep. 2006, 7, 956–960. [Google Scholar] [CrossRef] [Green Version]
  81. McKenzie, C.; Tan, J.; Macia, L.; Mackay, C.R. The nutrition-gut microbiome-physiology axis and allergic diseases. Immunol. Rev. 2017, 278, 277–295. [Google Scholar] [CrossRef]
  82. Koren, O.; Goodrich, J.K.; Cullender, T.C.; Spor, A.; Laitinen, K.; Bäckhed, H.K.; Gonzalez, A.; Werner, J.J.; Angenent, L.T.; Knight, R.; et al. Host Remodeling of the Gut Microbiome and Metabolic Changes during Pregnancy. Cell 2012, 150, 470–480. [Google Scholar] [CrossRef] [Green Version]
  83. Blaser, M.J.; Dominguez-Bello, M.G. The Human Microbiome before Birth. Cell Host Microbe 2016, 20, 558–560. [Google Scholar] [CrossRef] [Green Version]
  84. Ardissone, A.N.; De La Cruz, D.M.; Davis-Richardson, A.G.; Rechcigl, K.T.; Li, N.; Drew, J.C.; Murgas-Torrazza, R.; Sharma, R.; Hudak, M.L.; Triplett, E.W.; et al. Meconium Microbiome Analysis Identifies Bacteria Correlated with Premature Birth. PLoS ONE 2014, 9, e90784. [Google Scholar] [CrossRef] [Green Version]
  85. Collado, M.C.; Isolauri, E.; Laitinen, K.; Salminen, S. Distinct composition of gut microbiota during pregnancy in overweight and normal-weight women. Am. J. Clin. Nutr. 2008, 88, 894–899. [Google Scholar] [CrossRef]
  86. Aagaard, K.; Ma, J.; Antony, K.M.; Ganu, R.; Petrosino, J.; Versalovic, J. The Placenta Harbors a Unique Microbiome. Sci. Transl. Med. 2014, 6, 237ra65. [Google Scholar] [CrossRef] [Green Version]
  87. Han, Y.W.; Shen, T.; Chung, P.; Buhimschi, I.A.; Buhimschi, C.S. Uncultivated Bacteria as Etiologic Agents of Intra-Amniotic Inflammation Leading to Preterm Birth. J. Clin. Microbiol. 2008, 47, 38–47. [Google Scholar] [CrossRef] [Green Version]
  88. Lauder, A.P.; Roche, A.M.; Sherrill-Mix, S.; Bailey, A.; Laughlin, A.L.; Bittinger, K.; Leite, R.; Elovitz, M.A.; Parry, S.; Bushman, F.D. Comparison of placenta samples with contamination controls does not provide evidence for a distinct placenta microbiota. Microbiome 2016, 4, 1–11. [Google Scholar] [CrossRef] [Green Version]
  89. Kuang, Y.-S.; Lu, J.-H.; Li, S.-H.; Li, J.-H.; Yuan, M.-Y.; He, J.-R.; Chen, N.-N.; Xiao, W.-Q.; Shen, S.-Y.; Qiu, L.; et al. Connections between the human gut microbiome and gestational diabetes mellitus. GigaScience 2017, 6, 1–12. [Google Scholar] [CrossRef]
  90. Pedersen, H.K.; Gudmundsdottir, V.; Nielsen, H.B.; Hyotylainen, T.; Nielsen, T.; Jensen, B.A.H.; Forslund, K.; Hildebrand, F.; Prifti, E.; Falony, G.; et al. Human gut microbes impact host serum metabolome and insulin sensitivity. Nat. Cell Biol. 2016, 535, 376–381. [Google Scholar] [CrossRef]
  91. Fugmann, M.; Breier, M.; Rottenkolber, M.; Banning, F.; Ferrari, U.; Sacco, V.; Grallert, H.; Parhofer, K.G.; Seissler, J.; Clavel, T.; et al. The stool microbiota of insulin resistant women with recent gestational diabetes, a high risk group for type 2 diabetes. Sci. Rep. 2015, 5, 1–8. [Google Scholar] [CrossRef]
