Planta Med 2022; 88(08): 650-663
DOI: 10.1055/a-1482-2343
Biological and Pharmacological Activity
Reviews

Prophylactic Use of Natural Products against Developmentally Programmed Metabolic Syndrome

Kasimu Ghandi Ibrahim
1   Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
2   Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
,
Kehinde Ahmad Adeshina
1   Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
2   Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
,
Muhammad Bashir Bello
2   Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
3   Department of Veterinary Microbiology, Faculty of Veterinary Medicine, Usmanu Danfodiyo University, Sokoto, Nigeria
,
Ibrahim Malami
2   Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
4   Department of Pharmacognosy and Ethnopharmacy, Faculty of Pharmaceutical Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
,
Bilyaminu Abubakar
2   Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
5   Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
,
Murtala Bello Abubakar
1   Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
2   Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
,
Mustapha Umar Imam
2   Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
6   Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
› Author Affiliations
Supported by: Institutional-based research (IBR) grant of the Tertiary Education Trust Fund (TETFUND) of Nigeria awarded to Usmanu Danfodiyo University, Sokoto TETFUND/DR&D/CE/UNIV/SOKOTO/RP/VOL.1
 

Abstract

Parental dietary choices and/or nutritional interventions in the offspring are critical to early life development, especially during the periods of active developmental plasticity in the offspring. Exposure to a high-fructose, high-fat diet during the fetal or neonatal period predisposes the affected individuals to the development of one or more features of metabolic syndrome, such as dyslipidemia, insulin resistance, diabetes, and associated cardiovascular diseases, later in their life. Owing to the increasing global prevalence of metabolic syndrome and multiple side effects that accompany conventional medicines, much attention is directed towards medicinal plants and phytochemicals as alternative interventions. Several studies have investigated the potential of natural agents to prevent programmed metabolic syndrome. This present review, therefore, highlights an inextricable relationship between the administration of medicinal plants or phytochemicals during the intrauterine or neonatal period, and the prevention of metabolic dysfunction in adulthood, while exploring the mechanisms by which they exert such an effect. The review also identifies plant products as a novel approach to the prevention and management of metabolic syndrome.


#

Introduction

The environment modifies the functions of biological systems. Exposure of the offspring to stressful conditions such as poor maternal diet during embryonic and/or fetal development exerts significant effects on the health of the affected individual [1]. Other environmental factors such as physical, social, psychological, occupational, or lifestyle stressors could equally program the individual for the risk of metabolic disorders later in adulthood [2]. One such disorder that could arise from early life perturbations is metabolic syndrome (MetS). MetS is a complex, multifactorial cluster of physiological and metabolically related factors, such as obesity, insulin resistance, and dyslipidemia, that collectively increase the risk of developing non-alcoholic fatty liver disease (NAFLD), type 2 diabetes mellitus, hypertension, and other cardiovascular diseases [3], [4], [5]. Globally, about two-thirds of deaths arising from noncommunicable diseases are mainly caused by MetS and related disorders [6]. Given the rising cases of obesity and being overweight worldwide, MetS is considered to be a principal nutritional problem and one of the most extensively studied disorders associated with early life programming [7]. Obesity confers a significant risk for the development of MetS [8]. According to a recent report by the World Health Organization (WHO), over 1.9 billion adults aged 18 and above were estimated to be overweight worldwide, with about 650 million obese adults [9]. Despite recent therapeutic advances, the alarming increase in the global prevalence of MetS [10] constitutes a huge public health concern.

The concept of the developmental origins of health and disease provides a broader definition of the period of developmental plasticity to include the entire developmental period and not just the prenatal period [11], [12]. This period of developmental plasticity offers unique opportunities for prophylactic interventions to reverse the effects of programmed responses to an adverse environment in early life, a phenomenon known as reprogramming [13]. A number of studies have examined the potential of natural products as reprogramming strategies to prevent MetS [14], [15], [16], [17]. Here, we review dietary phytochemical agents and plant extracts with prophylactic potential in programming and reprogramming against MetS. The authors systematically searched PubMed, SCOPUS, and Google Scholar for relevant studies until December 2020. The key words were “medicinal plant extracts and metabolic syndrome in offsprings”, “maternal dietary supplements and offspring metabolic syndrome”, “metabolic syndrome and phytochemicals”, “neonatal intake”, “supplement or phytochemicals”, “high fat or high fructose induced metabolic dysfunction”, “neonatal or fetal programming of metabolic dysfunction”, “phytochemicals and neonatal programming of metabolic syndrome”, “medicinal plant extracts and phytochemicals in programmed hypertension”, “plant extracts and phytochemicals on non-alcoholic fatty liver disease”, and “epigenetics and metabolic programming”. The retrieved articles were thoroughly screened for eligibility while non-English papers were excluded.


#

Experimental and Epidemiological Evidence for the Developmental Origins of Metabolic Syndrome

Accumulating epidemiological and experimental studies provide compelling evidence for the developmental origins of MetS [18]. Epidemiological reports have associated adverse prenatal and postnatal environmental exposure to the development of MetS in adult life. Most of these epidemiological reports came from natural and man-made disasters, such as famine, which provided unique opportunities to study developmental programming in human populations [19], [20]. The Dutch famine (1944 – 1945) represents principal evidence for the developmental origins of MetS and is the most studied. Intrauterine exposure to poor nutrition due to the Dutch famine has been consistently associated with impaired metabolic phenotypes such as dyslipidemia, raised body mass index, obesity, and cardiovascular diseases [21], [22], [23]. Findings similar to those of the Dutch famine were also established in populations of other countries with data obtained from affected populations of the Austria famine [24], the Great Ukrainian famine (Holodomor) [25], Leningrad Siege [26], Chinese Great Leap Forward famine [27], Nigeriaʼs Biafran war famine [28], Europeʼs Holocaust [29], and seasonal malnutrition in Spain [30], all of which occurred in the 20th century. In most of these studies, the initial exposure to poor nutrition in early life, as a result of the famine, was associated with the development of obesity, dyslipidemia, diabetes, hypertension, and other cardiovascular disorders in later life when compared to their control counterparts who were not exposed.

