Hostname: page-component-8448b6f56d-c4f8m Total loading time: 0 Render date: 2024-04-15T09:09:31.815Z Has data issue: false hasContentIssue false

Could gestational diabetes mellitus be managed through dietary bioactive compounds? Current knowledge and future perspectives

Published online by Cambridge University Press:  16 February 2016

Carmela Santangelo*
Affiliation:
Department of Veterinary Public Health and Food Safety, Unit of Nutrition, Italian National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy
Alessandra Zicari
Affiliation:
Department of Experimental Medicine, 2nd Section of Cell Pathology, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
Elisabetta Mandosi
Affiliation:
Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
Beatrice Scazzocchio
Affiliation:
Department of Veterinary Public Health and Food Safety, Unit of Nutrition, Italian National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy
Emanuela Mari
Affiliation:
Department of Experimental Medicine, 2nd Section of Cell Pathology, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
Susanna Morano
Affiliation:
Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
Roberta Masella
Affiliation:
Department of Veterinary Public Health and Food Safety, Unit of Nutrition, Italian National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy
*
*Corresponding author: C. Santangelo, email carmela.santangelo@iss.it
Rights & Permissions [Opens in a new window]

Abstract

Gestational diabetes mellitus (GDM) is a serious problem growing worldwide that needs to be addressed with urgency in consideration of the resulting severe complications for both mother and fetus. Growing evidence indicates that a healthy diet rich in fruit, vegetables, nuts, extra-virgin olive oil and fish has beneficial effects in both the prevention and management of several human diseases and metabolic disorders. In this review, we discuss the latest data concerning the effects of dietary bioactive compounds such as polyphenols and PUFA on the molecular mechanisms regulating glucose homoeostasis. Several studies, mostly based on in vitro and animal models, indicate that dietary polyphenols, mainly flavonoids, positively modulate the insulin signalling pathway by attenuating hyperglycaemia and insulin resistance, reducing inflammatory adipokines, and modifying microRNA (miRNA) profiles. Very few data about the influence of dietary exposure on GDM outcomes are available, although this approach deserves careful consideration. Further investigation, which includes exploring the ‘omics’ world, is needed to better understand the complex interaction between dietary compounds and GDM.

Type
Full Papers
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
Copyright © The Authors 2016

Gestational diabetes mellitus (GDM) is the most common metabolic disorder during pregnancy( Reference Chen, Wang and Ji 1 ), and its prevalence is increasing worldwide( Reference Yuen and Wong 2 ). Women with GDM are at a high risk of developing type 2 diabetes (T2D) later in life( Reference Chen, Wang and Ji 1 ); in addition, the higher baseline BMI and weight gain often found after GDM occurrence increase the risk of progression from GDM to T2D( Reference Bao, Yeung and Tobias 3 ). Moreover, uncontrolled GDM is associated with a detrimental intra-uterine environment, which leads to fetal complications and an increased risk for the child of developing obesity and metabolic disorders( Reference Harris, Weston and Harding 4 , Reference Yessoufou and Moutairou 5 ).

In response to the marked rise in GDM, it is of paramount importance to identify appropriate treatments to prevent maternal and fetal complications associated with this disease.

At present, the management of GDM is a big challenge because of its heterogeneity (i.e. ethnic as well as intra-, and inter-country differences)( Reference Yuen and Wong 2 , Reference Benhalima, Mathieu and Damm 6 , Reference Agarwal 7 ), and the incomplete knowledge of its pathophysiology( Reference Sacks, Hadden and Maresh 8 ). As a result, standardised guidelines for GDM are difficult to arrive at worldwide, as are the intervention strategies aimed at preventing or/and reducing the burden of this disorder. In addition, obesity and maternal overweight have been considered as the main risk factors for GDM( Reference Chen, Wang and Ji 1 ); the reduction of these risk factors is essential for the well-being of mother and offspring( Reference Yessoufou and Moutairou 5 ).

Pre-pregnancy and early pregnancy are the best periods for a dietary intervention to control weight in order to prevent the long-lasting effects of maternal diabetes or obesity( Reference Santangelo, Vari and Scazzocchio 9 ).

There is increasing evidence that the dietary patterns having beneficial effects both in prevention and management of diabetes are characterised by high consumption of plant foods (e.g. whole grains, fruit, vegetables and extra-virgin olive oil, nuts) and fish, and low consumption of animal-based, high-fat, processed foods, that is, the Mediterranean-style diet (MedDiet)( Reference Georgoulis, Kontogianni and Yiannakouris 10 ). The MedDiet is a primarily plant-based dietary pattern that has been strongly associated with lower incidence of CVD, and neoplastic diseases, and an overall reduced mortality( Reference Sofi, Macchi and Abbate 11 , Reference Capurso, Massaro and Scoditti 12 ). Adherence to the MedDiet correlates with better glycaemic control, a reduced risk of both total and cardiovascular mortality in diabetic subjects in Mediterranean populations( Reference Esposito, Maiorino and Bellastella 13 Reference Koloverou, Panagiotakos and Pitsavos 15 ) and a lower risk of metabolic syndrome and CVD in non-Mediterranean populations( Reference Yang, Farioli and Korre 16 Reference Grosso, Stepaniak and Micek 19 ). These findings indicate that the adoption of the MedDiet model by populations having different dietary habits is effective in reducing the risk of non-communicable disorders.

The short-term – 4 weeks – consumption of a DASH diet (Dietary Approaches to Stop Hypertension), rich in fruits, vegetables, whole grains and lower amounts of SFA, improved pregnancy outcomes among GDM women( Reference Asemi, Samimi and Tabassi 20 ). Pre-pregnancy adherence to healthy dietary patterns( Reference Tobias, Zhang and Chavarro 21 ) and the MedDiet is associated with lower incidence of GDM and a better degree of glucose tolerance in no-GDM pregnant women( Reference Karamanos, Thanopoulou and Anastasiou 22 ).

In any case, the mechanisms underlying the protective effects of the MedDiet are not clear as yet. The high MUFA:SFA ratio, the increased level of PUFA, the low content of trans-fatty acids (FA) and the high content of fibres, vitamins, mineral salts and phytochemicals compounds may contribute to the beneficial effects of MedDiet( Reference Salas-Salvado, Martinez-Gonzalez and Bullo 23 Reference Wang, Jiang and Yang 25 ). Over the past years, researchers have focused their attention on the role of plant-derived, functional foods and their bioactive compounds in the control of various aspects of diabetes mellitus( Reference Mirmiran, Bahadoran and Azizi 26 , Reference Xiao and Hogger 27 ). Among the known natural bioactive components, polyphenols have been shown to have anti-hyperglycaemic effects, antioxidant and anti-inflammatory activities and no side effects( Reference Xiao and Hogger 27 Reference Oh and Jun 29 ). In addition, the increasing demand of non-fish source of n-3 PUFA is worth considering, in view of the beneficial effects of these FA in GDM women( Reference Samimi, Jamilian and Asemi 24 ), and during pregnancy in general, not to mention the worldwide increase in the number of vegetarians and vegans( Reference Saunders, Davis and Garg 30 ).

The knowledge of how (i.e. molecular mechanisms) and where (i.e. targets) a given biocompound acts is of crucial importance to better understand the mechanisms governing the dietary impact on the metabolic system in GDM.

We found a good number of articles in English published up to August 2015 by searching in PubMed, using the key words ‘gestational diabetes mellitus’, ‘diabetes’, ‘insulin resistance’, ‘hyperglycaemia’, ‘adipokines’, ‘inflammation’, ‘microRNAs’, ‘PUFAs’, molecular mechanisms’; these key words were searched in combination with the key words ‘Mediterranean diet’, vegetables food’, ‘polyphenols’, ‘phytochemicals’, ‘bioactive compounds’. Data regarding the association between plant-derived compounds and GDM are scarce. This review discusses the current knowledge and issue about the impact of dietary polyphenols on the mechanisms and/or factors regulating glucose homoeostasis, inflammation and adipose tissue (AT) function in metabolic alterations associated with GDM. The role of n-3 FA in pregnancy is also addressed. From all these data, MedDiet bioactive compounds appear to be more and more useful players to be included in future research approaches designed to prevent and treat GDM.

Dietary polyphenols

The MedDiet is characterised by a high intake of vegetable food( Reference Trichopoulou, Bamia and Trichopoulos 31 ), and polyphenols are the biggest class of plant-derived bioactive compounds( Reference Bravo 32 Reference Neveu, Perez-Jimenez and Vos 34 ). These phytochemicals are found in quite variable quantities in fruit, vegetables, cereals, nuts, tea, wine, chocolate, olives, extra-virgin olive oil and plant-derived foodstuffs, as well as spices and algae( Reference Neveu, Perez-Jimenez and Vos 34 Reference Perez-Jimenez, Fezeu and Touvier 36 ). Even more data link polyphenol intake with both health promotion and the prevention of non-communicable diseases such as CVD, stroke, T2D and some cancers( Reference Xiao and Hogger 27 Reference Oh and Jun 29 , Reference Tsao 37 ). Polyphenols are a complex class of compounds having a phenolic ring in their structure; they can be classified on the basis of the numbers of phenol rings they contain and the structural elements that bind these rings( Reference Bravo 32 , Reference Manach, Scalbert and Morand 35 ). The majority of polyphenols exist as glycosides, esters, polymers and in hydroxylated form( Reference Manach, Scalbert and Morand 35 , Reference Tsao 37 ). They are extensively modified throughout stomach, small intestine, colon and liver( Reference Landete 38 ); thus, a single polyphenol can generate several active metabolites( Reference Del Rio, Costa and Lean 39 ). Because of their chemical structures, dietary polyphenols exert multiple activities by interacting with several molecular pathways and cellular components including microRNA (miRNA)( Reference Bahadoran, Mirmiran and Azizi 28 , Reference Tsao 37 , Reference Lancon, Michaille and Latruffe 40 , Reference Santangelo, Vari and Scazzocchio 41 ). The main classes of polyphenols are flavonoids, phenolic acids, stilbenes and lignans( Reference Bravo 32 , Reference Manach, Scalbert and Morand 35 ). Flavonoids are the most abundant class of polyphenols in our diet and include different subclasses, that is, flavonols, flavones, flavanols, flavanones, anthocyanidins and isoflavones( Reference Sebastian, Wilkinson Enns and Goldman 42 , Reference Zamora-Ros, Forouhi and Sharp 43 ) (Fig. 1). The evaluation of the actual contribution of dietary polyphenols to human health is challenging, as several factors come into play: food-related factors (i.e. amount consumed, food content and bioavailability) and host-related factors (i.e. genetics, obesity, pregnancy and gut microbiota)( Reference D’Archivio, Filesi and Vari 44 ). Moreover, chemical structure, synergistic effects among different polyphenols and interaction with cellular components (e.g. membrane, proteins, enzymes, receptors and transcription factors) render the evaluation quite hard( Reference D’Archivio, Filesi and Vari 44 , Reference Fraga, Galleano and Verstraeten 45 ). An accurate measurement of the exposure to specific compounds will make it possible to associate these factors with health status and disease outcomes. To this end, efforts have been made to assess the following: (i) the level of adherence to the MedDiet, by the Mediterranean Dietary Serving Score, which accounts for the consumption of foods and food groups per meal, day or week( Reference Monteagudo, Mariscal-Arcas and Rivas 46 ); (ii) the content of polyphenols in food and their metabolism( Reference Neveu, Perez-Jimenez and Vos 34 ), as well as the human intake of total and specific polyphenols, by the creation of online database (e.g. Phenol-Explorer and United States Department of Agriculture database)( Reference Neveu, Perez-Jimenez and Vos 34 , Reference Sebastian, Wilkinson Enns and Goldman 42 , Reference Zamora-Ros, Forouhi and Sharp 43 , Reference Gonzalez, Fernandez and Cuervo 47 49 ); and (iii) novel biomarkers for polyphenol ingestion in biological fluids, by measuring food metabolome( Reference Wang, Tang and Wang 50 Reference Scalbert, Brennan and Manach 52 ).

Fig. 1 Chemical structure and main dietary source of polyphenols discussed in this review regarding their capability to modulate glucose metabolism signalling.

Glucose homoeostasis regulation

GDM is defined as carbohydrate intolerance during pregnancy( Reference Chen, Wang and Ji 1 ). Pregnancy is characterised by a complex process of endocrine-metabolic changes, including physiological insulin resistance (IR), necessary to ensure the supply of nutrients to the fetus and to adequately prepare the maternal organism for childbirth and lactation( Reference Sonagra, Biradar and K 53 ). GDM develops when the pregnant woman is not able to produce an adequate insulin response to compensate for physiological IR( Reference Catalano, Huston and Amini 54 ). IR during pregnancy is uniquely associated with a decrease in insulin receptor substrate 1 (IRS-1) tyrosine phosphorylation, primarily because of a decreased expression of IRS-1 protein( Reference Harlev and Wiznitzer 55 ). Nevertheless, in GDM subjects, there is an additional decrease in tyrosine phosphorylation of the insulin receptor β subunit, associated with further decreases in glucose transport activity( Reference Harlev and Wiznitzer 55 , Reference Catalano 56 ). Lower levels of tyrosine kinase receptor activity (30–40 %), IRS-1 expression and activation, and GLUT4, in fact, have been detected in pregnancy with respect to pre-pregnancy status( Reference Harlev and Wiznitzer 55 , Reference Catalano 56 ). Specifically, IRS-1 tyrosine phosphorylation has been found to be significantly reduced in muscle from pregnant control and obese GDM subjects, as reflected by a 23 and 44 % reduction, respectively, compared with non-pregnant women( Reference Friedman, Ishizuka and Shao 57 ). AT, through a large arrays of secreted factors, participates in the induction and regulation of IR both in non-pregnant and pregnant subjects( Reference Tersigni, Di Nicuolo and D’Ippolito 58 ). Maternal pre-pregnancy obesity decreases insulin sensitivity and positively associates with the risk of GDM( Reference Li, Liu and Guo 59 ). A recent study has shown that AT inflammasome (caspase-1 and IL-1β) is involved in the development of IR in GDM-complicated pregnancies( Reference Lappas 60 ). Increased IL-1β expression has been observed in AT from GDM women; incubation of AT with IL-1β results in a significant attenuation of phosphorylated IRS-1 protein expression, GLUT4 mRNA and protein expression and glucose uptake, indicating the importance of inflammasome in the pathophysiology of GDM( Reference Lappas 60 ).