  92. Acuna, J.; Cohavy, O.; Solt, I.; Reeder, J.; Kim, M.; Lebovics, I.; Paster, B.; Knight, R.; Rotmensch, S. 257: Preliminary observations on the microbial phylogeny of the oral, vaginal, and rectal microbiome in gestational diabetes and healthy pregnancies. Am. J. Obstet. Gynecol. 2011, 204, S109–S110. [Google Scholar] [CrossRef]
  93. Bassols, J.; Serino, M.; Carreras-Badosa, G.; Burcelin, R.; Blasco-Baque, V.; Lopez-Bermejo, A.; Fernandez-Real, J.-M. Gestational diabetes is associated with changes in placental microbiota and microbiome. Pediatr. Res. 2016, 80, 777–784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Thorburn, A.N.; Macia, L.; Mackay, C.R. Diet, Metabolites, and “Western-Lifestyle” Inflammatory Diseases. Immunity 2014, 40, 833–842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Sultana, Z.; Maiti, K.; Aitken, J.; Morris, J.; Dedman, L.; Smith, R. Oxidative stress, placental ageing-related pathologies and adverse pregnancy outcomes. Am. J. Reprod. Immunol. 2017, 77, e12653. [Google Scholar] [CrossRef] [Green Version]
  96. Henriksen, E.J.; Diamond-Stanic, M.K.; Marchionne, E.M. Oxidative stress and the etiology of insulin resistance and type 2 diabetes. Free Radic. Biol. Med. 2011, 51, 993–999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Ozler, S.; Oztas, E.; Uygur, D.; Ersoy, A.O.; Ergin, M.; Koca, C.; Danisman, N.; Erkaya, S. The Value of Total antioxidant Status and Serum Tumor Necrosis Factor-α Levels at 24–28 Weeks of Gestation in the Prediction of Optimal Treatment Protocol in Gestational Diabetes Mellitus. Exp. Clin. Endocrinol. Diabetes 2015, 127, 485–491. [Google Scholar] [CrossRef]
  98. Shang, M.; Zhao, J.; Yang, L.; Lin, L. Oxidative stress and antioxidant status in women with gestational diabetes mellitus diagnosed by IADPSG criteria. Diabetes Res. Clin. Pr. 2015, 109, 404–410. [Google Scholar] [CrossRef]
  99. Maciejczyk, M.; Żebrowska, E.; Chabowski, A. Insulin Resistance and Oxidative Stress in the Brain: What’s New? Int. J. Mol. Sci. 2019, 20, 874. [Google Scholar] [CrossRef] [Green Version]
  100. Arnold, S.E.; Lucki, I.; Brookshire, B.R.; Carlson, G.C.; Browne, C.A.; Kazi, H.; Bang, S.; Choi, B.-R.; Chen, Y.; McMullen, M.F.; et al. High fat diet produces brain insulin resistance, synaptodendritic abnormalities and altered behavior in mice. Neurobiol. Dis. 2014, 67, 79–87. [Google Scholar] [CrossRef] [Green Version]
  101. Banks, W.A.; Owen, J.B.; Erickson, M.A. Insulin in the brain: There and back again. Pharmacol. Ther. 2012, 136, 82–93. [Google Scholar] [CrossRef] [Green Version]
  102. Scherer, T.; O’Hare, J.; Diggs-Andrews, K.; Schweiger, M.; Cheng, B.; Lindtner, C.; Zielinski, E.; Vempati, P.; Su, K.; Dighe, S.; et al. Brain Insulin Controls Adipose Tissue Lipolysis and Lipogenesis. Cell Metab. 2011, 13, 183–194. [Google Scholar] [CrossRef] [Green Version]