Furthermore, owing to the difficulties in developing animal models that present all the features of MetS [31], investigations into the developmental origin of MetS are done using models that manifest certain components of the syndrome. For instance, several animal studies have associated intrauterine protein and caloric restriction to the development of hypertension in adult offspring, as reviewed elsewhere [32]. Since poor nutrition also includes the intake of excess fat and calories, rodent models of maternal high-fat feeding during pregnancy have also been linked to the onset of hyperinsulinemia, glucose intolerance, endothelial dysfunction, and hypertension in adult offspring [33], [34], [35]. Further evidence came from a study that demonstrated an association between low-birth-weight neonates that experienced a catch-up growth and increased risk for the development of cardiovascular disease, one of the key complications of MetS [36]. These observations clearly indicate the link between adverse fetal and neonatal environments and the risk of developing MetS in adulthood.

Epigenetics and metabolic programming

The advent of epigenetics has provided an avenue for explaining the mechanisms by which environmental factors influence fetal and neonatal phenotypes as well as the subsequent development of diseases. Epigenetics is majorly concerned with heritable changes in DNA in the absence of structural modifications to the nucleotide sequence, enabling prompt regulation of gene expression in numerous cell types [37]. The developmental period is a time when the growing fetuses and neonates are prone to maternal and environmental stress that results in programmed morphological alterations, cellular responses, and gene expression that affect the metabolism and physiology of the offspring. The outcome of developmental programming may appear instantly, for instance, abnormal organ development or, later in adulthood, as impaired organ function [38]. A primary background that may form the basis of the latter scenario is traceable to the concept of the double hit hypothesis. The hypothesis states that a preliminary intervention usually regarded as “first hit” may sensitize an organ to produce physiological alterations [39], [40]. These alterations may manifest immediately, resulting in malfunctioned organs and eventual disease development or may be suppressed [41], [42], [43], [44]. A second intervention (second hit) may unmask the suppressed effects or exacerbate the existing effects of the “first hit” [45], [46].

Suboptimal nutrition during the critical periods of developmental plasticity may alter gene expression via three different epigenetic mechanisms: (i) modification of the chromatin architecture and lysine and/or arginine residue at the N-terminal tails of histone [47], (ii) alteration in the availability of methyl groups by distorting the activities of methyltransferase and DNA demethylation [48], and (iii) modification of expression levels of miRNA involved in regulating the principal proteins in the folate-mediated carbon metabolism pathway, which is known to regulate the metabolism of methionine, homocysteine, vitamin B complex, proteins, and histones as well as DNA and ribonucleic acid (RNA) [49], [50], [51].

The mechanisms of epigenetics include DNA methylation, modification of histones, packaging of chromatin, and alteration in non-coding RNA expression [52]. Adverse intrauterine milieu is translated into epigenetic modifications during gametogenesis and fetal development and are steadily preserved until adulthood [53]. These epigenetic modifications alter the expression of genes and hence the metabolic phenotype, producing diseases in adulthood.


#

Potential mechanism by which natural products reprogram against metabolic syndrome of developmental origin

The mechanism by which natural products offer protection against developmentally programmed MetS is poorly understood. Epigenetic mechanisms have been shown to play an important role in the regulation of cellular functions and are critical to the development of complex diseases, including MetS [54]. Epigenetic modification is a reversible process and can be achieved by diet, environment, and lifestyle choices [55]. Hence, epigenetic modification is suggested to play a role in the prophylactic effects of dietary phytochemicals [56]. The exact mechanism by which plant extracts and phytochemicals interact with the epigenome to modulate the expression of genes to protect against certain metabolic disorders has not been fully elucidated.

Several bioactive compounds derived from plants are epigenetic modulators [57]. For instance, polyphenols contained in green tea have been shown to improve the metabolism of offspring born to undernourished dams by modulating the expression of enzymes that influence epigenetic marks [58]. Although epigenetic modifications are rarely studied as potential preventive mechanisms that relate to the reprogramming effects of phytochemicals with the development of MetS, they have been implicated as a mechanism of prevention in non-programming models of some metabolic disorders [57]. Resveratrol protected the diabetic rat aorta from macroangiopathy by influencing DNA methylation [59]. Also, curcumin reportedly suppresses the hyperglycemia-induced inflammatory response via the modulation of histone acetylase and histone deacetylase activity [60]. In addition, phytochemicals have also been shown to improve metabolism by regulating the expression of microRNAs. Joven et al. [61] linked the inhibition of miR-103/107 by polyphenols from Hibiscus sabdariffa (HS) to improved glucose and lipid metabolism in hyperlipidemic mice. Similarly, quercetin and polyphenol extracts from HS and coffee prevented high-fat diet (HFD)-induced liver steatosis in mice via the upregulated expression of miR-122 [62].

Therefore, epigenetic mechanisms may explain the modification of gene expression by phytochemicals or medicinal extracts during the fetal and neonatal period of metabolically challenged rodents to protect against the development of MetS in adulthood. A potential mechanism by which medicinal plants and phytochemicals act during critical periods of developmental plasticity may modulate biological systems to prevent the development of MetS is summarized in [Fig. 1] below.

