Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Jul 14.
Published in final edited form as: Exp Biol Med (Maywood). 2013 May 29;238(5):482–490. doi: 10.1177/1535370212473704

Diabetic complications in pregnancy: is resveratrol a solution?

Chandra K Singh 1,2, Ambrish Kumar 1, Holly A LaVoie 3, Donald J DiPette 3, Ugra S Singh 1
PMCID: PMC4501251  NIHMSID: NIHMS706127  PMID: 23436883

Abstract

Diabetes is a metabolic disorder that, during pregnancy, may affect fetal development. Fetal outcome depends on the type of diabetes present, the concentration of blood glucose and the extent of fetal exposure to elevated or frequently fluctuating glucose concentrations. The result of some diabetic pregnancies will be embryonic developmental abnormalities, a condition referred to as diabetic embryopathy. Tight glycemic control in type 1 diabetes during pregnancy using insulin therapy together with folic acid supplementation are partially able to prevent diabetic embryopathy; however, the protection is not complete and additional interventions are needed. Resveratrol, a polyphenol found largely in the skins of red grapes, is known to have antidiabetic action and is in clinical trials for the treatment of diabetes, insulin resistance, obesity and metabolic syndrome. Studies of resveratrol in a rodent model of diabetic embryopathy reveal that it significantly improves the embryonic outcome in terms of diminishing developmental abnormalities. Improvements in maternal and embryonic outcomes observed in rodent models may arise from resveratrol’s antioxidative potential, antidiabetic action and antidyslipidemic nature. Whether resveratrol will have similar actions in human diabetic pregnancy is unknown. Here, we review the potential therapeutic use of resveratrol in diabetes and diabetic pregnancy.

Keywords: diabetic pregnancy, neural tube defects, resveratrol, hyperglycemia

Introduction

Diabetes is a metabolic disease in which high blood-glucose concentrations exist either because the body does not produce sufficient insulin (type 1 diabetes) or because cells fail to properly respond to insulin (type 2 diabetes).1 A third kind of diabetes, known as gestational diabetes, occurs when a pregnant woman develops high blood-glucose levels typically during the second half of pregnancy due to the hormones of pregnancy.2 Gestational diabetes can revert to normal following pregnancy, but sometimes precedes the development of type 2 diabetes. Recent data of the National Diabetes Fact Sheet shows that total 25.8 million children and adults in the USA (8.3% of the population) have diabetes (Centers for Disease Control and Prevention, 2011; http://www.cdc.gov/diabetes/pubs/pdf/ndfs_2011.pdf).3 All forms of diabetes increase the risk of long-term complications. Up to 67% of adults with diabetes have high blood pressure.3 Long-term diabetes damages blood vessels. Diabetics have double the risk of heart attack or stroke. Diabetes causes nerve damage in the heart, which can make a heart attack painless or ‘silent,’ and affects blood vessel formation in the retina of the eye, leading to visual symptoms and sometimes blindness (diabetic retinopathy).1,3 High blood sugar can lead to scarring of kidney tissues that may result in chronic kidney diseases (diabetic nephropathy). Diabetes also affects the nervous system (diabetic neuropathy), causing numbness, tingling and pain in the feet, and increasing the risk of skin damage because of altered sensation.1,3 As diabetes affects every part of the body, it can also have devastating complications to the mother and embryo.

Diabetic pregnancies are a serious concern

The incidence of diabetes mellitus and its sequelae is rising in the general population, and it is an additional risk factor in pregnancy, causing birth defects commonly referred to as diabetic embryopathies. Although most embryonic organs are affected by maternal diabetes, the central nervous system is comparatively more susceptible to high glucose.413 Among the various malformations in diabetic embryopathy, neural tube defects (NTDs) are one of the most common.5,1416 Although the incidence of NTDs in diabetic pregnancies has been reduced by intensive insulin treatment and glucose monitoring, they are still 2- to 6-fold greater than in normal pregnancies.1719 Folic acid consumption alone or in combination with vitamin E diminishes the diabetes-induced embryonic malformations in diabetic pregnancy.2022 Currently, folic acid supplementation is standard practice to avoid birth defects, but the protection afforded by folic acid supplementation against diabetes-induced birth defects is not enough. Approximately 50% of congenital malformations can be prevented if folic acid is supplemented before and during pregnancy.2325 The folic acid tolerable intake limit is thought to be 1 mg/day; however, a few studies advocate the use of high-dose folic acid (4–5 mg/day) during diabetic pregnancy.26 Folate is needed for DNA synthesis but can also serve as a methyl group donor for DNA methylation, and thereby alter the embryo’s epigenome by epigenetic modification of DNA.27 Recently, DNA methylation by folate and the C677T mutation of the enzyme methylenetetrahydrofolate reductase (MTHFR), and thereby impaired MTHFR function have been shown as one of the cause of recurrent fetal loss and potential factor in the etiology of autism.28,29 There are currently no drugs or nutritional supplements available for complete protection against embryonic malformation during diabetic pregnancy, and NTDs and other birth defects are still a serious concern in diabetic pregnancies. These gaps in knowledge provide a compelling reason to further investigate alternative options for the prevention of neural tube defects in diabetic pregnancies.

Resveratrol, a grape antioxidant, and prevention of diabetic embryopathy

Recent studies have demonstrated that resveratrol (trans-3,5,4′-trihydroxystilbene), found in red grapes and blueberries, lowers maternal blood sugar, improves maternal lipid profile30 and prevents developmental delays in embryos of rat diabetic dams.31 On the basis of these results, resveratrol could be a potential therapeutic agent for the prevention of embryonic malformations including NTDs.31 Our understanding of the signaling mechanisms involved in diabetes mellitus has progressed greatly, but very little is known about the mechanism(s) underlying NTDs in diabetic pregnancy. Further studies are needed to determine whether the combined effects of resveratrol and folic acid can completely abolish the birth defects in diabetic pregnancies. Here, we review the literature and provide some new insights to aid further understanding of the signaling mechanisms associated with NTDs and how resveratrol may prevent these defects.

Various plants produce resveratrol to help defend against invading fungi, stress, injury, infection and excessive sunlight. Resveratrol is found in both cis- and trans-stereoisomeric forms existing as a glucoside (bound to a glucose molecule). The roots of the plant Polygonum cuspidatum (KO-jo-kon), mainly cultivated in Asia, provides a rich source of resveratrol from which commercially available trans-resveratrol (>98% pure) is isolated by high-speed counter-current chromatography.32 Resveratrol has been reported to possess many salutary effects, including cardioprotection,33 chemoprevention,34,35 and extension of lifespan in several species.36,37 However, the molecular mechanisms underlying the resveratrol functions are different in different diseases. Resveratrol has been in use since ancient times as an herbal formulation termed ‘Darakchasava.’ The ancient medicinal book of Hindus, Ayurveda, more than 4500 years ago described Darakchasava, a heart tonic made from fermented grapes. The formulation is believed to counteract lethargy, weakness and heat exhaustion and stimulate cardiac function. Darakchasava is also thought to have antipyretic, diuretic and diaphoretic effects. Today, Darakchasava contains 1.3–6.0 mg/L of resveratrol, depending on the pharmaceutical company producing it.38