Over the past years, the role of miRNA in gene regulation has been gaining attention; miRNA are implicated in regulating cholesterol biosynthesis, carbohydrate and lipid metabolism( Reference Krutzfeldt and Stoffel 61 ), as well as in insulin production, secretion and action( Reference Mao, Mohan and Zhang 62 ) (i.e. in glucose homoeostasis)( Reference Mao, Mohan and Zhang 62 Reference Chen, Lan and Roukos 65 ), by interacting with specific mRNA targets( Reference Dweep and Gretz 66 ). Accordingly, miRNA have been explored in GDM with the aim of using them as early biomarkers of disease( Reference Collares, Evangelista and Xavier 67 Reference Zhu, Tian and Li 70 ), as they are present in the blood or other biological fluids( Reference Collares, Evangelista and Xavier 67 , Reference Etheridge, Lee and Hood 71 ). To date, few investigations have been performed; two studies conducted in China have found decreased levels of miR-29a, -132 -and -222( Reference Zhao, Dong and Jiang 69 ), and increased levels of miR-16-5p, miR-17-5p, miR-19a-3p, miR-19b-3p and miR-20a-5p, in gestational weeks 16–19 in the serum of pregnant women who later developed GDM in gestational weeks 25–28, with respect to women who did not develop GDM( Reference Zhu, Tian and Li 70 ). However, hyperglycemic non-pregnant rodents have shown opposite expression of mir-29 and mir-132 families. MiR-29 family is involved in the insulin-signalling pathway; muscle, fat and liver from hyperglycaemic Goto–Kakizaki rats exhibit up-regulation of miR-29a and miR-29b( Reference He, Zhu and Gupta 72 ). High glucose induces the up-regulation of miR29a associated( Reference He, Zhu and Gupta 72 ) with the decrease in IRS-1, downstream kinase Akt and glycogen synthase kinase 3β, (GSK3β) proteins, in both rat myocytes( Reference Yang, Jeong and Park 73 ) and human pancreatic β-cells( Reference Bagge, Clausen and Larsen 74 ). MiR-132 expression is up-regulated in pancreatic islets of pre-diabetic (6 weeks old) and diabetic db/db mice (14–20 weeks)( Reference Nesca, Guay and Jacovetti 75 ); this increase improved glucose-stimulated insulin release and increased cell proliferation, which suggests that the modification of miR-132 levels might contribute to compensatory β-cell mass expansion elicited in response to IR( Reference Nesca, Guay and Jacovetti 75 ). In GDM women, the up-regulation of miR-222 has been found to be associated with reduced protein levels of both oestrogen receptor (ER)-α and GLUT4 in omental AT, obtained at the time of caesarean delivery, and with increased serum estradiol levels( Reference Shi, Zhao and Guo 76 ). In consideration of these findings, we may assume that miRNA are expressed in a species-, tissue-specific and time-dependent manner.

Furthermore, miRNA profiles of peripheral blood mononuclear cells isolated from Brazilian GDM women, obtained by using microarray platforms, have identified ten miRNA that seemed to be specific for GDM, namely, miR-101, miR-1180, miR-1268, miR-181a, miR-181d, miR-26a, miR-29a, miR-29c, miR-30b and miR-595( Reference Collares, Evangelista and Xavier 67 ).The target genes of most of these miRNA are involved in insulin signalling, angiogenesis, IR and AT dysfunction( Reference Dweep and Gretz 66 ). It is noteworthy that miR-181a is increased in the serum of diabetic patients, as well as in IR cultured hepatocytes and liver( Reference Zhou, Li and Qi 77 ). Inhibition of miR-181a by antisense oligonucleotides restores insulin sensitivity in hepatocytes, thus providing evidence of a potential therapeutic strategy for treating IR and T2D( Reference Zhou, Li and Qi 77 ). On the other hand, miR-30 belonging to the miR-30 family that has a key role in angiogenesis( Reference Arola-Arnal and Blade 78 ) might be involved in the hyper capillarisation of the placenta in women with mild hyperglycaemia( Reference Bridge, Monteiro and Henderson 79 , Reference Pietro, Daher and Rudge 80 ). Furthermore, miR-30 family members increase in abundance during the differentiation of pancreatic islet-derived mesenchymal cells into hormone-producing islet-like cell aggregates, thus indicating their participation in the regulatory signalling of the embryonic development of human pancreatic islets( Reference Joglekar, Patil and Joglekar 81 ). However, the miRNA modifications so far observed in GDM women result from a limited number of screened subjects (from six to twenty). In addition, people from different countries appear to have different miRNA profiles, except for miR-29, in the same metabolic condition. In conclusion, although the above data provide evidence that miRNA are promising, non-invasive biomarkers of GDM, further research with larger cohorts is warranted.

Polyphenols effects

The maintenance of glucose homoeostasis is extremely important for human physiology( Reference Zhang, Tobias and Chavarro 82 ). Growing evidence indicates that polyphenols, contained in fruits and vegetables, might influence glucose homoeostasis by several mechanisms such as by (i) inhibiting carbohydrate digestion and glucose absorption in the intestine, (ii) stimulating insulin secretion from pancreatic β-cells, (iii) modulating glucose release from liver and (iv) activating insulin receptors and glucose uptake in insulin-sensitive tissues( Reference Hanhineva, Torronen and Bondia-Pons 83 ). The bioactive food components can modify gene expression and regulate different signalling pathways, thus affecting muscle, liver, pancreatic β-cells, hypothalamus and AT functions, thereby regulating glucose homoeostasis( Reference Berna, Oliveras-Lopez and Jurado-Ruiz 84 , Reference Babu, Liu and Gilbert 85 ). This regulatory activity occurs also by modulating miRNA gene expression( Reference Lancon, Michaille and Latruffe 40 , Reference Garcia-Segura, Perez-Andrade and Miranda-Rios 86 Reference Jimenez, Dorado and Hernandez-Perez 89 ) (Fig. 2). Isoflavones, anthocyanins, flavanols and catechins appear to enhance β-cell function and glucose tolerance in animal models and humans and to protect against diabetes( Reference Oh and Jun 29 ). Genistein and daidzein are the major dietary isoflavones present primarily in soya foods( Reference Crozier, Jaganath and Clifford 90 ). Long-term genistein exposure (1–10 µm for 48 h) improved glucose-stimulated insulin secretion (GSIS) in mouse and human pancreatic islets( Reference Fu and Liu 91 ); this effect appears to be mediated by cyclic AMP/protein kinase A signalling activation and increased intracellular Ca2+ levels( Reference Fu and Liu 91 ) (Table 1).

Fig. 2 Crosstalk among signalling pathways in regulating glucose metabolism. All of the factors that appear in this scheme are potential points of action of polyphenols. , Activation; , inhibition; , modulation; JAK/STAT, Janus kinase/signal transducer and activator of transcription; AMPK, AMP-activated protein kinase; JNK, c-Jun N-terminal kinase; IRS1/2, insulin receptor substrate 1/2; MAPK, mitogen-activated protein kinases; PDK1, 3-phosphoinositide-dependent protein kinase-1; PI3K, phosphatidylinositol-3-kinase; Akt, protein kinase B; SIRT-1, sirtuin 1; NO, nitric oxide; eNOS, endothelial NO synthase; FOXO1, forkhead box protein O1; TF, transcription factors; miRNA, microRNA; FA, fatty acids.

Table 1 Effects of dietary polyphenols on molecular mechanisms associated with gestational diabetes mellitus

↑, Increases; ↓, decreases; NOD, non-obese diabetic; JNK, c-Jun N-terminal kinase; Foxo1, forkhead box protein O1; GSIS, glucose-stimulated insulin secretion; cAMP, cyclic AMP; PKA, protein kinase A; EGCG, epigallocatechin-3-gallate; IRS1/2, insulin receptor substrate 1/2; AMPK, AMP-activated protein kinase; C3G, cyanidin-3-glucoside; Sirt-1, sirtuin 1; STZ, streptozotocin; PDX1, pancreatic and duodenal homeobox-1; PCA, protocatechuic acid; T2D, type 2 diabetes; Akt, protein kinase B; INS-1, insulin 1; GK, glycerol kinase; p38 MAPK, p38 mitogen-activated protein kinase; HbA1c, glycated Hb; PI3K, phosphatidylinositol-3-kinase; ERK1/2, extracellular signal-regulated kinase; LPS, lipopolysaccharides; NO, nitric oxide; eNOS, endothelial NO synthase; BAEC, bovine artery endothelial cells; Tyr, tyrosol; OL, oleuropein; HT, hydroxytyrosol; Tax, taxifolin; VEGFR2, vascular endothelial growth factor receptor 2; VEGF, vascular endothelial growth factor; HUVEC, human umbilical vein endothelial cells.

The flavanol epigallocatechin gallate (EGCG), a main component of green tea extract, preserves the insulin secretory machinery; EGCG (10 µm for 48 h) stimulates the activation of the IRS-2 signalling in rat insulinoma pancreatic β-cells (RIN-m5F), also under chronic hyperglycaemia( Reference Cai and Lin 92 ). EGCG increases the activity of AMP-activated protein kinase (AMPK) through Thr172 phosphorylation, further strengthening the hypothesis that AMPK is involved in counteracting the glucolipotoxicity induced by high-glucose condition( Reference Cai and Lin 92 ). AMPK is an energy-sensing enzyme recognised as a master regulator of whole-body energy homoeostasis; its activation increases GLUT4 expression and membrane translocation in skeletal muscle, thereby improving glucose uptake( Reference Yu, Qiu and Nan 93 ). In this regard, AMPK has been considered as one of the targets of the insulin-sensitising drugs metformin and thiazolidinediones( Reference Yu, Qiu and Nan 93 ).

EGCG (20, 40 μm for 24 h) improved the insulin-stimulated glucose uptake also in dexamethasone-treated skeletal muscle cells L6, associated with an increased AMPK phosphorylation and GLUT4 membrane translocation( Reference Zhang, Li and Liang 94 ). Furthermore, the dexamethasone-stimulated inactivation of IRS-1 because of increased Ser307 phosphorylation has been significantly reversed by EGCG treatment( Reference Zhang, Li and Liang 94 ). Results from the miRNA profiling suggest that EGCG may exert its biological functions through up- or down-regulating multiple miRNA as well( Reference Tsang and Kwok 95 ). In particular, EGCG (50 mg/l for 5 h), grape seed (100 mg/l for 5 h) and cocoa proanthocyanidin extracts (100 mg/l for 5 h) down-regulated miR-30b expression in human hepatocellular carcinoma HepG2 cells( Reference Arola-Arnal and Blade 78 ). A longer EGCG (100 μm for 24 h) treatment reduced miR-181d and miR-222 expression as well, in HepG2 cells( Reference Tsang and Kwok 95 ).

Anthocyanins are widely distributed in the human diet through berries, fruits, vegetables and red wine( Reference Takikawa, Inoue and Horio 96 ). Pretreatment with bayberry fruit extracts (0·5 μmol/l cyanidin-3-glucoside (C3G) for 12 h) up-regulated pancreatic duodenal homeobox-1 gene expression, which is associated with increased levels of insulin-like growth factor-II gene transcript and insulin, in rat insulinoma cell line INS-1β-cells( Reference Sun, Zhang and Zhang 97 ).

We have shown that C3G (10·50 µm for 18 h) and its metabolite protocatechuic acid (PCA; 100 µm for 18 h) increased glucose uptake and GLUT4 membrane translocation in murine adipocytes and visceral human adipocytes( Reference Scazzocchio, Vari and Filesi 98 Reference Scazzocchio, Vari and Filesi 100 ). Specifically, PCA appeared to exert insulin-like effects by stimulating IRS-1 tyrosine phosphorylation and Akt activation( Reference Scazzocchio, Vari and Filesi 98 Reference Scazzocchio, Vari and Filesi 100 ).

Caffeic acid, naringenin and quercetin (10−6 µm for 72 h each), present in many plants, enhance GSIS and glucose sensitivity in rat pancreatic INS-1E cells( Reference Bhattacharya, Oksbjerg and Young 101 ). These compounds differently modulate gene expression profiles to improve β-cell survival and function during glucotoxicity (e.g. naringenin and quercetin up-regulated GLUT2, glucokinase, AKT1, whereas caffeic acid increased AKT2 and all the three compounds increased INS-1, mRNA expression)( Reference Bhattacharya, Oksbjerg and Young 101 ). Similarly, curcumin (5–15 µm for 24 h), a yellow pigment isolated from Curcuma Longa, increases insulin gene expression and GSIS in a dose-dependent manner in INS-1 cells( Reference Song, Wang and Zhu 102 ). In doing so, curcumin up-regulates the expression of GLUT2, the phosphorylation of IRS-1, phosphatidylinositol-3-kinase (PI3K) and Akt, suggesting that curcumin prevents the high-glucose-induced reduction of insulin expression and secretion by activating the PI3K/Akt/GLUT2 pathway in INS-1 cells( Reference Song, Wang and Zhu 102 ). Curcumin treatment modulates the expression profile of several miRNA, including up-regulation of miRNA-26a in A549 human lung adenocarcinoma cell line (curcumin: 15 µm for 48 h)( Reference Zhang, Du and Wu 103 ) and miR-181a in BxPC-3 human pancreatic carcinoma cell line (curcumin: 10 µm for 72 h)( Reference Sun, Estrov and Ji 104 ).