Zoom Image
Fig. 1 Influence of medicinal plants and phytochemicals on metabolic programming. The figure illustrates the epigenetic mechanisms by which phytochemicals or extracts of medicinal plants may upregulate or downregulate the expression of genes associated with metabolism or its control, thereby preventing the development of MetS induced by a high-fructose or high-fat diet. HFRT = high fructose, HF = high fat, RNA = ribonucleic acid.

#

Phytochemicals and their classification

Phytochemicals are plant-sourced medicinal agents with fewer side effects compared to synthetic compounds [63]. They are broadly classified into phenolic compounds, terpenoids, and alkaloids including other nitrogen-containing plant constituents [64].

The phenolic compounds are the most abundant group of phytochemicals and are readily available in most plants. They include anthocyanins, anthochlors, benzofurans, chromones, coumarins, minor flavonoids, flavonones and flavonols, isoflavonoids, lignans, phenols and phenolic acids, phenolic ketones, phenylpropanoids, quinonoids, stilbenoids, tannins, and xanthones. Among these, flavonoids, phenolic acids, and polyphenols are the three major categories of dietary phenolics [65].

On the other hand, the terpenes (terpenoids), otherwise referred to as isoprenoids [66], are a class of natural products formed from five-carbon isoprene units. They include phytosterols (including β sitosterol), sesquiterpenes, monoterpenoids, hemiterpenoids, diterpenoids, triterpenoids, and saponins [67]. The third group, alkaloids, includes peptide, pyrrolidine and piperidine, pyrrolizidine, quinoline, betalain, indole, isoquinoline, lycopodium, quinolizidine, and tropane compounds. Other nitrogen-containing constituents include purines and pyrimidines, non-protein amino acids, and amines [67]. Most phytochemicals are sourced from fruits and vegetables and are classified according to their corresponding constituents as indicated in [Table 1] below. These phytochemicals have been investigated for their potential prophylactic activity against developmentally programmed MetS.

Table 1 Selected phytochemicals and their dietary sources.

Phytochemicals

Class

Subclass

Sources

Phytosterol

Terpenes

Vegetable oils (rapeseed oil), cereals, vegetables, fruits

Ursolic acid

Terpenes

Triterpenes

Apples, bilberries, peppermint, cranberries, elder flower, rosemary, lavender, thyme, hawthorn, oregano, prunes

Oleanolic acid

Terpenes

Triterpenes

Olives

Resveratrol

Phenolic compounds

Polyphenols

Grapes, wine, peanuts, soy

Curcumin

Phenolic compounds

Polyphenols

Tumeric, mustard

Quercetin

Phenolic compounds

Flavonoids

Cranberries, apples, onions, beans

Genistein

Phenolic compounds

Isoflavones

Soybean, fava beans, coffee, lupin

S-allyl cysteine

Nitrogen-containing compounds

Organosulphur

Garlic

Citrulline

Nitrogen-containing compounds

α-amino acid

Watermelon, legumes, nuts


#
#

Beneficial Effects of Phytochemicals and Plant Extracts against Principal Features of Metabolic Syndrome

Dyslipidemia

Dyslipidemia is one of the hallmarks of MetS. It is characterized by abnormal lipid levels, usually presenting as an increased plasma concentration of low-density lipoprotein cholesterol (LDL-C) and triglycerides, coupled with low levels of high-density lipoprotein cholesterol (HDL-C) [68]. Reports from several experimental studies have improved the understanding of natural products and their mechanisms of action towards dyslipidemia. From existing studies ([Table 2]), it is apparent that natural products offer long-term protection against MetS by targeting dyslipidemia, especially when introduced during the early periods of development. [Table 2] shows animal studies reporting on the beneficial effects of phytochemicals or plant extracts on lipid metabolism following a high-fructose (HF) diet or HFD [69], [70], [71], [72].

Table 2 Impact of various phytochemicals and plant extracts on features of metabolic syndrome.

Intervention

Experimental animal

Model

Duration of intervention

Age at evaluation

Measured parameters

Outcome

References

8-OHdG: 8-oxo-2′-deoxyguanosine; ACC2: acetyl-coenzyme A carboxylase beta; AdipoR1: adiponectin receptor 1; AdipoR2: adiponectin receptor 2; ADMA: asymmetric dimethylarginine; AGRP: agouti-related peptide; AHR: aryl hydrocarbon receptor; AMPK: 5′ adenosine monophosphate-activated protein kinase; ALT: alanine aminotransferase; AUC: area under curve; BAT: brown adipose tissue; BM: body mass; BP: blood pressure; BPA: bisphenol A; BW: body weight; CHO: cholesterol; CPT-1: carnitine palmitoyl transferase I; eNOS: endothelial nitric oxide synthase; EpiVAT: epididymal visceral adipose tissue; EpiWAT: epididymal white adipose tissue, FABP: fatty acid binding protein 1; FAS: fatty acid synthase; FGF21: fibroblast growth factor 21; FPG: fasting plasma glucose, FPI: fasting plasma insulin; GLUT-4: glucose transporter type 4; GTT: glucose tolerance test; HDL-C: high-density lipoprotein cholesterol; HH: hepatic histomorphometry; HLC: hepatic lipid content; HMGc1: high-mobility group cluster; HOMA-IR: homeostatic model assessment of insulin resistance; IL-6: interleukin 6; IngSAT: inguinal subcutaneous adipose tissue; IngWAT: inguinal white adipose tissue; IR: insulin resistance; LDL-C: low-density lipoprotein cholesterol; LFM: liver function markers; LM: liver mass; L-NAME: NG-nitro-L-arginine-methyl ester; LW: liver weight; MAP: mean arterial pressure; MCP: monocyte chemoattractant protein-1; NAFLD: non-alcoholic fatty liver disease; nNOS: neuronal nitric oxide synthase; NO: nitric oxide; NPY: neuropeptide Y; OGTT: oral glucose tolerance test; PGC1α: PPAR-gamma coactivator 1-alpha; PND: post-natal day; p-NF-κB p50: phosphorylated protein p50 subunit of nuclear factor kappa B; POMC: pro-opiomelanocortin; PPARα: peroxisome proliferator-activated receptor alpha; PPARγ: peroxisome proliferator-activated receptor gamma; SCFA: short chain fatty acid; SDMA: symmetric dimethylarginine; SIRT1: nicotinamide adenine dinucleotide-dependent deacetylase sirtuin-1; SREBP1: sterol regulatory element binding transcription factor 1; TBM: terminal body mass; TG: triglyceride; TOTAL-C: total cholesterol; VEGF: vascular endothelial growth factor