Managing diabetes is one of modern medicine’s major challenges, and it remains one of the leading causes of morbidity and mortality among tens of millions of people in the USA alone. Resveratrol has shown promising results in the management of diabetes. In diet-induced obese and diabetic mice, long-term intracerebroventricular infusion of resveratrol corrects hyperglycemia and improves hyperinsulinemia.39 In high-fat-diet-fed mice, it ameliorates insulin resistance, increases the mitochondrial content of cells and prolongs survival.40 Resveratrol has beneficial effects on dyslipidemia and obesity. In another study, resveratrol demonstrated significant antihyperglycemic activity, improving insulin levels and oral glucose tolerance tests in ob/ob mice.41 Resveratrol action has been linked to the activation of a histone deacetylase Sirtuin 1 (Sirt1).40,41 Administration of resveratrol to diabetic rats results in a significant decrease in blood-glucose concentrations, glycosylated hemoglobin, blood urea and other blood compounds that are usually elevated in type II diabetes and cause damage to body organs.39,42 During starvation or limited food, Sirt1 promotes the breakdown of fat stores as a source of energy. Sirt1 is activated through caloric restriction and lengthens rodent lifespan and improves overall health by aiding fat metabolism, increasing immune function, and boosting cardiovascular health.43 Notably, when Sirt1 is activated by resveratrol, diabetes is improved.41

Resveratrol also activates the 5′-AMP-activated protein kinase (AMPK), which also regulates insulin sensitivity and mitochondrial biogenesis.43 The net effect of AMPK activation is stimulation of hepatic fatty-acid oxidation and ketogenesis; increase of skeletal muscle fatty acid oxidation and muscle glucose uptake; inhibition of cholesterol synthesis, lipogenesis, triglyceride synthesis, adipocyte lipolysis; and modulation of insulin secretion by pancreatic beta-cells. In mice deficient in the catalytic subunit of AMPK (α1 or α2), resveratrol fails to increase insulin sensitivity, glucose tolerance, mitochondrial biogenesis and physical endurance,44 indicating the necessity of AMPK in multiple resveratrol-mediated effects.

Resveratrol is safe, but its bioavailability is poor

Resveratrol use in humans has no pronounced toxicity,45 and orally administered trans-resveratrol is well-absorbed. A study performed by Walle et al.46 using 14C-trans-resveratrol (oral dose of 25 mg) in humans showed that 70% of the resveratrol dose was absorbed by the body. Metabolites of resveratrol (conjugated forms) in plasma peaked at 30–60 min postadministration with a plasma half-life of 9.2 h.46 In contrast, only small amounts of unmodified resveratrol (<5 ng/mL) were detected in plasma in a similar timeframe. Resveratrol absorbed from the gastrointestinal tract is rapidly metabolized to glucuronide (3-O-glucuronide and 4′-O-glucuronide), sulfate (resveratrol 3-O-sulfate), and hydroxylate forms. The majority of orally dosed resveratrol is found in urine as sulfate- or glucuronic acid-conjugates. Food intake with a single dose of 400 mg trans-resveratrol delays absorption.47 Thus, orally ingested resveratrol is both rapidly metabolized and excreted, accounting for its low bioavailability. However, a few studies show that the bioavailability of resveratrol can be enhanced by using more potent resveratrol analogs (i.e. SRT501)48 and enhanced delivery methods such as liposomal encapsulation,49 or combining it with another natural product from black pepper (Piper spp.), piperine.50 However, further validation of these strategies will be needed before resveratrol can be used in diabetic pregnancy.

Health benefits of resveratrol

According to ClinicalTrials.gov (http://clinicaltrials.gov/), there are more than 50 recent studies involving resveratrol. These trials investigate the possible role of resveratrol in different diseases such as diabetes, Alzheimer’s disease and cancer. There are 12 clinical trials where resveratrol is specifically being focused for type 2 diabetes, obesity, insulin resistance or metabolic syndrome.51,52 One of the human clinical trial is investigating whether resveratrol when given orally to obese and type 2 diabetic subjects, will decrease reactive oxygen species (ROS) generation and prevent the production of the pro-inflammatory transcription factor nuclear factor-κB.51 A second trial is investigating the cellular and molecular effects of resveratrol on inflammatory mediators and insulin resistance in obese non-diabetic and type 2 diabetic subjects (http://clinicaltrials.gov/). Another clinical trial is being conducted to determine the effect of resveratrol on endothelial function.53 The endothelium participates in the control of blood vessel function, in part by regulating vessel dilation via nitric oxide; however, endothelial function becomes abnormal in diabetic patients, and this abnormality contributes to the development of cardiovascular disease.33 In diabetic (Leprdb) mice, resveratrol restores endothelial function by interfering with tumor necrosis factor α-mediated activation of NADPH oxidase and thereby preserves nitric oxide availability.54 Preclinical observations suggest that resveratrol is safe and has enormous potential in the treatment of obesity and insulin resistance in humans. Current investigations are evaluating whether resveratrol is able to counteract the detrimental effects of obesity. One trial is examining the effect of resveratrol on improving the metabolic profile of adults with insulin resistance (http://clinical-trials.gov/). Resveratrol’s antioxidative potential is being compared with calorie restriction in clinical trials to determine: (i) gene expression and lipid profile changes, (ii) insulin’s efficacy to control blood sugar and (iii) changes in other blood and tissue markers of metabolic and cardiovascular health.51 Another current study with obese subjects is aimed to test resveratrol’s ability to improve mitochondrial activity and fatty acid oxidative capacity in skeletal muscle upon a high-fat challenge (http://clinicaltrials.gov/).

Resveratrol improves maternal glucose and lipid profile during diabetic pregnancy

The ability of resveratrol to lower blood glucose is well documented.42,55 In rodent studies, we tested this ability of resveratrol to lower blood glucose in pregnant diabetic dams where diabetes was induced with streptozotocin. Glucose concentrations were measured in dams on every other day of pregnancy and significant decreases at gestation day 9 (22.80%) and 12 (33.32%) were found in the resveratrol-treated diabetic group compared with the diabetic only group.31 In addition, significantly increased insulin levels were present in the resveratrol-fed diabetic groups, although it was still much lower than that in the control group.31 Diabetes also adversely affects lipid profile, which may indirectly affect embryogenesis and organogenesis.56 Furthermore, combined disruption of normal glucose and lipid status may be detrimental to fetal and neonatal growth; some evidence indicates that these effects may be lifelong and contribute to adult obesity. In our studies, excess lipids accumulate in the serum of diabetic dams; however, less lipid accumulation (comparable to that observed in controls) was found in the serum of rat diabetic dams treated with resveratrol.31 We further analyzed the lipid profile and found that resveratrol significantly decreased the cholesterol and triglycerides concentrations in the serum of diabetic dams.31 Recently, resveratrol was shown to reduce mRNA levels for HMG-CoA reductase (a rate-limiting enzyme for cholesterol synthesis and target for ‘statin’ drugs) in high-fat-diet-fed hamsters.57 Statin molecules are used as drugs to lower the cholesterol and triglycerides in humans, but are not recommended for pregnant or nursing mothers.58,59 Therefore, resveratrol could be used as substitute for statins to control excess cholesterol production. Lipid profile is correlated with biologically active estrogen. So, the significant reduction in triglyceride level of diabetic female rat might be due to the ability of resveratrol to act on estrogen receptor signaling.60,61 However, whether resveratrol activity is estrogenic or antiestrogenic remains controversial.62,63