Consistent with in vitro studies, animal intervention studies showed that supplementation with genistein (0·2 g/kg diet) or daidzein (0·2 g/kg diet) for 9 weeks preserved β-cells and reduced blood glucose levels by inducing insulin secretion, in non-obese diabetic mice( Reference Choi, Jung and Yeo 105 ). Similarly, administration of 0·1 ml of bayberry fruit extracts (containing 150 μg of C3G)/10 g of body weight twice per day for 30 d significantly reduced blood glucose and improved glucose tolerance in streptozotocin-induced diabetic mice( Reference Sun, Zhang and Zhang 97 ).

Changes in the expression of a large number of different miRNA have been observed after each polyphenol supplementation( Reference Milenkovic, Jude and Morand 88 ). MiRNA profile expression was evaluated in the livers of wild-type (C57B6/J) mice or apoE–/– mice fed diets supplemented with different polyphenols (including quercetin, hesperidin, naringenin, proanthocyanidin, caffeic acid, curcumin)( Reference Milenkovic, Jude and Morand 88 , Reference Milenkovic, Deval and Gouranton 106 ). MiR-29a and miR-30b expression were down-regulated by almost all the tested polyphenols; miR-222 was down-regulated by caffeic acid (300 mg/d for 2 weeks) and hesperidin (30 mg/d for 2 weeks); miR-181a was down-regulated by curcumin (30 mg/d for 2 weeks) and hesperidin and up-regulated by naringin (30 mg/d for 2 weeks), quercetin (30 mg/d for 2 weeks) and proanthocyanidin (300 mg/d for 2 weeks); miR-132 was up-regulated by naringin( Reference Milenkovic, Jude and Morand 88 ). This study highlights the importance of miRNA as modulators of dietary polyphenols activities, in vivo ( Reference Milenkovic, Jude and Morand 88 ).

Human intervention trials and cohort studies have reported that dietary supplementation with 22·5 g of whole blueberry twice daily (1462 mg of total phenolics, 668 mg of anthocyanins) for 6 weeks resulted in blood glucose concentration reduction and in insulin sensitivity improvement in obese, non-diabetic, IR subjects( Reference Stull, Cash and Johnson 107 ). Data from three prospective cohort studies involving 200 000 US men and women showed that a higher consumption of anthocyanin-rich fruit is associated with a lower risk of T2D( Reference Wedick, Pan and Cassidy 108 ). Collectively, anthocyanins and its glycosides, alone or in combination, improve glucose homoeostasis by influencing β-cell mass and function, insulin sensitivity, glucose uptake and insulin signalling( Reference Babu, Liu and Gilbert 85 ).

There is no doubt that dietary polyphenols have beneficial effects on β-cell function and glucose homoeostasis taking into account that most of them are capable of modulating almost all the players, including several miRNA, in the glucose uptake machinery. Although promising data have been obtained so far, no information on the effects of these bioactive compounds in GDM setting are available as yet.

Adipokines and gestational diabetes

It is well known that adipocyte-derived cytokines (adipokines) are involved in the modulation of a wide range of physiological processes including lipid metabolism, atherosclerosis, angiogenesis and insulin sensitivity( Reference Lappas 109 , Reference D’Ippolito, Tersigni and Scambia 110 ). The human gestational tissues, as well as AT, produce and release a large array of pro-inflammatory cytokines; the activation of some inflammatory pathways is necessary to induce maternal IR, which is required for the progression of normal gestation( Reference Santangelo, Vari and Scazzocchio 9 , Reference Hauguel-de Mouzon and Guerre-Millo 111 , Reference Friis, Paasche Roland and Godang 112 ). GDM associates with the amplification of the low-grade inflammation already existing in normal pregnancy( Reference Barbour, McCurdy and Hernandez 113 , Reference Li, Chen and Li 114 ). Indeed, serum TNFα, IL-1β and IL-6 levels are higher in GDM women as compared with control pregnant women( Reference Xu, Jin and Sun 115 ). Notably, pre-pregnant obesity in GMD women worsens this inflammatory picture by up-regulating TNFα, IL-1β and/or leptin, and by down-regulating genes involved in AT lipid metabolism such as PPARα, PPARδ, PPARγ, retinoid X receptor-α (RXRα) and sterol regulatory element-binding protein-1c (SREBP1c), in AT( Reference Lappas 109 ).

Over the past years, various adipokines such as adiponectin and leptin have been shown to play a role in normal pregnancy, as well as in complications of pregnancy including GDM( Reference Santangelo, Vari and Scazzocchio 9 , Reference D’Ippolito, Tersigni and Scambia 110 , Reference Miehle, Stepan and Fasshauer 116 , Reference Retnakaran, Hanley and Raif 117 ). Low adiponectin serum levels appear to be linked with T2D and IR( Reference Rasouli and Kern 118 ). A study conducted on 445 pregnant women demonstrated that lower levels of adiponectin during the first trimester of pregnancy are associated with increased risk of developing GDM during the second trimester, independently of other early pregnancy major risk factors( Reference Lacroix, Battista and Doyon 119 ). Further, a nested case–control study showed that lower adiponectin levels measured 6 years before pregnancy were associated with a 5-fold increased risk of developing GDM( Reference Hedderson, Darbinian and Havel 120 ). Thus, an early low adiponectin expression, which most likely reflects pre-existing IR, could be considered a useful biomarker to identify women at high risk for GDM and allow for early therapeutic interventions( Reference Hedderson, Darbinian and Havel 120 ). Adiponectin acts as an insulin-sensitising agent, and its action is complex and incompletely defined, but the collected evidence suggests that, by binding with its receptors, adiponectin activates three key signalling pathways in muscle and liver: AMPK, p38 mitogen-activated protein kinase (p38 MAPK) and PPAR( Reference Yamauchi, Kamon and Minokoshi 121 ). The activation of these pathways results in FA oxidation (FAO) and glucose uptake in skeletal muscle, and in the inhibition of gluconeogenesis in the liver( Reference Yamauchi, Kamon and Minokoshi 121 ). Reduced adiponectin could be held accountable for the lower FAO observed in placentas of GDM women compared with the control group( Reference Visiedo, Bugatto and Sanchez 122 ). Adiponectin gene expression, processing and secretion is up-regulated by PPARγ agonist; PPARγ activation results in increased insulin sensitivity, in skeletal muscle and liver, by inducing the release of insulin-sensitising adipokines including adiponectin( Reference Astapova and Leff 123 ). Administration of adiponectin to diabetic mice( Reference Berg and Scherer 124 ) and obese pregnant mice enhances insulin activity( Reference Aye, Rosario and Powell 125 ), prevents fetal overgrowth( Reference Aye, Rosario and Powell 125 ), normalises placental insulin/mammalian target of rapamycin complex 1 (mTORC1), PPARα signalling and placental nutrient transport( Reference Aye, Rosario and Powell 125 ). Adiponectin also mediates the crosstalk between AT and the vessel wall; thus, in virtue of its pleiotropic activities on multiple targets, it seems to have a protective role against vascular dysfunction induced by obesity and diabetes( Reference Li, Cheng and Lam 126 ).

TNFα and leptin have been suggested as the strongest predictors of pregnancy-related IR( Reference Lepercq, Cauzac and Lahlou 127 , Reference Lappas 128 ). Hyperleptinaemia in early pregnancy (13 weeks) appears to be a good predictor of GDM risk, independent of maternal adiposity( Reference Qiu, Williams and Vadachkoria 129 , Reference Al-Badri, Zantout and Azar 130 ). In week 30 of pregnancy, leptin levels have been found to be similar in both GDM and no-GDM subjects, but they remained elevated after delivery in the GDM group( Reference Saucedo, Zarate and Basurto 131 ).

Recent systematic reviews have highlighted higher and lower levels of plasma leptin and adiponectin, respectively, in the first or second trimester of pregnancy( Reference Bao, Baecker and Song 132 ), and this condition persisted, associated also with high TNF levels, in the late second or third trimester of pregnancy( Reference Xu, Zhao and Chen 133 ), in GDM patients compared with normal pregnancies. These findings corroborate the role exerted by the altered balance pro-inflammatory/anti-inflammatory factors in the impaired glucose homoeostasis in GDM.

Increased levels of leptin have been found in visceral AT of obese and GDM women( Reference Lappas 109 ). Leptin, predominantly produced by adipocytes, is considered an essential factor in maintaining energy homoeostasis, as it regulates food intake and energy expenditure via specific receptors in the hypothalamus( Reference Hegyi, Fulop and Kovacs 134 ). Leptin acts by activating Janus kinase/signal transducer and activator of transcription signalling, although other important intracellular signalling including PI3K, AMPK and MAPK appear to be involved as well( Reference Hegyi, Fulop and Kovacs 134 ).

In GDM women, TNFα and leptin levels are elevated; the increased TNFα expression causes a chronic inflammatory environment with enhanced leptin production, which, in turn, increases TNFα and IL-6 production by monocytes( Reference Santos-Alvarez, Goberna and Sanchez-Margalet 135 ), generating a vicious circle that amplifies the inflammatory situation and worsens the metabolic dysfunction in GDM. Although the significance of leptin and adiponectin is relatively well characterised as for their effects on glucose and lipid metabolism in diabetes, the molecular mechanisms by which these adipokines exert their effects on insulin action are not completely defined. Much more research is needed especially in the light of the growing list of molecules identified as adipokines, for example, visfatin, apelin, retinol-binding protein 4, vaspin, omentin and adiposity FA-binding protein 4 (FABP4), which renders the comprehension of the existing network and the interactions among them more complicated ( Reference Santangelo, Vari and Scazzocchio 9 , Reference Wojcik, Chmielewska-Kassassir and Grzywnowicz 136 , Reference Li, Xiao and Li 137 ).

Polyphenols effects

The beneficial effects of MedDiet dietary compounds on diabetes are associated with reduced biomarkers of inflammation( Reference Landberg, Naidoo and van Dam 138 Reference Schwingshackl and Hoffmann 140 ).

Taking into account the importance of the insulin-sensitising effects of adiponectin, enhancing adiponectin/AdipoR function may be an interesting therapeutic strategy against IR( Reference Caselli 141 ). Dietary compounds such as fish oil, linoleic acid (LA), seed extract, green tea extract and resveratrol (RSV) elevated adiponectin concentration; specifically, RSV (50 µm for 24 h) treatment stimulated adiponectin expression and reduced mRNA levels of leptin in 3T3-L1 adipocytes( Reference Wang, Liu and Liu 142 , Reference Eseberri, Lasa and Churruca 143 ). The stimulatory effect of RSV has been shown to be mediated via histone deacetylase sirtuin 1 (Sirt-1)-independent mechanism and is mainly because of phosphoinositide-dependent protein kinase-1/Akt signalling pathway suppression, in the activation of the transcription factor forkhead box O-1 (FOXO1), and the AMPK signalling pathway( Reference Wang, Liu and Liu 142 ). RSV (0·1–10 μm) reduced the expression and release of IL-6 and TNFα in RAW 264.7 macrophages and 3T3-L1 cells, in association with a reduced extracellular signal-regulated protein kinases 1/2 (ERK1/2) and NF-κB activation( Reference Kang, Heng and Yuan 144 ). The high-glucose-induced nitric oxide (NO) reduction was reverted by RSV (0·1–10 μm for 24 h) incubation in bovine artery endothelial cells, because of enhanced endothelial NO synthase mRNA and activity( Reference Peng, Qu and Wang 145 ). Altogether, these findings strongly indicate that RSV could be a therapeutic supplement to counteract the diabetic condition.

C3G treatment (12·5, 25, 50 µm for 24 h) induced adiponectin expression in 3T3-L1 and human adipocytes( Reference Scazzocchio, Vari and Filesi 98 , Reference Liu, Li and Zhang 146 ). As regards the molecular mechanisms, C3G induced FOXO1 deacetylation and transcriptional activity, as well as increased PPARγ activity( Reference Scazzocchio, Vari and Filesi 98 ), which, in turn, brought about adiponectin expression and secretion, in adipocytes( Reference Liu, Li and Zhang 146 ). In addition, flavonoid fisetin (0·1–10 μm for 24 h) increased adiponectin gene transcription by inducing SIRT-1-deacetylase activity and PPAR activation, in mouse adipocytes( Reference Jin, Kim and Shin 147 , Reference Khan, Syed and Ahmad 148 ).

Virgin olive oil components such as hydroxytyrosol (HT; 0·1–20 μm for 1 h pretreatment) and oleic acid (100 μm for 48 h pretreatment) alone or in combination in human adipocytes can prevent TNFα-induced down-regulation of adiponectin by attenuating PPARγ suppression mediated by c-Jun N-terminal kinase (JNK)( Reference Scoditti, Massaro and Carluccio 149 ). Moreover, HT, oleuropein, taxifolin and tyrosol (50 µm pretreatment for 24 h each) showed anti-angiogenesis activities as well( Reference Lamy, Ouanouki and Beliveau 150 ). Indeed, all these compounds suppressed angiogenesis in human umbilical vein endothelial cells by inhibiting vascular endothelial growth factor receptor 2 (VEGFR-2) signalling pathway. Specifically, HT and taxifolin inhibited VEGF-dependent tyrosine phosphorylation of VEGFR-2 by impairing phosphorylation of p42/p44 MAPK (ERK1/2) and p46/p54 stress-activated protein kinase (SAPK)/JNK( Reference Lamy, Ouanouki and Beliveau 150 ). Altogether, these data provide further molecular evidence of the beneficial effects of olive oil consumption in the MedDiet in the treatment of metabolic diseases.