Bitter melon extract (1%)

Sprague-Dawley rats

Fetal programming (high fructose – 60%)
Offspring weaned to high-fructose diet − 60% for 20 weeks

Throughout pregnancy and lactation

23 weeks

Serum CHO, TG, LW, hepatic TG and CHO, HLC, expression of SREBP-1, ACC2, FGF21, FABP 1, PPARα, PGC1α, antioxidant activities

Upregulation of PGC1α, FGF21, and FABP 1, which depicts enhanced lipid oxidation and a reduced lipogenesis, high antioxidant activity, reduced cholesterol and triglycerides

[77]

Oleanolic acid (60 mg/kg)

Sprague-Dawley rats

Neonatal programming (high-fructose solution – 20% w/v)

8 days (PND 7 – 14)

16 weeks

AMPK, GLUT-4, CPT-1, AdipoR1, AdipoR2, TNF-α, IL-6, VEGF, MCP-1, plasma adiponectin levels

Upregulated AMPK, Glut-4 gene, increased plasma adiponectin concentration

[80]

Sprague-Dawley rats

Neonatal programming (first hit high-fructose solution – 20% w/v)
Second hit (high fructose – 20% w/v) for 56 days (PND 56 – 112)

7 days (PND 7 – 13)

16 weeks (48 h earlier)

BM, visceral fat mass, epididymal fat mass, FPG, FPI, GTT, HOMA-IR

Protected against increase in TBM, prevented rise in AUC for OGTT, normalized FBG and HOMA-IR, prevented increase in plasma TG levels

[15]

Sprague-Dawley rats

Neonatal programming (high-fructose solution – 20% w/v)
Second hit (high fructose – 20% w/v) for 56 days (PND 56 – 112)

7 days (PND 7 – 13)

16 weeks

LFM, BM, LM HLC, HH, NAFLD score

Inhibited rise in BM, precluded increase in LM, blockage of hepatic lipid accumulation, prevented the occurrence of hepatic steatosis and fibrosis

[101]

Green tea extract (7.5 and 10 g/kg)

Sprague-Dawley rats

Fetal programming (high-fat diet)

Throughout pregnancy and lactation

13 weeks

FPG, FPI, index of IR, serum, muscle, and liver TG, serum leptin and adiponectin

Insulin resistance 57% lower, serum non-esterified fatty acid 23% lower, and liver TG was 26% lower, enhanced gene/protein expression related to lipid and glucose metabolism.

[16]

Grape skin extract (200 mg/kg)

Wistar rats

Fetal programming (high-fat diet 24%)

21 days (lactation period)

12 weeks 6 days; 25 weeks 5 days

FPG, FPI, HOMA-IR, plasma CHO, TG, BP, and vascular function, antioxidant activity

Protection against hypertriglyceridemia, hyperglycemia, lowered insulin resistance, inhibited increase in body weight, protection against hypertension

[17]

Phytosterol (2% solution)

Syrian Golden Hamsters

Fetal programming (high cholesterol – 0.5%)

Throughout pregnancy

3 weeks

Total-C, LDL-C, HDL-C, hepatic TG

Prevented hypercholesterolemia

[71]

Green tea extract (400 mg/kg)

Wistar rats

Fetal programming (high-fat diet)

Throughout pregnancy and lactation

13 weeks (OGTT – 12 weeks)

BW, serum analysis, OGTT, relative tissue weight, antioxidant enzyme activity, tissue cytokine levels, quantification of inflammatory protein

Prevented hypercholesterolemia, decreased levels of IL-10, IL-6, TNF-α, and IL-1β, and p-NF-κB p50 levels in gonadal adipose tissue

[69]

Genistein (0.6 g/kg)

C57BL/6 mice

Fetal programming (high-fat diet – 60%)

21 days pre-pregnancy + Throughout Pregnancy and lactation

24 weeks

IngSAT, EpiVAT, OGTT, serum insulin, and lipid levels, gut microbiota

Decreased BG during GTT, FPI, VAT, and serum TG levels, increased HDL-C, enrichment of Rikenella as well as SCFA-producing bacteria such as Alloprevotell,

Odoribacter, and Clostridium XlVa

[70]

Genistein (4, 40 and 160 mg/kg)

Sprague-Dawley rats

Neonatal programming (high-fat diet – 60%)

5 days

9 weeks

BW, LW, HH, hepatocyte apoptosis, plasma insulin, glucagon, ALT, expression of FAS, SREBP-1, PPARα, TNF-α

Decreased BW, inflammation and steatosis, reduced apoptosis, decreased FAS, SREBP-1, high PPARα (40 mg dose proved most effective)

[86]

Hibiscus sabdariffa calyx extract (50, 500 mg/kg)

Sprague-Dawley rats

Neonatal programming (high-fructose solution- 20% w/v)

9 days (PND 4 – 14)

7 weeks 2 days (OGTT – 48 h earlier)

BM, OGTT, FPG, plasma insulin, TG, HOMA-IR

Normalized hypercholesterolemia in male and hypertriglyceridemia in female

[72]