Antiteratogenic effect of resveratrol

An antiteratogenic effect of resveratrol has been demonstrated in pregnant mice (C57BL/6J) exposed to 2,3, 7,8-tetrachlorodibenzo-p-dioxin (TCDD). Pretreatment with resveratrol by gavage feeding (50 mg/kg body weight [bw]) from gestational day E8 to E13 significantly reduced the incidence and severity of fetal malformations caused by TCDD exposure (oral challenge, 14 μg/kg bw on E12).64 In addition, resveratrol inhibited TCDD-induced immunotoxicity in both the mother and fetus. Resveratrol protects normal non-pregnant mice and pregnant dams and their fetuses from TCDD-induced thymic atrophy and apoptosis, from alterations in the expression of T-cell receptor and co-stimulatory molecules, and from alterations in T-cell differentiation.65 In another study, the effect of dietary resveratrol on embryo-fetal survival and development was evaluated in groups of pregnant female Sprague–Dawley rats. Treatment with resveratrol with up to 750 mg/kg bw per day (from gestation day 5 to 20) was well tolerated with no signs of adverse effects on fetal weights, number of implantations, resorptions, live young or pre and post implantation losses. Maternal treatment with resveratrol at dietary doses up to 750 mg/kg bw/day had no effect on the incidence of major or minor fetal abnormalities or the incidence of visceral abnormalities. It was concluded that up to 750 mg/kg bw per day of resveratrol dose represented the no-observed-adverse-effect-level for maternal toxicity and embryo-fetal development.66 Recently, in a human clinical trial healthy subjects tolerated a 4.0 g resveratrol dose; however, at this high dose, diarrhea was observed in some participants.67 In another study from our lab, we showed that resveratrol exhibits neuroprotective effects in cerebellum by acting at redox regulating proteins in a rodent model of fetal alcohol spectrum disorders.68

Resveratrol prevents NTDs in diabetic pregnancies

Diabetic pregnancy can have many complications, especially in terms of embryonic development. Examples of embryonic impairments include improper or incomplete closure of the neural tube, developmental retardation, impaired rate of neurogenesis and failure to form appropriate neural connections.4,7 Furthermore, these developmental issues may be the basis of many future physical, neurological and psychiatric disorders. In humans, diabetic malformations most likely occur during the first trimester.69 In embryos of diabetic rat dams, embryonic day 12 is the stage where organogenesis is affected.70 We evaluated the outcome of diabetic pregnancy in E12 embryos after gavage feeding of resveratrol (100 mg/kg bw) to diabetic dams. No external malformations were observed in the embryos of control or control treated with resveratrol group (Figure 1a). Approximately 20% of the embryos of diabetic dams were malformed (Figures 1b and c). The malformed embryos were characterized as having defect in neural tube closing. Collectively, these malformed embryos were categorized as having malformations.71 The number of malformed embryos was significantly reduced when diabetic dams received resveratrol treatment (Figure 1c). Most embryos of diabetic dams, including those that lacked obvious malformations, had reduced weight, crown-rump length and somite number,31 and reduced embryo size indicating developmental delay. Resveratrol not only abolishes the diabetes-induced malformation in the embryos but it also rescues embryos from developmental delays.31 One way that resveratrol might be acting directly on embryos of diabetic pregnancies is by normalizing hyperglycemia-induced oxidative stress. Apoptosis subsequent to oxidative stress is associated with diabetic embryopathies.12 In embryos, resveratrol normalizes oxidative stress markers including the increased lipid peroxidation and total thiol concentrations and decreased amounts of reduced glutathione associated with hyperglycemia.31 Attenuation of oxidative stress further reduces the level of activated caspases, thereby reducing apoptosis, and in turn reduces embryonic malformations.31

Figure 1.

Figure 1

Resveratrol prevents neural tube defects in E12 rat embryos of diabetic dams. (a) Morphological analysis of E12 rat embryos of control (C), control treated with resveratrol (CR), diabetic (D) and diabetic treated with resveratrol (DR) dams; (b) E12 rat embryos of diabetic dams with neural tube defects; (c) Percentage of malformed embryos in C, CR, D and DR groups

Molecular mechanisms underlying the beneficial effects of resveratrol in diabetic embryopathy

Resveratrol crosses the blood–brain barrier and exerts protective effects against cerebral ischemic injury72 and also exerts potent antioxidant features in the brain of healthy rats.73 There have been no reports, however, on whether resveratrol crosses the placental barrier. Because resveratrol is beneficial against diabetes-induced embryonic malformation, we might cautiously assume that it does cross the placental barrier. Resveratrol works positively in different ways on mother and embryo in diabetic pregnancy to normalize the disease condition.31 Improvement in embryonic outcome might be because of improved maternal environment, direct effects on the embryos (as evident from normalized embryonic oxidative stress level), or both.31 Diabetes-dependent increases in oxidative stress and its fluctuation, and individual fetal antioxidative capacity determine the severity of embryonic deformity. Diabetic embryopathy studies revealed that the outcome of embryonic development depends on the glycemic, cholesterol and triglyceride levels and on the level of oxidative stress.12,31

We found that administration of resveratrol, a potent anti-oxidant74 in diabetic pregnancy works via three different ways by: (i) reducing excess maternal cholesterol and triglyceride, (ii) reducing maternal blood sugar level and (iii) modulating embryonic oxidative stress and apoptosis.31 Resveratrol is a free radical-scavenger and a potent antioxidant and can promote the activities of several antioxidative enzymes.75 Reactive oxygen species (ROS) are important intermediates in cellular signal transduction and transcriptional control but excess ROS generation leads to harmful oxidative modification of cellular lipids, proteins and nucleic acids. Indeed, diabetes-induced complications are associated with the increased amounts of ROS. In diabetic dams, with resveratrol treatment there were no changes in expression or activity of the antioxidative enzyme superoxide dismutase, but resveratrol reduced oxidative stress by normalizing the level of reduced glutathione, total thiol, lipid peroxidation and 4-hydroxy-2-non-enal (HNE) accumulation. Excess level of oxidative stress induces the formation of electrophilic aldehydes, such as HNE, as by-products of peroxidation of lipid membranes.76 HNE exerts its effects by alterations in cell proliferation, cell-cycle progression and apoptosis in various cell types.76,77

Diabetes impairs Rho GTPase signaling. Could resveratrol reverse impairments?

Rho GTPases (RhoA and Rac1), small-molecular weight G proteins of the Ras superfamily, have been implicated in cellular processes such as cytoskeletal rearrangement and cell-cycle control.7882 Active (GTP-bound) and inactive (GDP-bound) Rho conformations function as controls in a myriad of intracellular processes. Conformational cycling is regulated by three main classes of proteins: (i) those catalyzing exchange of GDP for GTP (e.g. guanosine nucleotide exchange factors);83 (ii) those stimulating hydrolysis of GTP to GDP (e.g. GTPase activating proteins [GAPs]);84 and (iii) those inhibiting the dissociation of GDP from GTPase (e.g. GDP dissociation inhibitors [GDIs]).85 Rho GTPases are also involved in neural tube closing8689 whereby RhoA accumulates at the apical region of neural plate to promote folding.87,90 Enrichment of RhoA at the apical region of the plate induces the constriction of the cells via Rho kinase. The resulting cytoskeletal rearrangement promotes the bending of the plate.90 G protein-coupled receptors such as Par1 and Par2 participate in neural tube closing. Genetic deletion of Par1/Par2 yields NTDs.91 These receptors function by coupling to heterotrimeric G proteins such as Go, Gi and Gz. Pertussis toxin, which inactivates these G proteins, reproduces the NTD-forming effects of Par1 and Par2 genetic. More importantly, Rac1 genetic deletion also produces exencephaly and spina bifida in mouse embryos.91