The soya isoflavones and their aglycones daidzein and genistein, as well as the equol metabolite (40 µm for 2 h for each), inhibited leptin secretion in mouse adipocytes( Reference Niwa, Yokoyama and Ito 151 ). A mechanistic study carried out in 3T3-L1 adipocytes demonstrated that genistein and daidzein inhibit TNFα-mediated down-regulation of adiponectin through different mechanisms; genistein inhibited TNFα-induced JNK signalling and daidzein inhibited TNFα-induced down-regulation of FOXO1( Reference Yanagisawa, Sugiya and Iijima 152 ). Naringin (80 µm pretreatment for 2 h) incubation down-regulated the high-glucose-induced leptin expression and inhibited leptin-induced activation of the p38 MAPK pathway in H9c2 cardiac cells( Reference Chen, Mo and Guo 153 ). An in vivo study showed that oral administration of naringin (50 mg/kg for 4 weeks) or hesperidin (50 mg/kg for 4 weeks) in diabetic rats significantly reduced the percentage of blood glycated Hb, and serum levels of TNFα and IL-6( Reference Mahmoud, Ashour and Abdel-Moneim 154 ). In humans, purified anthocyanin (320 mg/d for 12 weeks) supplementation significantly increased serum adiponectin concentrations in patients with T2D( Reference Liu, Li and Zhang 146 ).

The metabolites of polyphenols can be useful biomarkers of dietary intake to evaluate individual response to specific compounds; for example, equol is not produced in all adults who consume soya foods( Reference Sakane, Kotani and Tsuzaki 155 ). A recent study has shown that, in pre-diabetic and diabetic female population, the ‘equol producers’ subjects exhibited lower levels of leptin as compared with non-producers, indicating an association between isoflavone intake and leptin levels( Reference Sakane, Kotani and Tsuzaki 155 ). In healthy obese postmenopausal women, physical activity, diet and daily oral intake of a soya isoflavone extract (60·8 mg of genistein, 16 mg of daidzein and 3·2 mg of glycitein) resulted in reduced levels of serum leptin and TNFα associated with a significant increase in adiponectin, after 6 months of treatment( Reference Llaneza, Gonzalez and Fernandez-Inarrea 156 ).

A recent and exhaustive review describes the inhibitory effects of certain polyphenols on TNFα-activated inflammatory pathways both in vitro and in vivo, and in human studies( Reference Gupta, Tyagi and Deshmukh-Taskar 157 ).

The health effects of PUFA

A substantial amount of FA is required as additional source of energy to support fetal cellular growth, and gestational hyperlipidaemia normally occurring during late pregnancy enhances placental access to FA( Reference Rebholz, Burke and Yang 158 , Reference Jones, Mark and Waddell 159 ). GDM associates with an altered maternal lipid profile and affects the quantity and/or quality of lipids transferred to the fetus( Reference Herrera and Ortega-Senovilla 160 ). In GDM, a positive correlation between maternal TAG and NEFA levels, and fetal growth and fat mass, has been found even in diabetic mothers with appropriate glycaemic control( Reference Herrera and Ortega-Senovilla 160 ). Fetus development depends on the maternal supply of essential PUFA such as LA (18 : 2 n-6), α-linolenic acid (ALA, 18 : 3 n-3) and long-chain (LC)-PUFA, EPA (20 : 5 n-3), DHA (22 : 6 n-3) and arachidonic acid (AA, 20 : 4 n-6)( Reference Haggarty 161 ). The placenta regulates adequate LC-PUFA delivery to the fetus in a directional, preferential and timely manner( Reference Haggarty 161 ), preferentially transferring n-3 LC-PUFA( Reference Jones, Mark and Waddell 159 ). In GDM women, the placental transfer of LC-PUFA appears to be altered, with an impaired maternal-fetal transfer of DHA, which might affect neurodevelopment programming in the offspring( Reference Gil-Sanchez, Demmelmair and Parrilla 162 ). A reduced concentration of LC-PUFA in maternal, placental and fetal compartments in GDM women compared with healthy pregnant women has been highlighted( Reference Pagan, Prieto-Sanchez and Blanco-Carnero 163 , Reference Araujo, Correia-Branco and Ramalho 164 ).

However, increased, unchanged or lower hyperlipidaemia in diabetic pregnant women v. non-diabetic pregnant women has been observed too( Reference Herrera and Ortega-Senovilla 160 ). In this regard, it has been hypothesised that the degree of metabolic control and sex hormonal dysfunction may influence the different degree of dyslipidaemia observed in diabetic pregnant women( Reference Herrera and Ortega-Senovilla 160 ). NEFA are taken up by the placenta and FA are released from maternal lipoproteins by endothelial lipase (EL) and lipoprotein lipase (LPL), which are present in the maternal-facing microvillus membrane of the syncytiotrophoblast( Reference Gil-Sanchez, Demmelmair and Parrilla 162 ). Increased EL mRNA has been found in placenta from obese women with GDM compared with lean GDM women or normoglycemic pregnant women( Reference Gauster, Hiden and van Poppel 165 ). In contrast, a recent study has shown that no difference in the expression of LPL and EL exists between GDM and normoglycaemic placentas, suggesting that these lipases are not involved in the increased newborn adiposity( Reference Barrett, Kubala and Scholz Romero 166 ). NEFA may enter the cell by passive diffusion or by several plasma membrane-located transport/binding proteins including FA translocase (FAT/CD36), plasma membrane FABP(pm), FA transport proteins (FATP1–6) and intracellular FABP( Reference Duttaroy 167 ). DHA and EPA are endogenous ligands of several transcription factors such as PPAR, RXR and SREBP1( Reference Jawerbaum and Capobianco 168 ). A decreased expression of genes involved in FA uptake, intracellular transport, storage and synthesis (e.g. LPL, FATP2, FATP6, FABPpm, acyl-CoA synthetase long-chain family member 1), and of transcription factors involved in lipid metabolism regulation (e.g. liver X receptor (LXRα), PPARα, PPARδ, PPARγ, RXRα, SREBP1c), has been observed in AT obtained from obese pregnant women and women with GDM( Reference Lappas 109 ). These results indicate that pre-existing maternal obesity and GDM are associated with abnormal AT lipid metabolism, which may play a role in the pathogenesis of both diseases( Reference Lappas 109 ).

Dietary effects

PUFA are fundamental for human beings, and particularly important is the balance of n-6:n-3 FA in maintaining homoeostasis, normal development and mental health throughout the lifecycle( Reference Haggarty 161 ). Different studies have led to the conclusion that humans currently need a diet containing n-3 and n-6 PUFA in a ratio of about 1:5; on the contrary, in Western diets, this ratio is 1:10–20, indicating a deficiency in n-3 FA and/or an excess of n-6 FA( Reference Simopoulos 169 ). The essential FA, LA and ALA cannot be synthesised by humans and therefore must be supplied through the diet; a series of sequential desaturation and elongation reactions acting in concert transform LA and ALA into their unsaturated derivatives, namely AA from LA, EPA and DHA from ALA( Reference Calder 170 ). EPA and DHA are abundantly present in specific fishes, for example, fresh tuna, salmon and mackerel, as well as in fish oil and nuts( Reference Russo 171 , Reference Zinati, Zamansani and Hossein KayvanJoo 172 ). Nuts, vegetables, vegetable oils, for example, soyabean, flaxseed and rapeseed oil, and cereals are also food sources of ALA and LA( 49 , Reference Calder 170 ). Walnuts are widely consumed in Mediterranean regions and are rich in ALA (2·95 g/28·4 g)( Reference Burns-Whitmore, Haddad and Sabate 173 ). In the PREDIMED (PREvención con DIeta MEDiterránea) trial that involved subjects at high risk for CVD, a diet rich in fats of vegetable origin, olive oil and mixed nuts (30 g/d, as 15 g walnuts, 7·5 g hazelnuts and 7·5 g almonds) has been shown to be beneficial in the management of the metabolic syndrome( Reference Mayneris-Perxachs, Sala-Vila and Chisaguano 174 ). The fat content of vegetables is low, but a high consumption of vegetables will result in a substantial intake of ALA. The content of ALA in some edible plants growing in the Mediterranean regions is remarkable, as in the case with purslane (300–400 mg ALA/100 g)( Reference Galli and Marangoni 175 , Reference Uddin, Juraimi and Hossain 176 ).

Different cohort studies have shown a positive association between the adherence to MedDiet style, and plasma DHA and EPA content, indicating that the MedDiet ensures higher n-3 PUFA bioavailability than Western diets( Reference Panagiotakos, Kalogeropoulos and Pitsavos 177 Reference Scoditti, Capurso and Capurso 180 ).

The European Commission and the International Society for the Study of Fatty Acids and Lipids specifically recommend that pregnant and lactating women consume a minimum of 200 mg DHA/d( Reference Koletzko, Lien and Agostoni 181 ). Several researches indicate that maternal dietary supplementation with n-3 PUFA during pregnancy can reduce the risk of pregnancy complications by limiting placental inflammation and oxidative stress( Reference Jones, Mark and Waddell 159 , Reference Pietrantoni, Del Chierico and Rigon 182 , Reference Carvajal 183 ), with positive action on insulin function, improvement of glucose tolerance and lipid profiles( Reference Mohammadi, Rafraf and Farzadi 184 , Reference Lu, Borthwick and Hassanali 185 ) (Table 2).

Table 2 Maternal n-3 fatty acids (FA) supplementation

↑, Increases; ↓, decreases.

A daily dose of 600 mg of DHA, starting from the first trimester until delivery, can ameliorate red cell membrane anomaly in pregnant women with T2D and in neonates, and it prevents the decline of maternal DHA during pregnancy( Reference Min, Djahanbakhch and Hutchinson 186 ). These results suggest that DHA supplementation should be started in the antenatal care of pregnant women with T2D. An Italian pilot study revealed that daily administration of 200 mg of DHA, from the 8th week until delivery, reduced AA levels in erythrocytes and plasma during gestational time( Reference Pietrantoni, Del Chierico and Rigon 182 ). Thus, the increased DHA levels in the maternal plasma and the consequent reduction of AA-derived eicosanoids decrease inflammation, with a beneficial effect on fetal development and growth, and IR of amniochorial membranes as well( Reference Pietrantoni, Del Chierico and Rigon 182 ). New identified PUFA-derived mediators (specialised pro-resolving lipid mediators), namely resolvins, protectins, maresins and lipoxins, are involved in inflammatory resolution( Reference Recchiuti and Serhan 187 ). It has been observed that maternal dietary supplementation with n-3 PUFA might help placenta in resolving inflammation by increasing the levels of these pro-resolving mediators, in rat( Reference Jones, Mark and Keelan 188 ) and in human placenta( Reference Keelan, Mas and D’Vaz 189 ). GDM women who received 1000 mg/d n-3 FA supplementation (containing 180 mg of EPA and 120 mg of DHA) for 6 weeks, starting in weeks 24–28 of gestation, showed a significant decrease in serum insulin levels and in HOMA index (homoeostasis model of assessment–IR)( Reference Samimi, Jamilian and Asemi 24 ), high-sensitivity C-reactive protein (CRP) and better newborn outcome( Reference Jamilian, Samimi and Kolahdooz 190 ), compared with placebo groups.

The need of higher PUFA intake in pregnancy is highlighted by data collected from 600 women participating in the Alberta Pregnancy Outcomes and Nutrition study( Reference Jia, Pakseresht and Wattar 191 ). Through the development of a dietary database for n-3 PUFA, the authors evaluated PUFA intake each trimester of pregnancy and 3 months postpartum: results indicate that the majority of women did not meet the European Union recommendation for DHA during pregnancy( Reference Jia, Pakseresht and Wattar 191 ).

The mechanism through which n-3 FA intake might influence insulin metabolism is not well known. It has been hypothesised that n-3 PUFA supplementation might affect insulin metabolism and lipid profiles by activating AMPK( Reference Higuchi, Shirai and Saito 192 ). In agreement with this finding, a recent study showed that RSV supplementation, before, and throughout, pregnancy increased placental DHA uptake capacity in pregnant non-human primates fed a high-fat diet (i.e. 36 % fat supplemented with 0·37 % RSV)( Reference O’Tierney-Ginn, Roberts and Gillingham 193 ). This effect was associated with increased AMPK activity and FATP-4, CD36 and FABPpm mRNA expression( Reference O’Tierney-Ginn, Roberts and Gillingham 193 ). In addition, n-3 PUFA might modulate gene expression through a variety of transcription factors with tissue-specific effects; n-3 PUFA improve insulin metabolism by inhibiting pro-inflammatory cytokine release and NF-κB protein expression( Reference Bellenger, Bellenger and Bataille 194 ), and by activating their G-protein-coupled receptor 120( Reference Oh, Talukdar and Bae 195 ).

A recent meta-analysis showed that long-term supplementation of marine-derived n-3 PUFA results in a significant reduction of IL-6, TNFα and CRP plasma levels, in both healthy and non-healthy subjects( Reference Li, Huang and Zheng 196 ).