  103. Murthy, K.A.S.; Bhandiwada, A.; Chandan, S.L.; Gowda, S.L.; Sindhusree, G. Evaluation of oxidative stress and proinflammatory cytokines in gestational diabetes mellitus and their correlation with pregnancy outcome. Indian J. Endocrinol. Metab. 2018, 22, 79–84. [Google Scholar] [CrossRef]
  104. Jamilian, M.; Dizaji, S.H.; Bahmani, F.; Taghizadeh, M.; Memarzadeh, M.R.; Karamali, M.; Akbari, M.; Asemi, Z. A Randomized Controlled Clinical Trial Investigating the Effects of Omega-3 Fatty Acids and Vitamin E Co-Supplementation on Biomarkers of Oxidative Stress, Inflammation and Pregnancy Outcomes in Gestational Diabetes. Can. J. Diabetes 2017, 41, 143–149. [Google Scholar] [CrossRef] [PubMed]
  105. Wei, Y.; Sowers, J.R.; Nistala, R.; Gong, H.; Uptergrove, G.M.-E.; Clark, S.E.; Morris, E.M.; Szary, N.; Manrique, C.; Stump, C.S. Angiotensin II-induced NADPH Oxidase Activation Impairs Insulin Signaling in Skeletal Muscle Cells. J. Biol. Chem. 2006, 281, 35137–35146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Wei, Y.; Sowers, J.R.; Clark, S.E.; Li, W.; Ferrario, C.M.; Stump, C.S. Angiotensin II-induced skeletal muscle insulin resistance mediated by NF-κB activation via NADPH oxidase. Am. J. Physiol. Metab. 2008, 294, E345–E351. [Google Scholar] [CrossRef] [Green Version]
  107. Akaishi, T.; Abe, M.; Okuda, H.; Ishizawa, K.; Abe, T.; Ishii, T.; Ito, S. High glucose level and angiotensin II type 1 receptor stimulation synergistically amplify oxidative stress in renal mesangial cells. Sci. Rep. 2019, 9, 1–7. [Google Scholar] [CrossRef]
  108. Ghelani, H.; Razmovski-Naumovski, V.; Pragada, R.R.; Nammi, S. (R)-α-Lipoic acid inhibits fructose-induced myoglobin fructation and the formation of advanced glycation end products (AGEs) in vitro. BMC Complement. Altern. Med. 2018, 18, 1–11. [Google Scholar] [CrossRef]
  109. Lewis, E.J.; Greene, T.; Spitalewiz, S.; Blumenthal, S.; Berl, T.; Hunsicker, L.G.; Pohl, M.A.; Rohde, R.D.; Raz, I.; Yerushalmy, Y.; et al. Pyridorin in Type 2 Diabetic Nephropathy. J. Am. Soc. Nephrol. 2011, 23, 131–136. [Google Scholar] [CrossRef] [Green Version]
  110. Williams, M.E.; Bolton, W.K.; Khalifah, R.G.; Degenhardt, T.P.; Schotzinger, R.J.; McGill, J.B. Effects of Pyridoxamine in Combined Phase 2 Studies of Patients with Type 1 and Type 2 Diabetes and Overt Nephropathy. Am. J. Nephrol. 2007, 27, 605–614. [Google Scholar] [CrossRef]
  111. Karam, B.S.; Chavez-Moreno, A.; Koh, W.; Akar, J.G.; Akar, F.G. Oxidative stress and inflammation as central mediators of atrial fibrillation in obesity and diabetes. Cardiovasc. Diabetol. 2017, 16, 1–9. [Google Scholar] [CrossRef] [PubMed]
  112. Capra, L.; Tezza, G.; Mazzei, F.; Boner, A.L. The origins of health and disease: The influence of maternal diseases and lifestyle during gestation. Ital. J. Pediatr. 2013, 39, 1–12. [Google Scholar] [CrossRef] [Green Version]