Rho GTPases play key roles in cytoskeletal rearrangement and neuronal differentiation. During normal development, activation of RhoA leads to formation of stress fibers and focal adhesion complexes.79 On the other hand, Rac1 activation participates in membrane ruffling and lamellipodia formation.81 Studies conducted in primary neurons suggest that Rho GTPases help to regulate the growth of axons and other neuronal processes. In general, Rac1 has a positive effect on neurite extension, whereas RhoA has a positive effect on neurite retraction.9294 Experimental evidence indicates that formation of synaptic connections in the neuronal network and their rearrangement are governed by the neuronal actin cytoskeleton.95,96 Actin assembly and polymerization as well as actomycin contraction are chiefly regulated by Rho GTPases.97,98 Thus, by controlling neuronal morphology, Rho GTPases might also be essential for learning and memory. Indeed, activation of Rho GTPases by CNF1 toxin induces cerebral actin cytoskeletal re-arrangement and improves learning and memory, as judged by various behavioral tests.99 Although many aspects of Rho signaling in the central nervous system are still unclear, the emerging details reveal that alterations or impairments in Rho GTPase signaling result in abnormal neuronal connectivity and deficient cognitive functioning in humans.100 Rho GTPases participate in retinoic acid (RA)-induced neuronal differentiation. Whereas activation of RhoA is involved in cytoskeletal rearrangement, Rac1 regulates induction of neurite outgrowth.80,82 After activation, Rho GTPases bind to their kinase effectors (ROCK-2 for RhoA and PAK1 for Rac1) and translocate to the plasma membrane.79,101 Oxidative stress activates RhoA; the ROS produced during oxidative stress react with the redox-sensitive cysteine residues in the phosphoryl-binding loop of RhoA, promoting its activation. This activation does not require the participation of exchange factors or other regulating proteins.102 In another study, ROS promoted the activation of RhoA by dissociating Rho GDI. The removal of ROS by the scavenger Tiron prevented the activation of RhoA in vivo.103 Whether hyperglycemia also activates RhoA by increasing oxidative stress and whether resveratrol prevents oxidative stress to neutralize activation of RhoA are not known.

Diabetes-induced impairments in the activation of Rho GTPase signaling may contribute to neuronal malformations in diabetic embryopathy. Resveratrol corrects the diabetes-induced impairment of retinoic acid receptor (RAR), retinoid X receptor (RXR) and mitogen-activated protein kinase (MAPKs) expression. All these signals play crucial roles during embryonic development. In addition, RARs (RARα/β/γ) and RXRs (RXRα/β/γ) function as transcription factors controlling RA-directed gene expression.104 RA (a physiological active metabolite of vitamin A) is necessary for proper embryonic development and rostrocaudal patterning of the nervous system.105 As heterodimers, RARs and RXRs bind to RA-response elements in DNA of promoters of target genes to regulate their transcription.104 Vitamin A deficiency results in suppression of RAR activity and contributes to embryonic skeleton hypoplasia.106 The curly tail-mouse mutant (a model for NTD) displays NTD because of downregulated RARβ and RARγ expression.107 Activation of RARs and RXRs has an anti-diabetic effect and helps to activate insulin-sensitizing genes.108 Suppressed expression of RAR and RXR receptors in E12 embryo of diabetic dams was improved with resveratrol treatment.109 MAPKs also contribute to growth, differentiation and developmental processes; their activity determines the fate of embryonic organogenesis when the embryo faces teratogenic conditions.110 Hyperglycemia increases ROS, which modulate MAPK signaling.111 In the embryo, oxidative stress results in DNA, protein and lipid modifications that disrupt normal development. Resveratrol can also selectively inhibit stress activated MAPK signaling.112 Increased phospho-ERK1/2 and decreased phospho-JNK1/2 and phospho-p38 occurs in the embryos of resveratrol-treated diabetic dams compared with those not exposed to resveratrol.109

After reviewing the literature on resveratrol, and on the basis of our own studies, we conclude that resveratrol possesses significant blood sugar-lowering activity and anti-oxidative capacity without any observable side-effects31,45 and prevents embryonic oxidative stress and apoptosis associated with diabetic embryopathy. Thus, resveratrol may be suitable alone or in combination with insulin and folic acid for use during diabetic pregnancies and for nursing mothers. Animal studies to test whether resveratrol supplementation in combination with folic acid alone and/or with insulin can prevent all embryonic malformations and developmental delays are needed to provide data to support clinical trials in humans.

ACKNOWLEDGEMENTS

This research was supported in part by NIH R21AA016121 (US) and Health Sciences Distinguished Professorship (DJD).

Footnotes

Author contributions: CKS and USS designed the study; CKS conducted the study; AK contributed to the study design; CKS, HAL and USS wrote the manuscript; CKS, DD and USS were involved in the data analyses and interpretation and review of the manuscript.