Moreover, the increasing vegetarian and vegan population requires alternative sources of LC-PUFA to guarantee them the appropriate intake of n-3 FA( Reference Saunders, Davis and Garg 30 , Reference Harris 197 , Reference Rajaram 198 ). In addition to eating a larger quantity of vegetables, with a higher content of essential n-3 FA, a promising source of n-3 PUFA is represented by different species of microalgae( Reference Adarme-Vega, Lim and Timmins 199 , Reference Lenihan-Geels, Bishop and Ferguson 200 ). To this purpose, the European Commission has authorised the use in certain foods of oil derived from microalgae Schizochytrium sp. rich in DHA and EPA (EU decision no. 2015/545 and no. 2015/546),

Altogether, these data provide useful scientific information for planning future investigations aimed at exploring PUFA effects when considering a nutritional therapy of inflammatory diseases and GDM.

Conclusions and future research

GDM is increasing worldwide; obesity worsens this condition with increased risk of developing metabolic disorders in both mother and offspring later in life. The adoption of healthy lifestyle, with the adherence to a healthy dietary pattern, has positive effects on the prevention and management of diabetes. The Mediterranean diet is considered the model of healthy diet, and even more studies demonstrate its power in reducing the burden of several disorders in human being.

The few studies investigating the impact of healthy diet on GDM indicate that adherence to a healthy dietary pattern before pregnancy can reduce the risk of developing GDM; very few data about the association between dietary exposure and GDM outcomes are available.

Accordingly, this descriptive review tried to provide a picture on what we know about the impact of specific dietary biocompounds (i.e. polyphenols and PUFA) on the molecular mechanisms involved in glucose homoeostasis. Literature data, mostly derived from in vitro or animals studies, indicate that almost all subclasses of flavonoids, as well as the stilbene RSV and some olive oil phenolic compounds, interact and modulate several molecular pathways regulating insulin sensitivity in pancreatic β-cell, adipocyte, liver and muscle. Polyphenols drive activation and/ or silencing of transcription factors and consequently influence genes expression. Moreover, the fact that miRNA are the target of polyphenols action is to be taken into account in the future studies aimed at improving the understanding of the biological effects of polyphenols. However, how these compounds might work in GDM setting is a matter of speculation. Although increasing data indicate the beneficial effect of PUFA on pregnancy, a high-level evidence has not yet been achieved to make recommendations for GDM.

Many gaps, in the nutritional field and in the knowledge of pathophysiological processes, have to be filled in to progress in GDM prevention and management. GDM results from a complex interaction between the genetic background and environment. Identifying a number of modifiable biomarkers (including adipokines and miRNA) and the nutritional factors (e.g. polyphenols and their metabolites) able to target them can be useful in both early diagnosis and disease management.

It will be necessary to carry out dietary intervention studies involving large cohorts, and take into account genetic and dietary differences among races and countries. The ‘omics’ techniques will be fundamental tools to monitor dietary changes in populations and to identify the association between dietary exposure and disease outcomes. The integration of data from an individual’s genetic predisposition, endogenous metabolome, food metabolome and mirRNome profiles by systems biology approaches will provide a holistic view (systems biology approaches) of the relations between diet and disease, and will make it possible to translate these data in nutritional recommendations and diseases management. In particular, differences in metabolic profiles will be useful to identify novel biomarkers of dietary intake associated with disease. In this regard, the recent creation of a database to estimate intake and dietary seafood source of LC-PUFA in pregnancy provides a useful tool and represents an important step for future intervention studies.

Sharing this database along with others providing information on dietary polyphenols and polyphenol metabolites will help researchers to better understand the mechanisms governing interaction between dietary active compounds and the human organism.

Acknowledgements

The authors thank Monica Brocco for the linguistic revision.

This research received no specific grant from any funding agency, commercial or not-for-profit sectors.

C. S., R. M., A. Z. and S. M. contributed to the idea and development of the research. E. Mandosi, B. S. and E. Mari assisted with literature review and data collection. C. S., R. M., A. Z. and S. M. developed the first draft of the paper. All authors reviewed multiple drafts of the paper and approved the final manuscript for submission.

The authors declare that there are no conflicts of interest.