  113. Piccoli, G.B.; Clari, R.; Vigotti, F.N.; Leone, F.; Attini, R.; Cabiddu, G.; Mauro, G.; Castelluccia, N.; Colombi, N.; Capizzi, I.; et al. Vegan-vegetarian diets in pregnancy: Danger or panacea? A systematic narrative review. BJOG Int. J. Obstet. Gynaecol. 2015, 122, 623–633. [Google Scholar] [CrossRef] [PubMed]
  114. Longo-Mbenza, B.; Kadima-Tshimanga, B.; Buassa-Bu-Tsumbu, B.; M’Buyamba, K. Diets rich in vegetables and physical activity are associated with a decreased risk of pregnancy induced hypertension among rural women from Kimpese, DR Congo. Niger. J. Med. 2008, 17, 45–49. [Google Scholar]
  115. Chuang, S.-Y.; Chiu, T.H.; Lee, C.-Y.; Liu, T.-T.; Tsao, C.K.; Hsiung, C.A.; Chiu, Y.-F. Vegetarian diet reduces the risk of hypertension independent of abdominal obesity and inflammation. J. Hypertens. 2016, 34, 2164–2171. [Google Scholar] [CrossRef]
  116. Orlich, M.J.; Singh, P.N.; Sabaté, J.; Fan, J.; Sveen, L.; Bennett, H.; Knutsen, S.F.; Beeson, W.L.; Jaceldo-Siegl, K.; Butler, T.L.; et al. Vegetarian Dietary Patterns and the Risk of Colorectal Cancers. JAMA Intern. Med. 2015, 175, 767–776. [Google Scholar] [CrossRef] [PubMed]
  117. Tantamango-Bartley, Y.; Knutsen, S.F.; Knutsen, R.; Jacobsen, B.K.; Fan, J.; Beeson, W.L.; Sabate, J.; Hadley, D.; Jaceldo-Siegl, K.; Penniecook, J.; et al. Are strict vegetarians protected against prostate cancer? Am. J. Clin. Nutr. 2016, 103, 153–160. [Google Scholar] [CrossRef] [Green Version]
  118. Kerley, C.P. A Review of Plant-based Diets to Prevent and Treat Heart Failure. Card. Fail. Rev. 2018, 4, 54–61. [Google Scholar] [CrossRef]
  119. Tobore, T.O. Towards a comprehensive theory of obesity and a healthy diet: The causal role of oxidative stress in food addiction and obesity. Behav. Brain Res. 2020, 384, 112560. [Google Scholar] [CrossRef] [PubMed]
  120. Davis, C.R.; Bryan, J.; Hodgson, J.M.; Murphy, K.J. Definition of the Mediterranean Diet; A Literature Review. Nutrients 2015, 7, 9139–9153. [Google Scholar] [CrossRef]
  121. De Franciscis, P.; Conte, A.; Schiattarella, A.; Riemma, G.; Cobellis, L.; Colacurci, N. Non-hormonal Treatments for Menopausal Symptoms and Sleep Disturbances: A Comparison Between Purified Pollen Extracts and Soy Isoflavones. Curr. Pharm. Des. 2020, 26, 4509–4514. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Beneficial influences of a plant-based diet on gestational diabetes mellitus (Images by smart.sevier.com).
Figure 1. Beneficial influences of a plant-based diet on gestational diabetes mellitus (Images by smart.sevier.com).
Antioxidants 10 00557 g001
Table 1. Main findings of the studies concerning the impact of a plant-based diet on gestational diabetes mellitus.
Table 1. Main findings of the studies concerning the impact of a plant-based diet on gestational diabetes mellitus.