REFERENCES

  • 1.Polonsky KS. The past 200 years in diabetes. N Eng J Med. 2012;367:1332–40. doi: 10.1056/NEJMra1110560. [DOI] [PubMed] [Google Scholar]
  • 2.Evensen AE. Update on gestational diabetes mellitus. Prim Care. 2012;39:83–94. doi: 10.1016/j.pop.2011.11.011. [DOI] [PubMed] [Google Scholar]
  • 3.Centers for Disease Control and Prevention . National Diabetes Fact Sheet: National Estimates and General Information on Diabetes and Prediabetes in the United States, 2011. US Department of Health and Human Services, Centers for Disease Control and Prevention; Atlanta, GA: 2011. [Google Scholar]
  • 4.Becerra JE, Khoury MJ, Cordero JF, Erickson JD. Diabetes mellitus during pregnancy and the risks for specific birth defects: a population-based case-control study. Pediatrics. 1990;85:1–9. [PubMed] [Google Scholar]
  • 5.Mills JL. Malformations in infants of diabetic mothers. Teratology. 1982;25:385–94. doi: 10.1002/tera.1420250316. [DOI] [PubMed] [Google Scholar]
  • 6.Phelan SA, Ito M, Loeken MR. Neural tube defects in embryos of diabetic mice: role of the Pax-3 gene and apoptosis. Diabetes. 1997;46:1189–97. doi: 10.2337/diab.46.7.1189. [DOI] [PubMed] [Google Scholar]
  • 7.Cederberg J, Picard JJ, Eriksson UJ. Maternal diabetes in the rat impairs the formation of neural-crest derived cranial nerve ganglia in the offspring. Diabetologia. 2003;46:1245–51. doi: 10.1007/s00125-003-1100-1. [DOI] [PubMed] [Google Scholar]
  • 8.Pedersen LM, Tygstrup I, Pedersen J. Congenital malformations in newborn Infants of diabetic women. Correlation with maternal diabetic vascular complications. Lancet. 1964;1:1124–26. doi: 10.1016/s0140-6736(64)91805-7. [DOI] [PubMed] [Google Scholar]
  • 9.Zhao Z, Reece EA. Experimental mechanisms of diabetic embryopathy and strategies for developing therapeutic interventions. J Soc Gynecol Investig. 2005;12:549–57. doi: 10.1016/j.jsgi.2005.07.005. [DOI] [PubMed] [Google Scholar]
  • 10.Eriksson UJ, Cederberg J, Wentzel P. Congenital malformations in offspring of diabetic mothers – animal and human studies. Rev Endocr Metab Disord. 2003;4:79–93. doi: 10.1023/a:1021879504372. [DOI] [PubMed] [Google Scholar]
  • 11.Salbaum JM, Kappen C. Neural tube defect genes and maternal diabetes during pregnancy. Birth Defects Res A Clin Mol Teratol. 2010;88:601–11. doi: 10.1002/bdra.20680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Ornoy A. Embryonic oxidative stress as a mechanism of teratogenesis with special emphasis on diabetic embryopathy. Reprod Toxicol. 2007;24:31–41. doi: 10.1016/j.reprotox.2007.04.004. [DOI] [PubMed] [Google Scholar]
  • 13.Zabihi S, Wentzel P, Eriksson UJ. Maternal blood glucose levels determine the severity of diabetic embryopathy in mice with different expression of copper-zinc superoxide dismutase (CuZnSOD) Toxicol Sci. 2008;105:166–72. doi: 10.1093/toxsci/kfn101. [DOI] [PubMed] [Google Scholar]
  • 14.Yang P, Zhao Z, Reece EA. Involvement of c-Jun N-terminal kinases activation in diabetic embryopathy. Biochem Biophys Res Commun. 2007;357:749–54. doi: 10.1016/j.bbrc.2007.04.023. [DOI] [PubMed] [Google Scholar]
  • 15.Soler NG, Walsh CH, Malins JM. Congenital malformations in infants of diabetic mothers. Q J Med. 1976;45:303–13. [PubMed] [Google Scholar]
  • 16.Zabihi S, Loeken MR. Understanding diabetic teratogenesis: where are we now and where are we going? Birth Defects Res A Clin Mol Teratol. 2010;88:779–90. doi: 10.1002/bdra.20704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Evers IM, de Valk HW, Visser GH. Risk of complications of pregnancy in women with type 1 diabetes: nationwide prospective study in the Netherlands. Br Med J. 2004;328:915. doi: 10.1136/bmj.38043.583160.EE. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Suhonen L, Hiilesmaa V, Teramo K. Glycaemic control during early pregnancy and fetal malformations in women with type I diabetes mellitus. Diabetologia. 2000;43:79–82. doi: 10.1007/s001250050010. [DOI] [PubMed] [Google Scholar]
  • 19.Loeken MR. Challenges in understanding diabetic embryopathy. Diabetes. 2008;57:3187–8. doi: 10.2337/db08-1201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Mills JL, Signore CC. Folic acid and the prevention of neural-tube defects. N Engl J Med. 2004;350:2209–11. doi: 10.1056/NEJM200405203502120. [DOI] [PubMed] [Google Scholar]
  • 21.Gareskog M, Eriksson UJ, Wentzel P. Combined supplementation of folic acid and vitamin E diminishes diabetes-induced embryotoxicity in rats. Birth Defects Res A Clin Mol Teratol. 2006;76:483–90. doi: 10.1002/bdra.20278. [DOI] [PubMed] [Google Scholar]
  • 22.Wentzel P, Gareskog M, Eriksson UJ. Folic acid supplementation diminishes diabetes- and glucose-induced dysmorphogenesis in rat embryos in vivo and in vitro. Diabetes. 2005;54:546–53. doi: 10.2337/diabetes.54.2.546. [DOI] [PubMed] [Google Scholar]
  • 23.Mills JL, Signore C. Neural tube defect rates before and after food fortification with folic acid. Birth Defects Res A Clin Mol Teratol. 2004;70:844–45. doi: 10.1002/bdra.20075. [DOI] [PubMed] [Google Scholar]
  • 24.Wolff T, Witkop CT, Miller T, Syed SB. Folic acid supplementation for the prevention of neural tube defects: an update of the evidence for the US Preventive Services Task Force. Ann Intern Med. 2009;150:632–39. doi: 10.7326/0003-4819-150-9-200905050-00010. [DOI] [PubMed] [Google Scholar]
  • 25.Banhidy F, Acs N, Puho EH, Czeizel AE. Congenital abnormalities in the offspring of pregnant women with type 1, type 2 and gestational diabetes mellitus: a population-based case-control study. Congenit Anom (Kyoto) 2010;50:115–21. doi: 10.1111/j.1741-4520.2010.00275.x. [DOI] [PubMed] [Google Scholar]
  • 26.Wilson RD, Davies G, Desilets V, Reid GJ, Summers A, Wyatt P, Young D. The use of folic acid for the prevention of neural tube defects and other congenital anomalies. J Obstet Gynaecol Can. 2003;25:959–73. doi: 10.1016/s1701-2163(16)30248-1. [DOI] [PubMed] [Google Scholar]
  • 27.Kim KC, Friso S, Choi SW. DNA methylation, an epigenetic mechanism connecting folate to healthy embryonic development and aging. J Nutr Biochem. 2009;20:917–26. doi: 10.1016/j.jnutbio.2009.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Rogers EJ. Has enhanced folate status during pregnancy altered natural selection and possibly Autism prevalence? A closer look at a possible link. Med Hypotheses. 2008;71:406–10. doi: 10.1016/j.mehy.2008.04.013. [DOI] [PubMed] [Google Scholar]
  • 29.Callejon G, Mayor-Olea A, Jimenez AJ, Gaitan MJ, Palomares AR, Martinez F, Ruiz M, Reyes-Engel A. Genotypes of the C677T and A1298C polymorphisms of the MTHFR gene as a cause of human spontaneous embryo loss. Hum Reprod. 2007;22:3249–54. doi: 10.1093/humrep/dem337. [DOI] [PubMed] [Google Scholar]
  • 30.Do GM, Jung UJ, Park HJ, Kwon EY, Jeon SM, McGregor RA, Choi MS. Resveratrol ameliorates diabetes-related metabolic changes via activation of AMP-activated protein kinase and its downstream targets in db/db mice. Mol Nutr Food Res. 2012;56:1282–91. doi: 10.1002/mnfr.201200067. [DOI] [PubMed] [Google Scholar]