References

1. Chen, P, Wang, S, Ji, J, et al. (2015) Risk factors and management of gestational diabetes. Cell Biochem Biophys 71, 689694.CrossRefGoogle ScholarPubMed
2. Yuen, L & Wong, VW (2015) Gestational diabetes mellitus: challenges for different ethnic groups. World J Diabetes 6, 10241032.CrossRefGoogle ScholarPubMed
3. Bao, W, Yeung, E, Tobias, DK, et al. (2015) Long-term risk of type 2 diabetes mellitus in relation to BMI and weight change among women with a history of gestational diabetes mellitus: a prospective cohort study. Diabetologia 58, 12121219.CrossRefGoogle ScholarPubMed
4. Harris, DL, Weston, PJ & Harding, JE (2012) Incidence of neonatal hypoglycemia in babies identified as at risk. J Pediatr 161, 787791.CrossRefGoogle ScholarPubMed
5. Yessoufou, A & Moutairou, K (2011) Maternal diabetes in pregnancy: early and long-term outcomes on the offspring and the concept of ‘metabolic memory’. Exp Diabetes Res 2011, 218598.CrossRefGoogle ScholarPubMed
6. Benhalima, K, Mathieu, C, Damm, P, et al. (2015) A proposal for the use of uniform diagnostic criteria for gestational diabetes in Europe: an opinion paper by the European Board & College of Obstetrics and Gynaecology (EBCOG). Diabetologia 58, 14221429.CrossRefGoogle ScholarPubMed
7. Agarwal, MM (2015) Gestational diabetes mellitus: an update on the current international diagnostic criteria. World J Diabetes 6, 782791.CrossRefGoogle ScholarPubMed
8. Sacks, DA, Hadden, DR, Maresh, M, et al. (2012) Frequency of gestational diabetes mellitus at collaborating centers based on IADPSG consensus panel-recommended criteria: the Hyperglycemia and Adverse Pregnancy Outcome (HAPO) Study. Diabetes Care 35, 526528.CrossRefGoogle ScholarPubMed
9. Santangelo, C, Vari, R, Scazzocchio, B, et al. (2014) Management of reproduction and pregnancy complications in maternal obesity: which role for dietary polyphenols? Biofactors 40, 79102.CrossRefGoogle ScholarPubMed
10. Georgoulis, M, Kontogianni, MD & Yiannakouris, N (2014) Mediterranean diet and diabetes: prevention and treatment. Nutrients 6, 14061423.CrossRefGoogle ScholarPubMed
11. Sofi, F, Macchi, C, Abbate, R, et al. (2014) Mediterranean diet and health status: an updated meta-analysis and a proposal for a literature-based adherence score. Public Health Nutr 17, 27692782.CrossRefGoogle Scholar
12. Capurso, C, Massaro, M, Scoditti, E, et al. (2014) Vascular effects of the Mediterranean diet part I: anti-hypertensive and anti-thrombotic effects. Vascul Pharmacol 63, 118126.CrossRefGoogle ScholarPubMed
13. Esposito, K, Maiorino, MI, Bellastella, G, et al. (2015) A journey into a Mediterranean diet and type 2 diabetes: a systematic review with meta-analyses. BMJ Open 5, e008222.CrossRefGoogle ScholarPubMed
14. Bonaccio, M, Di Castelnuovo, A, Costanzo, S, et al. (2015) Adherence to the traditional Mediterranean diet and mortality in subjects with diabetes. Prospective results from the MOLI-SANI study. Eur J Prev Cardiol (epublication ahead of print version 3 February 2015).Google Scholar
15. Koloverou, E, Panagiotakos, DB, Pitsavos, C, et al. (2016) Adherence to Mediterranean diet and 10-year incidence (2002–2012) of diabetes: correlations with inflammatory and oxidative stress biomarkers in the ATTICA cohort study. Diabetes Metab Res Rev 32, 7381.CrossRefGoogle ScholarPubMed
16. Yang, J, Farioli, A, Korre, M, et al. (2014) Modified Mediterranean diet score and cardiovascular risk in a North American working population. PLOS ONE 9, e87539.CrossRefGoogle Scholar
17. Steffen, LM, Van Horn, L, Daviglus, ML, et al. (2014) A modified Mediterranean diet score is associated with a lower risk of incident metabolic syndrome over 25 years among young adults: the CARDIA (Coronary Artery Risk Development in Young Adults) study. Br J Nutr 112, 16541661.CrossRefGoogle ScholarPubMed
18. Hoscan, Y, Yigit, F & Muderrisoglu, H (2015) Adherence to Mediterranean diet and its relation with cardiovascular diseases in Turkish population. Int J Clin Exp Med 8, 28602866.Google ScholarPubMed
19. Grosso, G, Stepaniak, U, Micek, A, et al. (2015) A Mediterranean-type diet is associated with better metabolic profile in urban Polish adults: results from the HAPIEE study. Metabolism 64, 738746.CrossRefGoogle ScholarPubMed
20. Asemi, Z, Samimi, M, Tabassi, Z, et al. (2014) The effect of DASH diet on pregnancy outcomes in gestational diabetes: a randomized controlled clinical trial. Eur J Clin Nutr 68, 490495.CrossRefGoogle ScholarPubMed
21. Tobias, DK, Zhang, C, Chavarro, J, et al. (2012) Prepregnancy adherence to dietary patterns and lower risk of gestational diabetes mellitus. Am J Clin Nutr 96, 289295.CrossRefGoogle ScholarPubMed
22. Karamanos, B, Thanopoulou, A, Anastasiou, E, et al. (2014) Relation of the Mediterranean diet with the incidence of gestational diabetes. Eur J Clin Nutr 68, 813.CrossRefGoogle ScholarPubMed
23. Salas-Salvado, J, Martinez-Gonzalez, MA, Bullo, M, et al. (2011) The role of diet in the prevention of type 2 diabetes. Nutr Metab Cardiovasc Dis 21, Suppl. 2, B32B48.CrossRefGoogle ScholarPubMed
24. Samimi, M, Jamilian, M, Asemi, Z, et al. (2015) Effects of omega-3 fatty acid supplementation on insulin metabolism and lipid profiles in gestational diabetes: randomized, double-blind, placebo-controlled trial. Clin Nutr 34, 388393.CrossRefGoogle ScholarPubMed
25. Wang, H, Jiang, H, Yang, L, et al. (2015) Impacts of dietary fat changes on pregnant women with gestational diabetes mellitus: a randomized controlled study. Asia Pac J Clin Nutr 24, 5864.Google ScholarPubMed
26. Mirmiran, P, Bahadoran, Z & Azizi, F (2014) Functional foods-based diet as a novel dietary approach for management of type 2 diabetes and its complications: a review. World J Diabetes 5, 267281.CrossRefGoogle ScholarPubMed
27. Xiao, JB & Hogger, P (2015) Dietary polyphenols and type 2 diabetes: current insights and future perspectives. Curr Med Chem 22, 2338.CrossRefGoogle ScholarPubMed
28. Bahadoran, Z, Mirmiran, P & Azizi, F (2013) Dietary polyphenols as potential nutraceuticals in management of diabetes: a review. J Diabetes Metab Disord 12, 43.CrossRefGoogle ScholarPubMed
29. Oh, YS & Jun, HS (2014) Role of bioactive food components in diabetes prevention: effects on beta-cell function and preservation. Nutr Metab Insights 7, 5159.CrossRefGoogle ScholarPubMed
30. Saunders, AV, Davis, BC & Garg, ML (2013) Omega-3 polyunsaturated fatty acids and vegetarian diets. Med J Aust 199, S22S26.CrossRefGoogle ScholarPubMed
31. Trichopoulou, A, Bamia, C & Trichopoulos, D (2009) Anatomy of health effects of Mediterranean diet: Greek EPIC prospective cohort study. BMJ 338, b2337.CrossRefGoogle ScholarPubMed
32. Bravo, L (1998) Polyphenols: chemistry, dietary sources, metabolism, and nutritional significance. Nutr Rev 56, 317333.CrossRefGoogle ScholarPubMed
33. Scalbert, A, Manach, C, Morand, C, et al. (2005) Dietary polyphenols and the prevention of diseases. Crit Rev Food Sci Nutr 45, 287306.CrossRefGoogle ScholarPubMed
34. Neveu, V, Perez-Jimenez, J, Vos, F, et al. (2010) Phenol-Explorer: an online comprehensive database on polyphenol contents in foods. Database (Oxford) 2010, bap024.CrossRefGoogle ScholarPubMed
35. Manach, C, Scalbert, A, Morand, C, et al. (2004) Polyphenols: food sources and bioavailability. Am J Clin Nutr 79, 727747.CrossRefGoogle ScholarPubMed
36. Perez-Jimenez, J, Fezeu, L, Touvier, M, et al. (2011) Dietary intake of 337 polyphenols in French adults. Am J Clin Nutr 93, 12201228.CrossRefGoogle ScholarPubMed
37. Tsao, R (2010) Chemistry and biochemistry of dietary polyphenols. Nutrients 2, 12311246.CrossRefGoogle ScholarPubMed
38. Landete, JM (2012) Updated knowledge about polyphenols: functions, bioavailability, metabolism, and health. Cri Rev Food Sci Nutr 52, 936948.CrossRefGoogle ScholarPubMed
39. Del Rio, D, Costa, LG, Lean, ME, et al. (2010) Polyphenols and health: what compounds are involved? Nutr Metab Cardiovasc Dis 20, 16.CrossRefGoogle ScholarPubMed
40. Lancon, A, Michaille, JJ & Latruffe, N (2013) Effects of dietary phytophenols on the expression of microRNAs involved in mammalian cell homeostasis. J Sci Food Agric 93, 31553164.CrossRefGoogle ScholarPubMed
41. Santangelo, C, Vari, R, Scazzocchio, B, et al. (2007) Polyphenols, intracellular signalling and inflammation. Ann Ist Super Sanita 43, 394405.Google ScholarPubMed
42. Sebastian, RS, Wilkinson Enns, C, Goldman, JD, et al. (2015) A new database facilitates characterization of flavonoid intake, sources, and positive associations with diet quality among US adults. J Nutr 145, 12391248.CrossRefGoogle ScholarPubMed
43. Zamora-Ros, R, Forouhi, NG, Sharp, SJ, et al. (2014) Dietary intakes of individual flavanols and flavonols are inversely associated with incident type 2 diabetes in European populations. J Nutr 144, 335343.CrossRefGoogle ScholarPubMed
44. D’Archivio, M, Filesi, C, Vari, R, et al. (2010) Bioavailability of the polyphenols: status and controversies. Int J Mol Sci 11, 13211342.CrossRefGoogle ScholarPubMed
45. Fraga, CG, Galleano, M, Verstraeten, SV, et al. (2010) Basic biochemical mechanisms behind the health benefits of polyphenols. Mol Aspects Med 31, 435445.CrossRefGoogle ScholarPubMed
46. Monteagudo, C, Mariscal-Arcas, M, Rivas, A, et al. (2015) Proposal of a Mediterranean Diet Serving Score. PLOS ONE 10, e0128594.CrossRefGoogle ScholarPubMed
47. Gonzalez, S, Fernandez, M, Cuervo, A, et al. (2014) Dietary intake of polyphenols and major food sources in an institutionalised elderly population. J Hum Nutr Diet 27, 176183.CrossRefGoogle Scholar
48. Murphy, MM, Barraj, LM, Spungen, JH, et al. (2014) Global assessment of select phytonutrient intakes by level of fruit and vegetable consumption. Br J Nutr 112, 10041018.CrossRefGoogle ScholarPubMed
49. United States Department of Agriculture (USDA) Agricultural Research Service (ARS) (2012) Nutrient Data Laboratory USDA National Nutrient Database for Standard reference. http://www.nal.usda.gov/fnic/foodcomp/search Google Scholar
50. Wang, J, Tang, L & Wang, JS (2015) Biomarkers of dietary polyphenols in cancer studies: current evidence and beyond. Oxid Med Cell Longev 2015, 732302.CrossRefGoogle ScholarPubMed
51. Edmands, WM, Ferrari, P, Rothwell, JA, et al. (2015) Polyphenol metabolome in human urine and its association with intake of polyphenol-rich foods across European countries. Am J Clin Nutr 102, 905913.CrossRefGoogle ScholarPubMed
52. Scalbert, A, Brennan, L, Manach, C, et al. (2014) The food metabolome: a window over dietary exposure. Am J Clin Nutr 99, 12861308.CrossRefGoogle ScholarPubMed
53. Sonagra, AD, Biradar, SM, K, D, et al. (2014) Normal pregnancy – a state of insulin resistance. J Clin Diagn Res 8, CC01CC03.Google ScholarPubMed
54. Catalano, PM, Huston, L, Amini, SB, et al. (1999) Longitudinal changes in glucose metabolism during pregnancy in obese women with normal glucose tolerance and gestational diabetes mellitus. Am J Obstet Gynecol 180, 903916.CrossRefGoogle ScholarPubMed
55. Harlev, A & Wiznitzer, A (2010) New insights on glucose pathophysiology in gestational diabetes and insulin resistance. Curr Diab Rep 10, 242247.CrossRefGoogle ScholarPubMed
56. Catalano, PM (2014) Trying to understand gestational diabetes. Diabet Med 31, 273281.CrossRefGoogle ScholarPubMed
57. Friedman, JE, Ishizuka, T, Shao, J, et al. (1999) Impaired glucose transport and insulin receptor tyrosine phosphorylation in skeletal muscle from obese women with gestational diabetes. Diabetes 48, 18071814.CrossRefGoogle ScholarPubMed
58. Tersigni, C, Di Nicuolo, F, D’Ippolito, S, et al. (2011) Adipokines: new emerging roles in fertility and reproduction. Obstet Gynecol Surv 66, 4763.CrossRefGoogle ScholarPubMed
59. Li, N, Liu, E, Guo, J, et al. (2013) Maternal prepregnancy body mass index and gestational weight gain on pregnancy outcomes. PLOS ONE 8, e82310.CrossRefGoogle ScholarPubMed
60. Lappas, M (2014) Activation of inflammasomes in adipose tissue of women with gestational diabetes. Mol Cell Endocrinol 382, 7483.CrossRefGoogle ScholarPubMed
61. Krutzfeldt, J & Stoffel, M (2006) MicroRNAs: a new class of regulatory genes affecting metabolism. Cell Metab 4, 912.CrossRefGoogle ScholarPubMed
62. Mao, Y, Mohan, R, Zhang, S, et al. (2013) MicroRNAs as pharmacological targets in diabetes. Pharmacol Res 75, 3747.CrossRefGoogle ScholarPubMed
63. Guay, C, Roggli, E, Nesca, V, et al. (2011) Diabetes mellitus, a microRNA-related disease? Trans Res 157, 253264.CrossRefGoogle ScholarPubMed
64. Lynn, FC (2009) Meta-regulation: microRNA regulation of glucose and lipid metabolism. Trends Endocrinol Metab 20, 452459.CrossRefGoogle ScholarPubMed
65. Chen, H, Lan, HY, Roukos, DH, et al. (2014) Application of microRNAs in diabetes mellitus. J Endocrinol 222, R1R10.CrossRefGoogle ScholarPubMed
66. Dweep, H & Gretz, N (2015) miRWalk2.0: a comprehensive atlas of microRNA-target interactions. Nat Methods 12, 697.CrossRefGoogle ScholarPubMed
67. Collares, CV, Evangelista, AF, Xavier, DJ, et al. (2013) Identifying common and specific microRNAs expressed in peripheral blood mononuclear cell of type 1, type 2, and gestational diabetes mellitus patients. BMC Res Notes 6, 491.CrossRefGoogle ScholarPubMed
68. Fu, G, Brkic, J, Hayder, H, et al. (2013) MicroRNAs in human placental development and pregnancy complications. Int J Mol Sci 14, 55195544.CrossRefGoogle ScholarPubMed
69. Zhao, C, Dong, J, Jiang, T, et al. (2011) Early second-trimester serum miRNA profiling predicts gestational diabetes mellitus. PLoS ONE 6, e23925.CrossRefGoogle ScholarPubMed
70. Zhu, Y, Tian, F, Li, H, et al. (2015) Profiling maternal plasma microRNA expression in early pregnancy to predict gestational diabetes mellitus. Int J Gynaecol Obstet 130, 4953.CrossRefGoogle ScholarPubMed
71. Etheridge, A, Lee, I, Hood, L, et al. (2011) Extracellular microRNA: a new source of biomarkers. Mutat Res 717, 8590.CrossRefGoogle ScholarPubMed
72. He, A, Zhu, L, Gupta, N, et al. (2007) Overexpression of micro ribonucleic acid 29, highly up-regulated in diabetic rats, leads to insulin resistance in 3T3-L1 adipocytes. Mol Endocrinol 21, 27852794.CrossRefGoogle Scholar