ReferenceType of StudyMain OutcomeNumber of ParticipantsEventDefinition of Plant-Based Diet
Arora et al., India [41]Observational, cross-sectionalAn increased risk of developing GDM was associated with a non-vegetarian diet5100 womenThe prevalence of GDM was 35% using WHO 2013 criteriaVegetarian diet
Barrett et al., Australia [44]RCTA vegetarian diet in early pregnancy increased the presence of short-chain fatty acid bacteria producers without any influence on GDM risk9 following a vegetarian diet and 18 an omnivorous oneMicrobiome alpha diversity was similar, while beta diversity was reduced, in vegetariansVegetarian diet
De Filippis et al., Italy [42]ObservationalAn increased consumption of plant foodstuffs based on a MedDiet was associated with beneficial microbiota improvements51 vegetarians, 51 vegans, and 51 omnivoresPositive correlation between consumption of vegetables and short-chain fatty acids, Prevotella, and Firmicutes in the gut microbiomeMedDiet
García de la Torre et al., Spain [35]Observational, prospectiveFollowing a MedDiet with EVOO and pistachio supplementation before 12 gestational weeks showed a lower GDM incidence and better maternal–fetal outcomes932 womenThe incidence of GDM was lower in the intervention group than in the controls (RR 0.81)MedDiet
Izadi et al., Iran [39]Observational, case-controlAdherence to the DASH and MedDiet was associated with a reduced risk for GDM200 women with GDM and 260 without GDMA higher adherence to DASH was related to 71% reduced risk for GDMDASH and MedDiet
Jali et al., India [40]Observational, cross-sectionalNon-vegetarian pregnant women showed an increased risk for glucose intolerance325 women: 202 vegetarian and 123 non-vegetarian52 women (16%) presented GDM. An increased prevalence of GDM in patients following a non-vegetarian diet compared to a vegetarian diet (65.5% vs. 38.5%)Vegetarian diet
Kahleova et al., Czech Republic [33]RCTA low calorie vegetarian diet improved insulin sensitivity37 following a vegetarian diet and 37 following a conventional diabetic dietA vegetarian diet improved adipokine levels and oxidative stress markers compared to a conventional diabetic diet over 24 weeksVegetarian diet
Karamanos et al., Mediterranean countries [36]Observational, prospectiveAdhering to a MedDiet pattern decreased the incidence of GDM1076 womenThe incidence of GDM was lower in subjects with better adherence to the MedDiet (8.0% vs. 12.3%)MedDiet
Kesary et al., Israel [19]Observational, retrospectiveA vegan diet is a protective factor from maternal weight gain but increased the risk for a lower birth weight234 vegans, 133 vegetarian, and 1052 omnivoresA vegan diet in pregnancy was associated with a lower birth weight centile compared to omnivores (42.6 ± 25.9 vs. 52.5 ± 27.0; p  <  0.001)Vegan and vegetarian diet
Mak et al., China [38]Observational, prospectiveFollowing an early pregnancy dietary pattern did not significantly increase the risk of GDM in patients. However, a high protein–low starch diet was associated with a decrease in risk for GDM among obese women1337 women199 women (14.9%) developed GDMPlant-based and a high protein–low starch pattern diet
Olmedo-Requena et al., Spain [37]Observational, case-controlA high adherence to a MedDiet before pregnancy was strongly associated with a decreased risk in GDM291 with GDM and 1175 without GDMA high MedDiet adherence was associated with lower GDM risk (aOR 0.61; p = 0.028), while a very high MedDiet adherence was more strongly associated (aOR 0.33; p = 0.005)MedDiet
Zhang et al., USA [45]Observational, prospectiveA low fiber and high sugar intake diet increased the risk for GDM13,100 women758 with GDM. Each 10-g/day increment in total fiber intake was associated with a 26% reduction in GDM riskDiet rich in fiber
Zulyniak et al., Canada [34]Observational, prospectiveA plant-based diet was associated with lowering the birth weight for women of Caucasian ethnicity and increasing it in Asiatic women living in Canada3997 womenThe plant-based diet was inversely associated with birth weight (β = −67.6 g per 1-unit increase; p < 0.001)Plant-based diet
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Schiattarella, A.; Lombardo, M.; Morlando, M.; Rizzo, G. The Impact of a Plant-Based Diet on Gestational Diabetes: A Review. Antioxidants 2021, 10, 557. https://doi.org/10.3390/antiox10040557

AMA Style

Schiattarella A, Lombardo M, Morlando M, Rizzo G. The Impact of a Plant-Based Diet on Gestational Diabetes: A Review. Antioxidants. 2021; 10(4):557. https://doi.org/10.3390/antiox10040557

Chicago/Turabian Style

Schiattarella, Antonio, Mauro Lombardo, Maddalena Morlando, and Gianluca Rizzo. 2021. "The Impact of a Plant-Based Diet on Gestational Diabetes: A Review" Antioxidants 10, no. 4: 557. https://doi.org/10.3390/antiox10040557

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