  • 31.Singh CK, Kumar A, Hitchcock DB, Fan D, Goodwin R, Lavoie HA, Nagarkatti P, Dipette DJ, Singh US. Resveratrol prevents embryonic oxidative stress and apoptosis associated with diabetic embryopathy and improves glucose and lipid profile of diabetic dam. Mol Nutr Food Res. 2011;55:1186–96. doi: 10.1002/mnfr.201000457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Yang F, Zhang T, Ito Y. Large-scale separation of resveratrol, anthraglycoside A and anthraglycoside B from Polygonum cuspidatum Sieb. et Zucc by high-speed counter-current chromatography. J Chromatogr A. 2001;919:443–48. doi: 10.1016/s0021-9673(01)00846-9. [DOI] [PubMed] [Google Scholar]
  • 33.Magyar K, Halmosi R, Palfi A, Feher G, Czopf L, Fulop A, Battyany I, Sumegi B, Toth K, Szabados E. Cardioprotection by resveratrol: a human clinical trial in patients with stable coronary artery disease. Clin Hemorheol Microcirc. 2012;50:179–87. doi: 10.3233/CH-2011-1424. [DOI] [PubMed] [Google Scholar]
  • 34.Bishayee A, Politis T, Darvesh AS. Resveratrol in the chemoprevention and treatment of hepatocellular carcinoma. Cancer Treat Rev. 2010;36:43–53. doi: 10.1016/j.ctrv.2009.10.002. [DOI] [PubMed] [Google Scholar]
  • 35.Fulda S. Resveratrol and derivatives for the prevention and treatment of cancer. Drug Discov Today. 2010;15:757–65. doi: 10.1016/j.drudis.2010.07.005. [DOI] [PubMed] [Google Scholar]
  • 36.Howitz KT, Bitterman KJ, Cohen HY, Lamming DW, Lavu S, Wood JG, Zipkin RE, Chung P, Kisielewski A, Zhang LL, Scherer B, Sinclair DA. Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature. 2003;425:191–96. doi: 10.1038/nature01960. [DOI] [PubMed] [Google Scholar]
  • 37.Valenzano DR, Cellerino A. Resveratrol and the pharmacology of aging: a new vertebrate model to validate an old molecule. Cell Cycle. 2006;5:1027–32. doi: 10.4161/cc.5.10.2739. [DOI] [PubMed] [Google Scholar]
  • 38.Paul B, Masih I, Deopujari J, Charpentier C. Occurrence of resveratrol and pterostilbene in age-old darakchasava, an ayurvedic medicine from India. J Ethnopharmacol. 1999;68:71–76. doi: 10.1016/s0378-8741(99)00044-6. [DOI] [PubMed] [Google Scholar]
  • 39.Ramadori G, Gautron L, Fujikawa T, Vianna CR, Elmquist JK, Coppari R. Central administration of resveratrol improves diet-induced diabetes. Endocrinology. 2009;150:5326–33. doi: 10.1210/en.2009-0528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Milne JC, Lambert PD, Schenk S, Carney DP, Smith JJ, Gagne DJ, Jin L, Boss O, Perni RB, Vu CB, Bemis JE, Xie R, Disch JS, Ng PY, Nunes JJ, Lynch AV, Yang H, Galonek H, Israelian K, Choy W, Iffland A, Lavu S, Medvedik O, Sinclair DA, Olefsky JM, Jirousek MR, Elliott PJ, Westphal CH. Small molecule activators of SIRT1 as therapeutics for the treatment of type 2 diabetes. Nature. 2007;450:712–16. doi: 10.1038/nature06261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Sharma S, Misra CS, Arumugam S, Roy S, Shah V, Davis JA, Shirumalla RK, Ray A. Antidiabetic activity of resveratrol, a known SIRT1 activator in a genetic model for type-2 diabetes. Phytother Res. 2011;25:67–73. doi: 10.1002/ptr.3221. [DOI] [PubMed] [Google Scholar]
  • 42.Palsamy P, Subramanian S. Resveratrol, a natural phytoalexin, normalizes hyperglycemia in streptozotocin-nicotinamide induced experimental diabetic rats. Biomed Pharmacother. 2008;62:598–605. doi: 10.1016/j.biopha.2008.06.037. [DOI] [PubMed] [Google Scholar]
  • 43.Baur JA, Pearson KJ, Price NL, Jamieson HA, Lerin C, Kalra A, Prabhu VV, Allard JS, Lopez-Lluch G, Lewis K, Pistell PJ, Poosala S, Becker KG, Boss O, Gwinn D, Wang M, Ramaswamy S, Fishbein KW, Spencer RG, Lakatta EG, Le Couteur D, Shaw RJ, Navas P, Puigserver P, Ingram DK, de Cabo R, Sinclair DA. Resveratrol improves health and survival of mice on a high-calorie diet. Nature. 2006;444:337–42. doi: 10.1038/nature05354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Um JH, Park SJ, Kang H, Yang S, Foretz M, McBurney MW, Kim MK, Viollet B, Chung JH. AMP-activated protein kinase-deficient mice are resistant to the metabolic effects of resveratrol. Diabetes. 2010;59:554–63. doi: 10.2337/db09-0482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Cottart CH, Nivet-Antoine V, Laguillier-Morizot C, Beaudeux JL. Resveratrol bioavailability and toxicity in humans. Mol Nutr Food Res. 2010;54:7–16. doi: 10.1002/mnfr.200900437. [DOI] [PubMed] [Google Scholar]
  • 46.Walle T, Hsieh F, DeLegge MH, Oatis JE, Jr, Walle UK. High absorption but very low bioavailability of oral resveratrol in humans. Drug Metab Dispos. 2004;32:1377–82. doi: 10.1124/dmd.104.000885. [DOI] [PubMed] [Google Scholar]
  • 47.Vaz-da-Silva M, Loureiro AI, Falcao A, Nunes T, Rocha JF, Fernandes-Lopes C, Soares E, Wright L, Almeida L, Soares-da-Silva P. Effect of food on the pharmacokinetic profile of trans-resveratrol. Int J Clin Pharmacol Ther. 2008;46:564–70. doi: 10.5414/cpp46564. [DOI] [PubMed] [Google Scholar]
  • 48.Howells LM, Berry DP, Elliott PJ, Jacobson EW, Hoffmann E, Hegarty B, Brown K, Steward WP, Gescher AJ. Phase I randomized, double-blind pilot study of micronized resveratrol (SRT501) in patients with hepatic metastases – safety, pharmacokinetics, and pharmacodynamics. Cancer Prevention Res. 2011;4:1419–25. doi: 10.1158/1940-6207.CAPR-11-0148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Narayanan NK, Nargi D, Randolph C, Narayanan BA. Liposome encapsulation of curcumin and resveratrol in combination reduces prostate cancer incidence in PTEN knockout mice. Int J Cancer. 2009;125:1–8. doi: 10.1002/ijc.24336. [DOI] [PubMed] [Google Scholar]
  • 50.Johnson JJ, Nihal M, Siddiqui IA, Scarlett CO, Bailey HH, Mukhtar H, Ahmad N. Enhancing the bioavailability of resveratrol by combining it with piperine. Mol Nutr Food Res. 2011;55:1169–76. doi: 10.1002/mnfr.201100117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Patel KR, Scott E, Brown VA, Gescher AJ, Steward WP, Brown K. Clinical trials of resveratrol. Ann NY Acad Sci. 2011;1215:161–69. doi: 10.1111/j.1749-6632.2010.05853.x. [DOI] [PubMed] [Google Scholar]
  • 52.Smoliga JM, Baur JA, Hausenblas HA. Resveratrol and health – a comprehensive review of human clinical trials. Mol Nutr Food Res. 2011;55:1129–41. doi: 10.1002/mnfr.201100143. [DOI] [PubMed] [Google Scholar]
  • 53.Fujitaka K, Otani H, Jo F, Jo H, Nomura E, Iwasaki M, Nishikawa M, Iwasaka T, Das DK. Modified resveratrol Longevinex improves endothelial function in adults with metabolic syndrome receiving standard treatment. Nutr Res. 2011;31:842–47. doi: 10.1016/j.nutres.2011.09.028. [DOI] [PubMed] [Google Scholar]
  • 54.Zhang H, Zhang J, Ungvari Z, Zhang C. Resveratrol improves endothelial function: role of TNF-α and vascular oxidative stress. Arterioscler Thromb Vasc Biol. 2009;29:1164–71. doi: 10.1161/ATVBAHA.109.187146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Penumathsa SV, Thirunavukkarasu M, Zhan L, Maulik G, Menon VP, Bagchi D, Maulik N. Resveratrol enhances GLUT-4 translocation to the caveolar lipid raft fractions through AMPK/Akt/eNOS signalling pathway in diabetic myocardium. J Cell Mol Med. 2008;12:2350–61. doi: 10.1111/j.1582-4934.2008.00251.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Herrera E, Ortega-Senovilla H. Disturbances in lipid metabolism in diabetic pregnancy – Are these the cause of the problem? Best Pract Res Clin Endocrinol Metab. 2010;24:515–25. doi: 10.1016/j.beem.2010.05.006. [DOI] [PubMed] [Google Scholar]