Citrulline (2 g/kg)

Sprague-Dawley rats

Neonatal programming: pups from low-protein-fed dams – 4% throughout gestation, presented with high-fat diet challenge – 10% fructose w/v)

16 days (PND 6 – 21)

12 weeks 6 days

BW, plasma TG and HDL, insulin sensitivity index, liver TG, TC, FAS, SREBP1, HMGc1

Increased liver TG content, upregulated FAS and SREBP1, downregulation of HMGc1, no significant difference in BW, insulin sensitivity index, plasma TG and HDL

[88]

Resveratrol (200 mg/kg)

C57BL/6J mice

Fetal programming (high-fat diet – 45%)

Pregnancy + 21 days of lactation

11 weeks
(GTT – 10 weeks)

LM, BAT, IngWAT & EpiWAT, GTT, metabolic parameters, histology of adipose tissue, AMPKα, P- AMPKα, cytochrome C, PPARγ, SIRT1

Increased metabolism in BAT of offspring, decreased adiposity and promotion of insulin sensitivity, rise in energy expenditure, facilitated BAT activity, and IngWAT browning

[14]

Resveratrol (2 – 2.5 mg/kg)

Wistar rats

Fetal programming (high fat 61.6%)

Throughout pregnancy and lactation

3 weeks

Serum CHO, LDL-C, HDL-C, TG and phospholipid, blood glucose, leptin levels, VAT, SCAT, expression of POMC, AGRP, NPY, and orexin, SIRT-1, oxidative stress

Protected against increased BW, leptin, VAT, SCAT, neuropeptides expression unaffected, elevated BG

[96]

Quercetin (50, 100 and 200 mg/kg)

Sprague-Dawley rats

Fetal programming (high-fat diet – 42% fat)

Throughout gestation and lactation

14 weeks 2 days

BW, Serum CHO, HDL-C, TG, IL-6, TNF-α

Prevention against increased BW, improved insulin sensitivity, prevention against hyperlipidemia, decreased inflammatory cytokines (IL-6)

[95]

S-allyl cysteine (150 mg/kg)

Wistar rats

Neonatal programming (first hit- high fructose: 20% w/v)
Second hit (high fructose: 20% w/v)

15 days (PND 6 – 20)

16 weeks (OGTT – 48 h earlier)

BM, OGTT and AUC, FPG, fasting plasma TG and CHO, degree of adiposity, LM, HLC, HH

Prevented hepatic lipid accumulation, prevented the development of microvesicular steatosis

[97]

Wistar rats

Neonatal programming (high fructose 20% w/v)

15 days (PND 6 – 20)

3 weeks

TBM, non-fasted BG, TG, CHO, leptin and insulin concentrations and HOMA-IR, NAFLD scores

Increased insulin levels increased HOMA-IR in male, anti-insulinotropic effects in female

[98]

Resveratrol (50 mg/L)

Sprague-Dawley rats

Fetal programming
high-fat – 58%, high sucrose (25% carbohydrate) + BPA (50 µg/kg)

Throughout pregnancy and lactation

16 weeks

Systolic BP, L-citrulline, L-arginine, ADMA, SDMA, NO2 levels oxidative stress-8-OHdG staining, renal eNOS, nNOS, AHR and target genes

Lowered systolic BP, increased L-arginine, NO2 levels, increased renal eNOS and nNOS protein levels, reduced 8-OHdG density, inhibition of AHR signaling pathway

[107]

Resveratrol (50 mg/L)

Sprague-Dawley rats

Fetal programming
L-NAME (60 mg/kg), high-fat – 58%, high sucrose (25%) from weaning to adulthood

Throughout pregnancy and lactation

16 weeks

BP, L-citrulline, L-arginine, ADMA, SDMA, oxidative stress-8-OHdG, NO, gut microbiota composition

Significantly reduced systolic BP, and MAP, no effect on ADMA-NO pathway, reduced 8-OHdG density, modulated gut microbiota composition

[108]

Acai (Euterpe oleracea Mart) seed extract (200 mg/kg)

Wistar rats

Fetal programming
(low-protein diet – 6% protein diet)

Throughout pregnancy

16 weeks

BP, vascular function, oxidative stress

Prevented programmed hypertension by reducing systolic BP, decreased plasma renin levels, prevented oxidative stress

[109]

Ursolic acid (10 mg/kg)

Sprague-Dawley rats

Neonatal programming (first hit – high fructose diet – 50%); Second hit (high fructose diet – 20%) 56 days (PND70-126)

14 days (PND 6 – 20)

18 weeks 3 days

Circulating TG, CHO and glucose, BM, total calorie
intake and LFM, HLC, VF, HH

Reduced hepatic lipid accumulation, reduced hypertrophy, microvesicular and macrovesicular steatosis

[87]

Curcumin (500 mg/kg)

Sprague-Dawley rats

Neonatal programming (high-fructose solution – 20% w/v)

16 days

9 weeks

TBM and LM, HLC, steatosis scores, RNA expression of AMPKα: TNF-α

Decreased inflammation, inhibited upregulation of TNF-α, and downregulation of AMPKα

[100]

The biosynthesis of lipids is a tightly regulated process. Sterol regulatory element binding transcription factor 1 (SREBP-1), peroxisome proliferator-activated receptor delta, and peroxisome proliferator-activated receptor gamma are key regulators of lipogenesis [73]. Fatty acid synthase (FAS), acetyl-coenzyme A carboxylase (ACC), adenosine triphosphate citrate lyase, and stearoyl-CoA desaturase-1 are the target genes of SREBP-1c in the lipogenic pathway [74]. Conversely, proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) and fibroblast growth factor 21 (FGF21) promote fatty acid oxidation and regulate lipid metabolism [75], [76]. Maternal supplementation of bitter melon extract to HF-fed dams protected adult offspring against dyslipidemia via inhibition of lipogenesis and promotion of fatty acid oxidation [77]. The study by Ching et al. [77] demonstrated that this extract improved lipid metabolism via the downregulation of Srebp1, Acc2, and Fas expression levels and upregulated expression of Pgc1α and Fgf21.