73. Yang, WM, Jeong, HJ, Park, SY, et al. (2014) Induction of miR-29a by saturated fatty acids impairs insulin signaling and glucose uptake through translational repression of IRS-1 in myocytes. FEBS Lett 588, 21702176.CrossRefGoogle ScholarPubMed
74. Bagge, A, Clausen, TR, Larsen, S, et al. (2012) MicroRNA-29a is up-regulated in beta-cells by glucose and decreases glucose-stimulated insulin secretion. Biochem Biophys Res Commun 426, 266272.CrossRefGoogle ScholarPubMed
75. Nesca, V, Guay, C, Jacovetti, C, et al. (2013) Identification of particular groups of microRNAs that positively or negatively impact on beta cell function in obese models of type 2 diabetes. Diabetologia 56, 22032212.CrossRefGoogle ScholarPubMed
76. Shi, Z, Zhao, C, Guo, X, et al. (2014) Differential expression of microRNAs in omental adipose tissue from gestational diabetes mellitus subjects reveals miR-222 as a regulator of ERalpha expression in estrogen-induced insulin resistance. Endocrinology 155, 19821990.CrossRefGoogle ScholarPubMed
77. Zhou, B, Li, C, Qi, W, et al. (2012) Downregulation of miR-181a upregulates sirtuin-1 (SIRT1) and improves hepatic insulin sensitivity. Diabetologia 55, 20322043.CrossRefGoogle ScholarPubMed
78. Arola-Arnal, A & Blade, C (2011) Proanthocyanidins modulate microRNA expression in human HepG2 cells. PLoS ONE 6, e25982.CrossRefGoogle ScholarPubMed
79. Bridge, G, Monteiro, R, Henderson, S, et al. (2012) The microRNA-30 family targets DLL4 to modulate endothelial cell behavior during angiogenesis. Blood 120, 50635072.CrossRefGoogle ScholarPubMed
80. Pietro, L, Daher, S, Rudge, MV, et al. (2010) Vascular endothelial growth factor (VEGF) and VEGF-receptor expression in placenta of hyperglycemic pregnant women. Placenta 31, 770780.CrossRefGoogle ScholarPubMed
81. Joglekar, MV, Patil, D, Joglekar, VM, et al. (2009) The miR-30 family microRNAs confer epithelial phenotype to human pancreatic cells. Islets 1, 137147.CrossRefGoogle ScholarPubMed
82. Zhang, C, Tobias, DK, Chavarro, JE, et al. (2014) Adherence to healthy lifestyle and risk of gestational diabetes mellitus: prospective cohort study. BMJ 349, g5450.CrossRefGoogle ScholarPubMed
83. Hanhineva, K, Torronen, R, Bondia-Pons, I, et al. (2010) Impact of dietary polyphenols on carbohydrate metabolism. Int J Mol Sci 11, 13651402.CrossRefGoogle ScholarPubMed
84. Berna, G, Oliveras-Lopez, MJ, Jurado-Ruiz, E, et al. (2014) Nutrigenetics and nutrigenomics insights into diabetes etiopathogenesis. Nutrients 6, 53385369.CrossRefGoogle ScholarPubMed
85. Babu, PV, Liu, D & Gilbert, ER (2013) Recent advances in understanding the anti-diabetic actions of dietary flavonoids. J Nutr Biochem 24, 17771789.CrossRefGoogle ScholarPubMed
86. Garcia-Segura, L, Perez-Andrade, M & Miranda-Rios, J (2013) The emerging role of microRNAs in the regulation of gene expression by nutrients. J Nutrigenet Nutrigenomics 6, 1631.Google ScholarPubMed
87. Palmer, JD, Soule, BP, Simone, BA, et al. (2014) MicroRNA expression altered by diet: can food be medicinal? Ageing Res Rev 17, 1624.CrossRefGoogle ScholarPubMed
88. Milenkovic, D, Jude, B & Morand, C (2013) miRNA as molecular target of polyphenols underlying their biological effects. Free Radi Biol Med 64, 4051.CrossRefGoogle ScholarPubMed
89. Jimenez, M, Dorado, L, Hernandez-Perez, M, et al. (2014) Ankle-brachial index in screening for asymptomatic carotid and intracranial atherosclerosis. Atherosclerosis 233, 7275.CrossRefGoogle ScholarPubMed
90. Crozier, A, Jaganath, IB & Clifford, MN (2009) Dietary phenolics: chemistry, bioavailability and effects on health. Nat Prod Rep 26, 10011043.CrossRefGoogle ScholarPubMed
91. Fu, Z & Liu, D (2009) Long-term exposure to genistein improves insulin secretory function of pancreatic beta-cells. Eur J Pharmacol 616, 321327.CrossRefGoogle ScholarPubMed
92. Cai, EP & Lin, JK (2009) Epigallocatechin gallate (EGCG) and rutin suppress the glucotoxicity through activating IRS2 and AMPK signaling in rat pancreatic beta cells. J Agri Food Chem 57, 98179827.CrossRefGoogle ScholarPubMed
93. Yu, LF, Qiu, BY, Nan, FJ, et al. (2010) AMPK activators as novel therapeutics for type 2 diabetes. Curr Top Med Chem 10, 397410.CrossRefGoogle ScholarPubMed
94. Zhang, ZF, Li, Q, Liang, J, et al. (2010) Epigallocatechin-3-O-gallate (EGCG) protects the insulin sensitivity in rat L6 muscle cells exposed to dexamethasone condition. Phytomedicine 17, 1418.CrossRefGoogle ScholarPubMed
95. Tsang, WP & Kwok, TT (2010) Epigallocatechin gallate up-regulation of miR-16 and induction of apoptosis in human cancer cells. J Nutr Biochem 21, 140146.CrossRefGoogle ScholarPubMed
96. Takikawa, M, Inoue, S, Horio, F, et al. (2010) Dietary anthocyanin-rich bilberry extract ameliorates hyperglycemia and insulin sensitivity via activation of AMP-activated protein kinase in diabetic mice. J Nutr 140, 527533.CrossRefGoogle ScholarPubMed
97. Sun, CD, Zhang, B, Zhang, JK, et al. (2012) Cyanidin-3-glucoside-rich extract from Chinese bayberry fruit protects pancreatic beta cells and ameliorates hyperglycemia in streptozotocin-induced diabetic mice. J Med Food 15, 288298.CrossRefGoogle ScholarPubMed
98. Scazzocchio, B, Vari, R, Filesi, C, et al. (2011) Cyanidin-3-O-beta-glucoside and protocatechuic acid exert insulin-like effects by upregulating PPARgamma activity in human omental adipocytes. Diabetes 60, 22342244.CrossRefGoogle ScholarPubMed
99. Masella, R, Santangelo, C, D’Archivio, M, et al. (2012) Protocatechuic acid and human disease prevention: biological activities and molecular mechanisms. Curr Med Chem 19, 29012917.CrossRefGoogle ScholarPubMed
100. Scazzocchio, B, Vari, R, Filesi, C, et al. (2015) Protocatechuic acid activates key components of insulin signaling pathway mimicking insulin activity. Mol Nutr Food Res 59, 14721481.CrossRefGoogle ScholarPubMed
101. Bhattacharya, S, Oksbjerg, N, Young, JF, et al. (2014) Caffeic acid, naringenin and quercetin enhance glucose-stimulated insulin secretion and glucose sensitivity in INS-1E cells. Diabetes Obes Metab 16, 602612.CrossRefGoogle ScholarPubMed
102. Song, Z, Wang, H, Zhu, L, et al. (2015) Curcumin improves high glucose-induced INS-1 cell insulin resistance via activation of insulin signaling. Food Funct 6, 461469.CrossRefGoogle ScholarPubMed
103. Zhang, J, Du, Y, Wu, C, et al. (2010) Curcumin promotes apoptosis in human lung adenocarcinoma cells through miR-186* signaling pathway. Oncol Rep 24, 12171223.CrossRefGoogle ScholarPubMed
104. Sun, M, Estrov, Z, Ji, Y, et al. (2008) Curcumin (diferuloylmethane) alters the expression profiles of microRNAs in human pancreatic cancer cells. Mol Cancer Ther 7, 464473.CrossRefGoogle ScholarPubMed
105. Choi, MS, Jung, UJ, Yeo, J, et al. (2008) Genistein and daidzein prevent diabetes onset by elevating insulin level and altering hepatic gluconeogenic and lipogenic enzyme activities in non-obese diabetic (NOD) mice. Diabetes Metab Res Rev 24, 7481.CrossRefGoogle ScholarPubMed
106. Milenkovic, D, Deval, C, Gouranton, E, et al. (2012) Modulation of miRNA expression by dietary polyphenols in apoE deficient mice: a new mechanism of the action of polyphenols. PLOS ONE 7, e29837.CrossRefGoogle ScholarPubMed
107. Stull, AJ, Cash, KC, Johnson, WD, et al. (2010) Bioactives in blueberries improve insulin sensitivity in obese, insulin-resistant men and women. J Nutr 140, 17641768.CrossRefGoogle ScholarPubMed
108. Wedick, NM, Pan, A, Cassidy, A, et al. (2012) Dietary flavonoid intakes and risk of type 2 diabetes in US men and women. Am J Clin Nutr 95, 925933.CrossRefGoogle ScholarPubMed
109. Lappas, M (2014) Effect of pre-existing maternal obesity, gestational diabetes and adipokines on the expression of genes involved in lipid metabolism in adipose tissue. Metabolism 63, 250262.CrossRefGoogle ScholarPubMed
110. D’Ippolito, S, Tersigni, C, Scambia, G, et al. (2012) Adipokines, an adipose tissue and placental product with biological functions during pregnancy. Biofactors 38, 1423.CrossRefGoogle ScholarPubMed
111. Hauguel-de Mouzon, S & Guerre-Millo, M (2006) The placenta cytokine network and inflammatory signals. Placenta 27, 794798.CrossRefGoogle ScholarPubMed
112. Friis, CM, Paasche Roland, MC, Godang, K, et al. (2013) Adiposity-related inflammation: effects of pregnancy. Obesity 21, E124E130.CrossRefGoogle ScholarPubMed
113. Barbour, LA, McCurdy, CE, Hernandez, TL, et al. (2007) Cellular mechanisms for insulin resistance in normal pregnancy and gestational diabetes. Diabetes Care 30, Suppl. 2, S112S119.CrossRefGoogle ScholarPubMed
114. Li, HP, Chen, X & Li, MQ (2013) Gestational diabetes induces chronic hypoxia stress and excessive inflammatory response in murine placenta. Int J Clin Exp Pathol 6, 650659.Google ScholarPubMed
115. Xu, Y, Jin, B, Sun, L, et al. (2014) The expression of FoxO1 in placenta and omental adipose tissue of gestational diabetes mellitus. Exp Clin Endocrinol Diabetes 122, 287294.Google ScholarPubMed
116. Miehle, K, Stepan, H & Fasshauer, M (2012) Leptin, adiponectin and other adipokines in gestational diabetes mellitus and pre-eclampsia. Clin Endocrinol 76, 211.CrossRefGoogle ScholarPubMed
117. Retnakaran, R, Hanley, AJ, Raif, N, et al. (2004) Reduced adiponectin concentration in women with gestational diabetes: a potential factor in progression to type 2 diabetes. Diabetes Care 27, 799800.CrossRefGoogle ScholarPubMed
118. Rasouli, N & Kern, PA (2008) Adipocytokines and the metabolic complications of obesity. J Clin Endocrinol Metab 93, S64S73.CrossRefGoogle ScholarPubMed
119. Lacroix, M, Battista, MC, Doyon, M, et al. (2013) Lower adiponectin levels at first trimester of pregnancy are associated with increased insulin resistance and higher risk of developing gestational diabetes mellitus. Diabetes Care 36, 15771583.CrossRefGoogle ScholarPubMed
120. Hedderson, MM, Darbinian, J, Havel, PJ, et al. (2013) Low prepregnancy adiponectin concentrations are associated with a marked increase in risk for development of gestational diabetes mellitus. Diabetes Care 36, 39303937.CrossRefGoogle ScholarPubMed
121. Yamauchi, T, Kamon, J, Minokoshi, Y, et al. (2002) Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nat Med 8, 12881295.CrossRefGoogle ScholarPubMed
122. Visiedo, F, Bugatto, F, Sanchez, V, et al. (2013) High glucose levels reduce fatty acid oxidation and increase triglyceride accumulation in human placenta. Am J Physiol Endocrinol Metab 305, E205E212.CrossRefGoogle ScholarPubMed
123. Astapova, O & Leff, T (2012) Adiponectin and PPARgamma: cooperative and interdependent actions of two key regulators of metabolism. Vitam Horm 90, 143162.CrossRefGoogle ScholarPubMed
124. Berg, AH & Scherer, PE (2005) Adipose tissue, inflammation, and cardiovascular disease. Circ Res 96, 939949.CrossRefGoogle ScholarPubMed
125. Aye, IL, Rosario, FJ, Powell, TL, et al. (2015) Adiponectin supplementation in pregnant mice prevents the adverse effects of maternal obesity on placental function and fetal growth. Proc Natl Acad Sci U S A 112, 1285812863.CrossRefGoogle ScholarPubMed
126. Li, FY, Cheng, KK, Lam, KS, et al. (2011) Cross-talk between adipose tissue and vasculature: role of adiponectin. Acta Physiol (Oxf) 203, 167180.CrossRefGoogle ScholarPubMed
127. Lepercq, J, Cauzac, M, Lahlou, N, et al. (1998) Overexpression of placental leptin in diabetic pregnancy: a critical role for insulin. Diabetes 47, 847850.CrossRefGoogle ScholarPubMed
128. Lappas, M (2014) Markers of endothelial cell dysfunction are increased in human omental adipose tissue from women with pre-existing maternal obesity and gestational diabetes. Metabolism 63, 860873.CrossRefGoogle ScholarPubMed
129. Qiu, C, Williams, MA, Vadachkoria, S, et al. (2004) Increased maternal plasma leptin in early pregnancy and risk of gestational diabetes mellitus. Obstet Gynecol 103, 519525.CrossRefGoogle ScholarPubMed
130. Al-Badri, MR, Zantout, MS & Azar, ST (2015) The role of adipokines in gestational diabetes mellitus. Ther Adv Endocrinol Metab 6, 103108.CrossRefGoogle ScholarPubMed
131. Saucedo, R, Zarate, A, Basurto, L, et al. (2011) Relationship between circulating adipokines and insulin resistance during pregnancy and postpartum in women with gestational diabetes. Arch Med Res 42, 318323.CrossRefGoogle ScholarPubMed
132. Bao, W, Baecker, A, Song, Y, et al. (2015) Adipokine levels during the first or early second trimester of pregnancy and subsequent risk of gestational diabetes mellitus: a systematic review. Metabolism 64, 756764.CrossRefGoogle ScholarPubMed
133. Xu, J, Zhao, YH, Chen, YP, et al. (2014) Maternal circulating concentrations of tumor necrosis factor-alpha, leptin, and adiponectin in gestational diabetes mellitus: a systematic review and meta-analysis. ScientificWorldJournal 2014, 926932.CrossRefGoogle ScholarPubMed
134. Hegyi, K, Fulop, K, Kovacs, K, et al. (2004) Leptin-induced signal transduction pathways. Cell Biol Int 28, 159169.CrossRefGoogle ScholarPubMed
135. Santos-Alvarez, J, Goberna, R & Sanchez-Margalet, V (1999) Human leptin stimulates proliferation and activation of human circulating monocytes. Cell Immunol 194, 611.CrossRefGoogle ScholarPubMed
136. Wojcik, M, Chmielewska-Kassassir, M, Grzywnowicz, K, et al. (2014) The relationship between adipose tissue-derived hormones and gestational diabetes mellitus (GDM). Endokrynol Pol 65, 134142.Google ScholarPubMed
137. Li, YY, Xiao, R, Li, CP, et al. (2015) Increased plasma levels of FABP4 and PTEN is associated with more severe insulin resistance in women with gestational diabetes mellitus. Med Sci Monit 21, 426431.Google ScholarPubMed
138. Landberg, R, Naidoo, N & van Dam, RM (2012) Diet and endothelial function: from individual components to dietary patterns. Curr Opin Lipidol 23, 147155.CrossRefGoogle ScholarPubMed
139. Favero, G, Paganelli, C, Buffoli, B, et al. (2014) Endothelium and its alterations in cardiovascular diseases: life style intervention. Biomed Res Int 2014, 801896.CrossRefGoogle ScholarPubMed
140. Schwingshackl, L & Hoffmann, G (2014) Mediterranean dietary pattern, inflammation and endothelial function: a systematic review and meta-analysis of intervention trials. Nutr Metab Cardiovasc Dis 24, 929939.CrossRefGoogle ScholarPubMed
141. Caselli, C (2014) Role of adiponectin system in insulin resistance. Mol Genet Metab 113, 155160.CrossRefGoogle ScholarPubMed