  • 57.Cho IJ, Ahn JY, Kim S, Choi MS, Ha TY. Resveratrol attenuates the expression of HMG-CoA reductase mRNA in hamsters. Biochem Biophys Res Commun. 2008;367:190–94. doi: 10.1016/j.bbrc.2007.12.140. [DOI] [PubMed] [Google Scholar]
  • 58.Ofori B, Rey E, Berard A. Risk of congenital anomalies in pregnant users of statin drugs. Br J Clin Pharmacol. 2007;64:496–509. doi: 10.1111/j.1365-2125.2007.02905.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Statins: beware during pregnancy. Prescrire Int. 2006;15:18–19. [PubMed] [Google Scholar]
  • 60.Deng JY, Hsieh PS, Huang JP, Lu LS, Hung LM. Activation of estrogen receptor is crucial for resveratrol-stimulating muscular glucose uptake via both insulin-dependent and -independent pathways. Diabetes. 2008;57:1814–23. doi: 10.2337/db07-1750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Shearer GC, Joles JA, Jones H, Walzem RL, Kaysen GA. Estrogen effects on triglyceride metabolism in analbuminemic rats. Kidney Int. 2000;57:2268–74. doi: 10.1046/j.1523-1755.2000.00087.x. [DOI] [PubMed] [Google Scholar]
  • 62.Basly JP, Marre-Fournier F, Le Bail JC, Habrioux G, Chulia AJ. Estrogenic/antiestrogenic and scavenging properties of (E)- and (Z)-resveratrol. Life Sci. 2000;66:769–77. doi: 10.1016/s0024-3205(99)00650-5. [DOI] [PubMed] [Google Scholar]
  • 63.Gehm BD, McAndrews JM, Chien PY, Jameson JL. Resveratrol, a polyphenolic compound found in grapes and wine, is an agonist for the estrogen receptor. Proc Natl Acad Sci USA. 1997;94:14138–43. doi: 10.1073/pnas.94.25.14138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Jang JY, Park D, Shin S, Jeon JH, Choi BI, Joo SS, Hwang SY, Nahm SS, Kim YB. Antiteratogenic effect of resveratrol in mice exposed in utero to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Eur J Pharmacol. 2008;591:280–83. doi: 10.1016/j.ejphar.2008.05.033. [DOI] [PubMed] [Google Scholar]
  • 65.Singh NP, Singh US, Nagarkatti M, Nagarkatti PS. Resveratrol (3,5,4′-trihydroxystilbene) protects pregnant mother and fetus from the immunotoxic effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin. Mol Nutr Food Res. 2011;55:209–19. doi: 10.1002/mnfr.201000206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Williams LD, Burdock GA, Edwards JA, Beck M, Bausch J. Safety studies conducted on high-purity trans-resveratrol in experimental animals. Food Chem Toxicol. 2009;47:2170–82. doi: 10.1016/j.fct.2009.06.002. [DOI] [PubMed] [Google Scholar]
  • 67.Patel KR, Brown VA, Jones DJ, Britton RG, Hemingway D, Miller AS, West KP, Booth TD, Perloff M, Crowell JA, Brenner DE, Steward WP, Gescher AJ, Brown K. Clinical pharmacology of resveratrol and its metabolites in colorectal cancer patients. Cancer Res. 2010;70:7392–99. doi: 10.1158/0008-5472.CAN-10-2027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Kumar A, Singh CK, LaVoie HA, Dipette DJ, Singh US. Resveratrol restores Nrf2 level and prevents ethanol-induced toxic effects in the cerebellum of a rodent model of fetal alcohol spectrum disorders. Mol Pharmacol. 2011;80:446–57. doi: 10.1124/mol.111.071126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Nielsen GL, Norgard B, Puho E, Rothman KJ, Sorensen HT, Czeizel AE. Risk of specific congenital abnormalities in offspring of women with diabetes. Diabet Med. 2005;22:693–96. doi: 10.1111/j.1464-5491.2005.01477.x. [DOI] [PubMed] [Google Scholar]
  • 70.Reece EA, Wu YK. Prevention of diabetic embryopathy in offspring of diabetic rats with use of a cocktail of deficient substrates and an antioxidant. Am J Obstet Gynecol. 1997;176:790–97. doi: 10.1016/s0002-9378(97)70602-1. [DOI] [PubMed] [Google Scholar]
  • 71.Zabihi S, Wentzel P, Eriksson UJ. Altered uterine perfusion is involved in fetal outcome of diabetic rats. Placenta. 2008;29:413–21. doi: 10.1016/j.placenta.2008.02.005. [DOI] [PubMed] [Google Scholar]
  • 72.Wang Q, Xu J, Rottinghaus GE, Simonyi A, Lubahn D, Sun GY, Sun AY. Resveratrol protects against global cerebral ischemic injury in gerbils. Brain Res. 2002;958:439–47. doi: 10.1016/s0006-8993(02)03543-6. [DOI] [PubMed] [Google Scholar]
  • 73.Mokni M, Elkahoui S, Limam F, Amri M, Aouani E. Effect of resveratrol on antioxidant enzyme activities in the brain of healthy rat. Neurochem Res. 2007;32:981–87. doi: 10.1007/s11064-006-9255-z. [DOI] [PubMed] [Google Scholar]
  • 74.Ates O, Cayli SR, Yucel N, Altinoz E, Kocak A, Durak MA, Turkoz Y, Yologlu S. Central nervous system protection by resveratrol in streptozotocin-induced diabetic rats. J Clin Neurosci. 2007;14:256–60. doi: 10.1016/j.jocn.2005.12.010. [DOI] [PubMed] [Google Scholar]
  • 75.Venturini CD, Merlo S, Souto AA, Fernandes Mda C, Gomez R, Rhoden CR. Resveratrol and red wine function as antioxidants in the nervous system without cellular proliferative effects during experimental diabetes. Oxid Med Cell Longev. 2010;3:434–41. doi: 10.4161/oxim.3.6.14741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Kruman I, Bruce-Keller AJ, Bredesen D, Waeg G, Mattson MP. Evidence that 4-hydroxynonenal mediates oxidative stress-induced neuronal apoptosis. J Neurosci. 1997;17:5089–100. doi: 10.1523/JNEUROSCI.17-13-05089.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Ong WY, Lu XR, Hu CY, Halliwell B. Distribution of hydroxynonenal-modified proteins in the kainate-lesioned rat hippocampus: evidence that hydroxynonenal formation precedes neuronal cell death. Free Radic Biol Med. 2000;28:1214–21. doi: 10.1016/s0891-5849(00)00238-0. [DOI] [PubMed] [Google Scholar]
  • 78.Etienne-Manneville S, Hall A. Rho GTPases in cell biology. Nature. 2002;420:629–35. doi: 10.1038/nature01148. [DOI] [PubMed] [Google Scholar]
  • 79.Singh US, Kunar MT, Kao YL, Baker KM. Role of transglutaminase II in retinoic acid-induced activation of RhoA- associated kinase-2. Embo J. 2001;20:2413–23. doi: 10.1093/emboj/20.10.2413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Singh US, Pan J, Kao YL, Joshi S, Young KL, Baker KM. Tissue transglutaminase mediates activation of RhoA and MAP kinase pathways during retinoic acid-induced neuronal differentiation of SH-SY5Y cells. J Biol Chem. 2003;278:391–99. doi: 10.1074/jbc.M206361200. [DOI] [PubMed] [Google Scholar]
  • 81.Nobes CD, Hawkins P, Stephens L, Hall A. Activation of the small GTP-binding proteins rho and rac by growth factor receptors. J Cell Sci. 1995;108(Part 1):225–33. doi: 10.1242/jcs.108.1.225. [DOI] [PubMed] [Google Scholar]
  • 82.Pan J, Kao YL, Joshi S, Jeetendran S, Dipette D, Singh US. Activation of Rac1 by phosphatidylinositol 3-kinase in vivo: role in activation of mitogen-activated protein kinase (MAPK) pathways and retinoic acid-induced neuronal differentiation of SH-SY5Y cells. J Neurochem. 2005;93:571–83. doi: 10.1111/j.1471-4159.2005.03106.x. [DOI] [PubMed] [Google Scholar]
  • 83.Scita G, Tenca P, Frittoli E, Tocchetti A, Innocenti M, Giardina G, Di Fiore PP. Signaling from Ras to Rac and beyond: not just a matter of GEFs. EMBO J. 2000;19:2393–98. doi: 10.1093/emboj/19.11.2393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Sprang SR. G proteins, effectors and GAPs: structure and mechanism. Curr Opin Struct Biol. 1997;7:849–56. doi: 10.1016/s0959-440x(97)80157-1. [DOI] [PubMed] [Google Scholar]