In addition, adenosine monophosphate-activated protein kinase (AMPK) is a principal cellular regulator of metabolism known to activate catabolic pathways like β-oxidation and inhibit lipogenesis by modulating the expression of genes in the affected pathway [78], [79]. The administration of oleanolic acid (OA) in the neonatal period is believed to prevent dyslipidemia in adulthood through increased expression of Ampk [80]. These results suggest that plant extracts or phytochemicals acting during the critical window of developmental plasticity may target multiple genes involved in the biosynthesis and oxidation of fatty acids, thus facilitating improved lipid metabolism.

Furthermore, Zhou et al. [70] demonstrated that genistein, a phytochemical, prevents against hypertriglyceridemia and increases HDL-C levels in mice offsprings. Genistein also inhibited HFD-induced gut microbial dysbiosis. Certain intestinal bacteria such as Alloprevotella odoribacter and Clostridium XIVa have been established as being present in low quantities in mice or humans with type 2 diabetes or those presenting with obesity [81], [82], [83]. These bacteria are known to synthesize short-chain fatty acids (SCFAs), which are known for their roles in the regulation of glucose and insulin levels, probably through inhibition of lipolysis in the hepatocytes, skeletal muscle, and adipose tissue [84]. SCFAs may also function by triggering anti-inflammatory Treg cells or by lowering cytokine levels [85]. Increased quantities of SCFA-producing bacteria offer a beneficial effect in protecting the offspring against HFD-induced metabolic dysfunction [70]. This study suggests that natural products may not only act via the modulation of gene expression but also by restoring balance in the gut microbial composition of diet-induced animal models of MetS, athough, the mechanism by which this occurs has not been fully established.

Phytochemicals such as phytosterol, OA, ursolic acid (UA), and genistein have also proven effective in lowering serum or hepatic triglyceride (TG) levels and/or LDL-C levels, or increasing the levels of HDL-C, hence preventing dyslipidemia [15], [69], [70], [71]. A group of researchers [16] reported a marked decrease in the serum, liver, and muscle TG levels, including serum cholesterol and free fatty acid (FFA) of adult male offspring of green tea extract (GTE) supplemented HFD-fed dams. Consistent with these findings, grape skin extract (GSE) was effective at reducing plasma TG levels in adult offspring [17]. Although these studies did not investigate the detailed mechanisms of action by which these extracts or phytochemicals prevented the development of dyslipidemia in adult offspring, they did, however, established lipid-lowering effects in maternal or neonatal HF- or HFD-induced animal models. Further studies are therefore warranted to delineate the possible mechanism of the lipid-lowering effects of phytochemicals and plant extracts.

Phytosterol administration in hamster dams resulted in a 71% reduction in total cholesterol in the offspring. Similarly, significant decreases of 81, 50, and 36% in non-HDL-C, HDL-C, and TG levels were observed, respectively, in the progeny of high-cholesterol-fed dams [71]. Based on this analysis, phytosterol administration appears to cause a drop in HDL-C levels, however, this level is still within an acceptable range. Dietary intake of GTE by rat dams has also been shown to reduce offspring serum TG levels in adulthood from 122.9 mg/dL in the HFD controls to 88.8 mg/dL at 7.5 g/kg GTE and 78.7 mg/dL at 10 g/kg GTE [16]. Two studies on genistein reported its beneficial health effects. One of the studies investigated the influence of genistein on some features of MetS [70], while the other focused on its effect on NAFLD [86]. In the study by Zhou et al. [70], no significant changes were observed in the serum levels of total cholesterol, LDL-C, and FFA in the offspring as a result of maternal genistein intake. It was however established that genistein administration in dams increased offspring HDL-C levels by 7.8%. Although Huang et al. [86] reported a downregulated expression of lipogenic genes, the serum lipid profile was not accounted for but instead showed that genistein prevented hepatic lipid accumulation in the offspring.

Oral administration of 60 mg/kg OA to female neonatal rats challenged with a double hit of an HF diet reduced plasma TG and cholesterol levels by 40 and 20%, respectively [15]. However, the plasma lipid profiles of their male counterparts were not significantly different. A higher dose of OA might have produced a statistically significant change in the plasma lipid profile of the male rats. Although the authors assayed the total concentration of cholesterol across the various treatment groups in their work, they, however, did not consider the concentrations of its different subtypes, unlike the studies conducted on phytosterol and genistein as discussed above. Neonatal administration of UA was reported to have no significant impact on plasma TG and cholesterol levels of HF-fed rats [87].

In another study, HS aqueous extract had sex-specific actions in rats. In males, it reduced cholesterol level by 19% while in females, it lowered TG levels by 13% [72]. Similarly, maternal consumption of GSE in rats prevented a rise in TG levels in 90- and 180-day-old offspring, however, it had no significant effect on total cholesterol levels [17]. On the contrary, in a low-birth-weight and HF diet-induced model of metabolic dysfunction, a citrulline supplement given to neonates of protein-restricted dams increased liver TG content and lowered the hepatic cholesterol level, but did not alter plasma TG and HDL as well as glucose metabolism at post-natal day 90 in rats [88]. This can be explained in part by the action of citrulline in the upregulation of Fas and Srebp1, which promotes lipogenesis, as reported in this study. Conversely, previous studies in a curative approach suggested that citrulline downregulates Srebp1 [89] and prevents hypertriglyceridemia induced by HF intake [90]. Of course, a number of factors may account for the disparity in observations in response to citrulline supplementation. A lower dose of fructose (10%) for 8 weeks adopted by Tran et al. [88] compared to a higher dose of fructose (60%) for 8 weeks [89] is not likely to be responsible for increased hepatic TG levels in response to citrulline supplement as reported by Tran et al. [88]. The dosage and duration of both studies are also far apart. So, a higher dose of citrulline (2 g/kg) adopted by Tran et al. [88] compared to 0.15 – 1 g/kg used in previous studies [89], [90] may have contributed to the discrepancies in these studies. This suggests that citrulline at a higher dose may negatively impact TG metabolism.