142. Wang, A, Liu, M, Liu, X, et al. (2011) Up-regulation of adiponectin by resveratrol: the essential roles of the Akt/FOXO1 and AMP-activated protein kinase signaling pathways and DsbA-L. J Biol Chem 286, 6066.CrossRefGoogle ScholarPubMed
143. Eseberri, I, Lasa, A, Churruca, I, et al. (2013) Resveratrol metabolites modify adipokine expression and secretion in 3T3-L1 pre-adipocytes and mature adipocytes. PLOS ONE 8, e63918.CrossRefGoogle ScholarPubMed
144. Kang, L, Heng, W, Yuan, A, et al. (2010) Resveratrol modulates adipokine expression and improves insulin sensitivity in adipocytes: relative to inhibition of inflammatory responses. Biochimie 92, 789796.CrossRefGoogle ScholarPubMed
145. Peng, XL, Qu, W, Wang, LZ, et al. (2014) Resveratrol ameliorates high glucose and high-fat/sucrose diet-induced vascular hyperpermeability involving Cav-1/eNOS regulation. PLOS ONE 9, e113716.CrossRefGoogle ScholarPubMed
146. Liu, Y, Li, D, Zhang, Y, et al. (2014) Anthocyanin increases adiponectin secretion and protects against diabetes-related endothelial dysfunction. Am J Physiol Endocrinol Metab 306, E975E988.CrossRefGoogle ScholarPubMed
147. Jin, T, Kim, OY, Shin, MJ, et al. (2014) Fisetin up-regulates the expression of adiponectin in 3T3-L1 adipocytes via the activation of silent mating type information regulation 2 homologue 1 (SIRT1)-deacetylase and peroxisome proliferator-activated receptors (PPARs). J Agri Food Chem 62, 1046810474.CrossRefGoogle ScholarPubMed
148. Khan, N, Syed, DN, Ahmad, N, et al. (2013) Fisetin: a dietary antioxidant for health promotion. Antioxid Redox Signal 19, 151162.CrossRefGoogle ScholarPubMed
149. Scoditti, E, Massaro, M, Carluccio, MA, et al. (2015) Additive regulation of adiponectin expression by the Mediterranean diet olive oil components oleic acid and hydroxytyrosol in human adipocytes. PLOS ONE 10, e0128218.CrossRefGoogle ScholarPubMed
150. Lamy, S, Ouanouki, A, Beliveau, R, et al. (2014) Olive oil compounds inhibit vascular endothelial growth factor receptor-2 phosphorylation. Exp Cell Res 322, 8998.CrossRefGoogle ScholarPubMed
151. Niwa, T, Yokoyama, S, Ito, T, et al. (2010) Reduction of leptin secretion by soy isoflavonoids in murine adipocytes in vitro . Phytochem Lett 3, 122125.CrossRefGoogle Scholar
152. Yanagisawa, M, Sugiya, M, Iijima, H, et al. (2012) Genistein and daidzein, typical soy isoflavones, inhibit TNF-alpha-mediated downregulation of adiponectin expression via different mechanisms in 3T3-L1 adipocytes. Mol Nutr Food Res 56, 17831793.CrossRefGoogle ScholarPubMed
153. Chen, J, Mo, H, Guo, R, et al. (2014) Inhibition of the leptin-induced activation of the p38 MAPK pathway contributes to the protective effects of naringin against high glucose-induced injury in H9c2 cardiac cells. Int J Mol Med 33, 605612.CrossRefGoogle Scholar
154. Mahmoud, AM, Ashour, MB, Abdel-Moneim, A, et al. (2012) Hesperidin and naringin attenuate hyperglycemia-mediated oxidative stress and proinflammatory cytokine production in high fat fed/streptozotocin-induced type 2 diabetic rats. J Diabetes Complications 26, 483490.CrossRefGoogle ScholarPubMed
155. Sakane, N, Kotani, K, Tsuzaki, K, et al. (2014) Equol producers can have low leptin levels among prediabetic and diabetic females. Ann Endocrinol (Paris) 75, 2528.CrossRefGoogle ScholarPubMed
156. Llaneza, P, Gonzalez, C, Fernandez-Inarrea, J, et al. (2011) Soy isoflavones, diet and physical exercise modify serum cytokines in healthy obese postmenopausal women. Phytomedicine 18, 245250.CrossRefGoogle ScholarPubMed
157. Gupta, SC, Tyagi, AK, Deshmukh-Taskar, P, et al. (2014) Downregulation of tumor necrosis factor and other proinflammatory biomarkers by polyphenols. Arch Biochem Biophys 559, 9199.CrossRefGoogle ScholarPubMed
158. Rebholz, SL, Burke, KT, Yang, Q, et al. (2011) Dietary fat impacts fetal growth and metabolism: uptake of chylomicron remnant core lipids by the placenta. Am J Physiol Endocrinol Metab 301, E416E425.CrossRefGoogle ScholarPubMed
159. Jones, ML, Mark, PJ & Waddell, BJ (2014) Maternal dietary omega-3 fatty acids and placental function. Reproduction 147, R143R152.CrossRefGoogle ScholarPubMed
160. Herrera, E & Ortega-Senovilla, H (2010) Disturbances in lipid metabolism in diabetic pregnancy – are these the cause of the problem? Best Pract Res Clin Endocrinol Metab 24, 515525.CrossRefGoogle ScholarPubMed
161. Haggarty, P (2010) Fatty acid supply to the human fetus. Ann Rev Nutr 30, 237255.CrossRefGoogle Scholar
162. Gil-Sanchez, A, Demmelmair, H, Parrilla, JJ, et al. (2011) Mechanisms involved in the selective transfer of long chain polyunsaturated fatty acids to the fetus. Front Genet 2, 57.CrossRefGoogle ScholarPubMed
163. Pagan, A, Prieto-Sanchez, MT, Blanco-Carnero, JE, et al. (2013) Materno-fetal transfer of docosahexaenoic acid is impaired by gestational diabetes mellitus. Am J Physiol Endocrinol Metab 305, E826E833.CrossRefGoogle ScholarPubMed
164. Araujo, JR, Correia-Branco, A, Ramalho, C, et al. (2013) Gestational diabetes mellitus decreases placental uptake of long-chain polyunsaturated fatty acids: involvement of long-chain acyl-CoA synthetase. J Nutr Biochem 24, 17411750.CrossRefGoogle ScholarPubMed
165. Gauster, M, Hiden, U, van Poppel, M, et al. (2011) Dysregulation of placental endothelial lipase in obese women with gestational diabetes mellitus. Diabetes 60, 24572464.CrossRefGoogle ScholarPubMed
166. Barrett, HL, Kubala, MH, Scholz Romero, K, et al. (2014) Placental lipases in pregnancies complicated by gestational diabetes mellitus (GDM). PLOS ONE 9, e104826.CrossRefGoogle ScholarPubMed
167. Duttaroy, AK (2009) Transport of fatty acids across the human placenta: a review. Prog Lipid Res 48, 5261.CrossRefGoogle ScholarPubMed
168. Jawerbaum, A & Capobianco, E (2011) Review: effects of PPAR activation in the placenta and the fetus: implications in maternal diabetes. Placenta 32, Suppl. 2, S212S217.CrossRefGoogle ScholarPubMed
169. Simopoulos, AP (2011) Evolutionary aspects of diet: the omega-6/omega-3 ratio and the brain. Mol Neurobiol 44, 203215.CrossRefGoogle ScholarPubMed
170. Calder, PC (2012) Mechanisms of action of (n-3) fatty acids. J Nutr 142, 592S599S.CrossRefGoogle ScholarPubMed
171. Russo, GL (2009) Dietary n-6 and n-3 polyunsaturated fatty acids: from biochemistry to clinical implications in cardiovascular prevention. Biochem Pharmacol 77, 937946.CrossRefGoogle ScholarPubMed
172. Zinati, Z, Zamansani, F, Hossein KayvanJoo, A, et al. (2014) New layers in understanding and predicting alpha-linolenic acid content in plants using amino acid characteristics of omega-3 fatty acid desaturase. Comput Biol Med 54, 1423.CrossRefGoogle ScholarPubMed
173. Burns-Whitmore, B, Haddad, E, Sabate, J, et al. (2014) Effects of supplementing n-3 fatty acid enriched eggs and walnuts on cardiovascular disease risk markers in healthy free-living lacto-ovo-vegetarians: a randomized, crossover, free-living intervention study. Nutr J 13, 29.CrossRefGoogle ScholarPubMed
174. Mayneris-Perxachs, J, Sala-Vila, A, Chisaguano, M, et al. (2014) Effects of 1-year intervention with a Mediterranean diet on plasma fatty acid composition and metabolic syndrome in a population at high cardiovascular risk. PLOS ONE 9, e85202.CrossRefGoogle Scholar
175. Galli, C & Marangoni, F (2006) N-3 fatty acids in the Mediterranean diet. Prostaglandins Leukot Essent Fatty Acids 75, 129133.CrossRefGoogle ScholarPubMed
176. Uddin, MK, Juraimi, AS, Hossain, MS, et al. (2014) Purslane weed (Portulaca oleracea): a prospective plant source of nutrition, omega-3 fatty acid, and antioxidant attributes. ScientificWorldJournal 2014, 951019.CrossRefGoogle ScholarPubMed
177. Panagiotakos, D, Kalogeropoulos, N, Pitsavos, C, et al. (2009) Validation of the MedDietScore via the determination of plasma fatty acids. Int J Food Sci Nutr 60, Suppl. 5, 168180.CrossRefGoogle ScholarPubMed
178. Feart, C, Torres, MJ, Samieri, C, et al. (2011) Adherence to a Mediterranean diet and plasma fatty acids: data from the Bordeaux sample of the Three-City study. Br J Nutr 106, 149158.CrossRefGoogle ScholarPubMed
179. Ambring, A, Johansson, M, Axelsen, M, et al. (2006) Mediterranean-inspired diet lowers the ratio of serum phospholipid n-6 to n-3 fatty acids, the number of leukocytes and platelets, and vascular endothelial growth factor in healthy subjects. Am J Clin Nutr 83, 575581.CrossRefGoogle ScholarPubMed
180. Scoditti, E, Capurso, C, Capurso, A, et al. (2014) Vascular effects of the Mediterranean diet-part II: role of omega-3 fatty acids and olive oil polyphenols. Vascul Pharmacol 63, 127134.CrossRefGoogle ScholarPubMed
181. Koletzko, B, Lien, E, Agostoni, C, et al. (2008) The roles of long-chain polyunsaturated fatty acids in pregnancy, lactation and infancy: review of current knowledge and consensus recommendations. J Perinat Med 36, 514.CrossRefGoogle ScholarPubMed
182. Pietrantoni, E, Del Chierico, F, Rigon, G, et al. (2014) Docosahexaenoic acid supplementation during pregnancy: a potential tool to prevent membrane rupture and preterm labor. Int J Mol Sci 15, 80248036.CrossRefGoogle ScholarPubMed
183. Carvajal, JA (2014) Docosahexaenoic acid supplementation early in pregnancy may prevent deep placentation disorders. Biomed Res Int 2014, 526895.CrossRefGoogle ScholarPubMed
184. Mohammadi, E, Rafraf, M, Farzadi, L, et al. (2012) Effects of omega-3 fatty acids supplementation on serum adiponectin levels and some metabolic risk factors in women with polycystic ovary syndrome. Asia Pac J Clin Nutr 21, 511518.Google ScholarPubMed
185. Lu, J, Borthwick, F, Hassanali, Z, et al. (2011) Chronic dietary n-3 PUFA intervention improves dyslipidaemia and subsequent cardiovascular complications in the JCR:LA-cp rat model of the metabolic syndrome. Br J Nutr 105, 15721582.Google Scholar
186. Min, Y, Djahanbakhch, O, Hutchinson, J, et al. (2014) Effect of docosahexaenoic acid-enriched fish oil supplementation in pregnant women with type 2 diabetes on membrane fatty acids and fetal body composition-double-blinded randomized placebo-controlled trial. Diabet Med 31, 13311340.CrossRefGoogle ScholarPubMed
187. Recchiuti, A & Serhan, CN (2012) Pro-resolving lipid mediators (SPMs) and their actions in regulating miRNA in novel resolution circuits in inflammation. Front Immunol 3, 298.CrossRefGoogle ScholarPubMed
188. Jones, ML, Mark, PJ, Keelan, JA, et al. (2013) Maternal dietary omega-3 fatty acid intake increases resolvin and protectin levels in the rat placenta. J Lipid Res 54, 22472254.CrossRefGoogle ScholarPubMed
189. Keelan, JA, Mas, E, D’Vaz, N, et al. (2015) Effects of maternal n-3 fatty acid supplementation on placental cytokines, pro-resolving lipid mediators and their precursors. Reproduction 149, 171178.CrossRefGoogle ScholarPubMed
190. Jamilian, M, Samimi, M, Kolahdooz, F, et al. (2016) Omega-3 fatty acid supplementation affects pregnancy outcomes in gestational diabetes: a randomized, double-blind, placebo-controlled trial. J Matern Fetal Neonatal Med 29, 669675.CrossRefGoogle ScholarPubMed
191. Jia, X, Pakseresht, M, Wattar, N, et al. (2015) Women who take n-3 long-chain polyunsaturated fatty acid supplements during pregnancy and lactation meet the recommended intake. App Physiol Nutr Metab 40, 474481.CrossRefGoogle ScholarPubMed
192. Higuchi, T, Shirai, N, Saito, M, et al. (2008) Levels of plasma insulin, leptin and adiponectin, and activities of key enzymes in carbohydrate metabolism in skeletal muscle and liver in fasted ICR mice fed dietary n-3 polyunsaturated fatty acids. J Nutr Biochem 19, 577586.CrossRefGoogle ScholarPubMed
193. O’Tierney-Ginn, P, Roberts, V, Gillingham, M, et al. (2015) Influence of high fat diet and resveratrol supplementation on placental fatty acid uptake in the Japanese macaque. Placenta 36, 903910.CrossRefGoogle ScholarPubMed
194. Bellenger, J, Bellenger, S, Bataille, A, et al. (2011) High pancreatic n-3 fatty acids prevent STZ-induced diabetes in fat-1 mice: inflammatory pathway inhibition. Diabetes 60, 10901099.CrossRefGoogle ScholarPubMed
195. Oh, DY, Talukdar, S, Bae, EJ, et al. (2010) GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects. Cell 142, 687698.CrossRefGoogle ScholarPubMed
196. Li, K, Huang, T, Zheng, J, et al. (2014) Effect of marine-derived n-3 polyunsaturated fatty acids on C-reactive protein, interleukin 6 and tumor necrosis factor alpha: a meta-analysis. PLOS ONE 9, e88103.CrossRefGoogle ScholarPubMed
197. Harris, WS (2014) Achieving optimal n-3 fatty acid status: the vegetarian’s challenge… or not. Am J Clin Nutr 100, Suppl. 1, 449S452S.CrossRefGoogle ScholarPubMed
198. Rajaram, S (2014) Health benefits of plant-derived alpha-linolenic acid. Am J Clin Nutr 100, Suppl. 1, 443S448S.CrossRefGoogle ScholarPubMed
199. Adarme-Vega, TC, Lim, DK, Timmins, M, et al. (2012) Microalgal biofactories: a promising approach towards sustainable omega-3 fatty acid production. Microb Cell Fact 11, 96.CrossRefGoogle ScholarPubMed
200. Lenihan-Geels, G, Bishop, KS & Ferguson, LR (2013) Alternative sources of omega-3 fats: can we find a sustainable substitute for fish? Nutrients 5, 13011315.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1 Chemical structure and main dietary source of polyphenols discussed in this review regarding their capability to modulate glucose metabolism signalling.

Figure 1

Fig. 2 Crosstalk among signalling pathways in regulating glucose metabolism. All of the factors that appear in this scheme are potential points of action of polyphenols. , Activation; , inhibition; , modulation; JAK/STAT, Janus kinase/signal transducer and activator of transcription; AMPK, AMP-activated protein kinase; JNK, c-Jun N-terminal kinase; IRS1/2, insulin receptor substrate 1/2; MAPK, mitogen-activated protein kinases; PDK1, 3-phosphoinositide-dependent protein kinase-1; PI3K, phosphatidylinositol-3-kinase; Akt, protein kinase B; SIRT-1, sirtuin 1; NO, nitric oxide; eNOS, endothelial NO synthase; FOXO1, forkhead box protein O1; TF, transcription factors; miRNA, microRNA; FA, fatty acids.

Figure 2

Table 1 Effects of dietary polyphenols on molecular mechanisms associated with gestational diabetes mellitus

Figure 3

Table 2 Maternal n-3 fatty acids (FA) supplementation