  • 85.Hart MJ, Maru Y, Leonard D, Witte ON, Evans T, Cerione RA. A GDP dissociation inhibitor that serves as a GTPase inhibitor for the Ras-like protein CDC42Hs. Science. 1992;258:812–15. doi: 10.1126/science.1439791. [DOI] [PubMed] [Google Scholar]
  • 86.Brouns MR, Matheson SF, Hu KQ, Delalle I, Caviness VS, Silver J, Bronson RT, Settleman J. The adhesion signaling molecule p190 RhoGAP is required for morphogenetic processes in neural development. Development. 2000;127:4891–903. doi: 10.1242/dev.127.22.4891. [DOI] [PubMed] [Google Scholar]
  • 87.Ybot-Gonzalez P, Savery D, Gerrelli D, Signore M, Mitchell CE, Faux CH, Greene ND, Copp AJ. Convergent extension, planar-cell-polarity signalling and initiation of mouse neural tube closure. Development. 2007;134:789–99. doi: 10.1242/dev.000380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Lindqvist M, Horn Z, Bryja V, Schulte G, Papachristou P, Ajima R, Dyberg C, Arenas E, Yamaguchi TP, Lagercrantz H, Ringstedt T. Vang-like protein 2 and Rac1 interact to regulate adherens junctions. J Cell Sci. 2010;123:472–83. doi: 10.1242/jcs.048074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Suzuki M, Morita H, Ueno N. Molecular mechanisms of cell shape changes that contribute to vertebrate neural tube closure. Dev Growth Differ. 2012;54:266–76. doi: 10.1111/j.1440-169X.2012.01346.x. [DOI] [PubMed] [Google Scholar]
  • 90.Kinoshita N, Sasai N, Misaki K, Yonemura S. Apical accumulation of Rho in the neural plate is important for neural plate cell shape change and neural tube formation. Mol Biol Cell. 2008;19:2289–99. doi: 10.1091/mbc.E07-12-1286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Camerer E, Barker A, Duong DN, Ganesan R, Kataoka H, Cornelissen I, Darragh MR, Hussain A, Zheng YW, Srinivasan Y, Brown C, Xu SM, Regard JB, Lin CY, Craik CS, Kirchhofer D, Coughlin SR. Local protease signaling contributes to neural tube closure in the mouse embryo. Dev Cell. 2010;18:25–38. doi: 10.1016/j.devcel.2009.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Jalink K, van Corven EJ, Hengeveld T, Morii N, Narumiya S, Moolenaar WH. Inhibition of lysophosphatidate- and thrombin-induced neurite retraction and neuronal cell rounding by ADP ribosylation of the small GTP-binding protein Rho. J Cell Biol. 1994;126:801–10. doi: 10.1083/jcb.126.3.801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Kozma R, Sarner S, Ahmed S, Lim L. Rho family GTPases and neuronal growth cone remodelling: relationship between increased complexity induced by Cdc42Hs, Rac1, and acetylcholine and collapse induced by RhoA and lysophosphatidic acid. Mol Cell Biol. 1997;17:1201–11. doi: 10.1128/mcb.17.3.1201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Leeuwen FN, Kain HE, Kammen RA, Michiels F, Kranenburg OW, Collard JG. The guanine nucleotide exchange factor Tiam1 affects neuronal morphology; opposing roles for the small GTPases Rac and Rho. J Cell Biol. 1997;139:797–807. doi: 10.1083/jcb.139.3.797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Fukazawa Y, Saitoh Y, Ozawa F, Ohta Y, Mizuno K, Inokuchi K. Hippocampal LTP is accompanied by enhanced F-actin content within the dendritic spine that is essential for late LTP maintenance in vivo. Neuron. 2003;38:447–60. doi: 10.1016/s0896-6273(03)00206-x. [DOI] [PubMed] [Google Scholar]
  • 96.Zito K, Knott G, Shepherd GM, Shenolikar S, Svoboda K. Induction of spine growth and synapse formation by regulation of the spine actin cytoskeleton. Neuron. 2004;44:321–34. doi: 10.1016/j.neuron.2004.09.022. [DOI] [PubMed] [Google Scholar]
  • 97.Luo L. Rho GTPases in neuronal morphogenesis. Nat Rev Neurosci. 2000;1:173–80. doi: 10.1038/35044547. [DOI] [PubMed] [Google Scholar]
  • 98.Luo L. Actin cytoskeleton regulation in neuronal morphogenesis and structural plasticity. Annu Rev Cell Dev Biol. 2002;18:601–35. doi: 10.1146/annurev.cellbio.18.031802.150501. [DOI] [PubMed] [Google Scholar]
  • 99.Diana G, Valentini G, Travaglione S, Falzano L, Pieri M, Zona C, Meschini S, Fabbri A, Fiorentini C. Enhancement of learning and memory after activation of cerebral Rho GTPases. Proc Natl Acad Sci USA. 2007;104:636–41. doi: 10.1073/pnas.0610059104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Ramakers GJ. Rho proteins, mental retardation and the cellular basis of cognition. Trends Neurosci. 2002;25:191–99. doi: 10.1016/s0166-2236(00)02118-4. [DOI] [PubMed] [Google Scholar]
  • 101.Sin WC, Chen XQ, Leung T, Lim L. RhoA-binding kinase alpha translocation is facilitated by the collapse of the vimentin intermediate filament network. Mol Cell Biol. 1998;18:6325–39. doi: 10.1128/mcb.18.11.6325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Aghajanian A, Wittchen ES, Campbell SL, Burridge K. Direct activation of RhoA by reactive oxygen species requires a redox-sensitive motif. PLoS One. 2009;4:e8045. doi: 10.1371/journal.pone.0008045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Kondrikov D, Caldwell RB, Dong Z, Su Y. Reactive oxygen species-dependent RhoA activation mediates collagen synthesis in hyperoxic lung fibrosis. Free Radic Biol Med. 2011;50:1689–98. doi: 10.1016/j.freeradbiomed.2011.03.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Mark M, Ghyselinck NB, Chambon P. Function of retinoic acid receptors during embryonic development. Nucl Recept Signal. 2009;7:e002. doi: 10.1621/nrs.07002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Wilson L, Gale E, Maden M. The role of retinoic acid in the morphogenesis of the neural tube. J Anat. 2003;203:357–68. doi: 10.1046/j.1469-7580.2003.00230.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Li N, Sun S, Wang D, Yao P, Yang X, Yan H, Du Y, Ying C, Liu L. Suppression of retinoic acid receptors may contribute to embryonic skeleton hypoplasia in maternal rats with chronic vitamin A deficiency. J Nutr Biochem. 2010;21:710–16. doi: 10.1016/j.jnutbio.2009.04.011. [DOI] [PubMed] [Google Scholar]
  • 107.Chen WH, Morriss-Kay GM, Copp AJ. Genesis and prevention of spinal neural tube defects in the curly tail mutant mouse: involvement of retinoic acid and its nuclear receptors RAR-beta and RAR-gamma. Development. 1995;121:681–91. doi: 10.1242/dev.121.3.681. [DOI] [PubMed] [Google Scholar]
  • 108.Lenhard JM. PPAR gamma/RXR as a molecular target for diabetes. Recept Channel. 2001;7:249–58. [PubMed] [Google Scholar]
  • 109.Singh CK, Kumar A, LaVoie HA, Dipette DJ, Singh US. Resveratrol prevents impairment in activation of retinoic acid receptors and MAP kinases in the embryos of a rodent model of diabetic embryopathy. Reprod Sci. 2012;19:949–61. doi: 10.1177/1933719112438972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Yan J, Hales BF. p38 and c-Jun N-terminal kinase mitogen-activated protein kinase signaling pathways play distinct roles in the response of organogenesis-stage embryos to a teratogen. J Pharmacol Exp Ther. 2008;326:764–72. doi: 10.1124/jpet.108.139907. [DOI] [PubMed] [Google Scholar]
  • 111.Torres M, Forman HJ. Redox signaling and the MAP kinase pathways. Biofactors. 2003;17:287–96. doi: 10.1002/biof.5520170128. [DOI] [PubMed] [Google Scholar]
  • 112.Malemud CJ. Inhibitors of stress-activated protein/mitogen-activated protein kinase pathways. Curr Opin Pharmacol. 2007;7:339–43. doi: 10.1016/j.coph.2006.11.012. [DOI] [PubMed] [Google Scholar]

RESOURCES