Based on the available evidence, an insight into both the molecular mechanism and gut bacteria-associated activity of natural products would enable a proper grasp of their ability to program against MetS. In addition, harmonizing the doses of plant extracts and phytochemicals in experimental studies, with very minimal variation in experimental design, is crucial to making reasonable comparisons between different studies and would help improve our understanding of their roles and mechanism of action.


#

Insulin resistance and glucose metabolism

Insulin resistance is a condition characterized by an inability of insulin to act on target tissues [91]. In this condition, insulin is present at a normal concentration, but the tissues are unresponsive to its stimulation and as a result, more insulin is secreted, thereby leaving fasting plasma insulin at high levels [92], [93]. It usually results from a disrupted metabolism. Insulin resistance is a component of MetS and a risk factor for cardio-metabolic disorders [94].

With respect to insulin resistance, certain phytochemicals and plant extracts have demonstrated a protective effect ([Table 1]). Some plant extracts and phytochemicals have been shown to lower insulin resistance, thereby improving insulin action on target tissues. The resultant effect is the normalization of blood glucose levels, although some studies reported that some of these phytochemicals or plant extracts exert no significant effect on glucose levels and insulin sensitivity [72], [88]. Two studies on GTE have given conflicting reports on the glucose profile and insulin sensitivity [16], [69]. Decreases in the serum insulin level and insulin resistance index were observed in offspring of high-fat-fed dams supplemented with GTE compared to those fed an HFD only [16]. GTE was shown to affect serum glucose levels in a dose-dependent manner, where 10 g/kg of GTE lowered the blood glucose concentration, but a dose of 7.5 g/kg produced no significant change in the glucose levels. Additionally, the same study recorded that post-weaning intake of GTE in rats born to high-fat-fed dams did not result in a decrease in insulin and the insulin resistance index value [16]. This constitutes yet another evidence of metabolic programming, which suggests that GTE acted during pregnancy or lactation or both to bring about its effect.

Also, maternal GTE consumption lowered basal glucose levels but had no effect on the HFD-induced increase in insulin levels in rat offspring [69]. Interestingly, both studies on GTE reported its beneficial effect in reducing serum cholesterol levels. The discrepancy in the effect of GTE on glucose metabolism in these studies may be dose related as the dose adopted by Li et al. [16] was higher than that used by Hachul et al. [69], even though both had a similar duration of intervention.

Improved insulin sensitivity was also observed in HFD-fed adult offspring following maternal resveratrol supplementation (200 mg/kg) during pregnancy and lactation as indicated by enhanced glucose tolerance. However, resveratrol did not lower glucose levels in the HFD-fed mice offspring [14]. Consistent with this finding, maternal quercetin administration enhanced glucose metabolism and insulin sensitivity in adult offspring of obese dams [95]. However, a contrary observation was made by Ros et al. [96], who demonstrated an increased glycemic level in pups following maternal resveratrol supplementation in HFD-fed dams (2.5 mg/kg). The disparity in the findings may be partly explained by the difference in the experimental design. Zou et al. [14] administered a high dose of resveratrol and subjected the dams to 11 weeks of HFD, while Ros et al. [96] fed the dams a very low dose of resveratrol and euthanized the pups and dams immediately after the suckling period was over. Oral administration of OA in neonatal rats demonstrated a remarkable degree of protection from the adverse effects of HF consumption in adulthood. At 60 mg/kg, OA for 7 days normalized fasting blood glucose and homeostatic model assessment of insulin resistance (HOMA-IR) in adulthood [15]. OA offered significant protection against the development of insulin resistance via the upregulation of adiponectin levels by approximately 1.5-fold. Increased adiponectin levels in neonatal OA-administered rats are indicative of increased insulin sensitivity, hence, normalizing glucose levels compared to HFD-fed rats with lower levels of adiponectin [80]. The same observation was reported for maternal genistein intake on the offspringʼs glucose profile [70]. The progenies of genistein-supplemented dams experienced a pronounced improvement in the glucose profile. A significant reduction was observed in glycemic levels and insulin concentrations as well as HOMA-IR of the adult offspring [70].

Supplementation with GSE to lactating dams also supported the foregoing assertions regarding the benefits of these natural products on glucose homeostasis. At days 90 and 180, the study by Resende et al. [17] showed that while offspring of HFD-fed dams without supplementation developed considerable resistance to insulin, the reverse was the case for offspring from HFD-fed dams supplemented with 0.6 g/kg GSE. High-performance liquid chromatography analysis of the GSE identified four different anthocyanins as its major phytochemical composition responsible for the physiological activity [17]. S-allyl cysteine, administered during the neonatal period for 15 days in rats, did not alter insulin levels [97]. Another study by the same investigators reported that s-allyl-cysteine produced an insulinotropic effect immediately after the weaning period in the absence of a second hit of fructose insult [98], thus demonstrating a time-bound effect of s-allyl-cysteine. The time-bound effect could also explain the increased glucose levels observed in the offspring of HFD-fed dams upon weaning in response to resveratrol intake [96]. In separate studies, neonatal administration of UA [87] and HS extract [72] exhibited no prominent effect on the glucose profile and HOMA-IR. Collectively, these results show that plant extracts and phytochemicals may improve glucose metabolism by enhancing the sensitivity of target organs to insulin and the upregulation of adiponectin levels, hence abating HFD-induced hyperglycemia.


#

Body weight, fat mass, and adiposity

Increased body weight and adiposity are potential risk factors for the development of obesity. Since obesity is one of the principal predisposing factors for MetS, increased consumption of certain medicinal plants, fruits, vegetables, nuts, vegetable oils, and other rich sources of phytochemicals by expectant obese mothers could substantially minimize the vulnerability of their offspring to obesity in later life. Although limited studies are available to arrive at this conclusion, some animal studies, as reported in [Table 2], have established improved body weight and adiposity of offspring as well as overall metabolic activity following intervention by phytochemical or medicinal plant extracts in obese dams or neonates. Developmental events occurring in the neonatal period of altricial species such as rats and mice are equivalent to the events occurring in the third trimester of precocious species like humans [99]. Hence, neonatal studies in animals are translatable to the critical window of developmental plasticity in humans.

Phytochemicals that have exhibited a demonstrable impact in preventing a high-fructose or high-fat diet-induced increase in body weight include resveratrol, oleanolic acid, quercetin, and genistein [15], [70], [95], [96]. These phytochemicals were administered to pregnant or lactating dams and their effects were subsequently observed in the offspring. Alternatively, some of these phytochemicals were given during the neonatal period and their effects observed in later life after an HF or HFD challenge. On the contrary, curcumin, s-allyl cysteine, GTE, and HS have not been reported to cause any significant changes in body weight [69], [72], [97], [100].

Maternal resveratrol supplementation reduced the birth weight in offsprings of HFD-fed dams in mice [14]. A reduction was also observed in white adipose tissue (WAT) but not in brown adipose tissue (BAT) mass. Resveratrol was reported to stimulate increased energy expenditure in BAT of HFD-fed offspring via the activation of AMPKα and nicotinamide adenine dinucleotide-dependent deacetylase sirtuin-1 (SIRT1) [14]. Resveratrol also promoted thermogenesis by upregulating the expression of thermogenic genes (Prdm16, Cidea, Elovl3 and Pgc1α, p-Ampkα, and Sirt1) in inguinal WAT and epididymal WAT in the offspring. Hence, by the mechanisms highlighted above, resveratrol protected against obesity in the offspring of obese dams fed a HFD post-weaning. A similar outcome was observed at weaning in a study by Ros et al. [96] in which body weight was significantly decreased in the offspring following resveratrol supplementation in the dams. Accordingly, resveratrol reduced adiposity in subcutaneous adipose tissue in male and female offspring and had a sex-dependent effect on visceral adipose tissue (VAT) [96]. However, resveratrol decreased VAT only in female offspring in the same study.

Furthermore, resveratrol was also found to prevent excessive fat accumulation by downregulating adiponectin and fas, though no significant change in SIRT1 levels was observed. OA administration in neonatal rats prevented weight gain in adulthood following a high-fructose insult [15], [101]. Oral administration of quercetin to rat dams during pregnancy and lactation prevented an HFD-induced increase in body weight in adult offspring [95]. The regulation of lipid metabolism at the level of transcription and increased energy expenditure in BAT and WAT is therefore a potential mechanism by which plant products program protection against obesity.


#

Hypertension

Adulthood hypertension can be programmed in response to a suboptimal environment in early life [102]. Blood pressure is primarily regulated by the kidneys. The developing kidney is vulnerable to early-life insults which may produce renal programming and programmed hypertension [103]. HFD intake, nitric oxide (NO) deficiency, and oxidative stress have been implicated in the developmental programming of hypertension [104], [105]. NO is a vasodilator. Asymmetric dimethylarginine (ADMA), an endogenous NO synthase inhibitor, plays a key role in the regulation of NO-reactive oxygen species (ROS) balance [106]. An imbalance between NO and ROS produces oxidative stress, which is involved in the development of programmed hypertension [105]. Resveratrol supplementation (50 mg/L in drinking water) to rat dams during pregnancy and lactation prevented hypertension via the restoration of NO bioavailability (increased L-arginine to ADMA ratio, increased NO2 levels) and reduction of oxidative stress in offspring of HDF plus bisphenol A induced hypertension in adulthood [107]. Similarly, supplementation of resveratrol during gestation and lactation in rats prevents hypertension by reducing oxidative stress and modulating gut microbial composition in offspring challenged with N-nitro-L-arginine-methyl ester treatment and an HFD in prenatal and postnatal periods, respectively [108].

Apart from resveratrol, two plant extracts have been reported to confer protection against programmed hypertension in different programming models [17], [109]. GSE prevented adulthood hypertension in offspring born to high-fat-fed rat dams [17]. Euterpe oleracea extract has been reported to prevent programmed hypertension in adult offspring of low-protein-fed dams [109]. These findings further support the use of some medicinal plants and phytochemicals to prevent adulthood hypertension of developmental origin.


#

Non-alcoholic fatty liver disease

A few studies have investigated the potential of plant products as prophylactic agents against NAFLD in the context of metabolic programming. This disease is characterized by pathological conditions such as hepatic steatosis, non-alcoholic steatohepatitis (NASH), and secondary complications such as hepatic fibrosis, which may eventually lead to cirrhosis and hepatocellular carcinoma if not properly managed [110]. NASH is associated with lipid peroxidation and production of free radicals that elicit inflammation and activate stellate cells in the liver, which collectively result in fibrosis [111], [112].

When neonatal Sprague-Dawley rats were administered 10 mg/kg of UA and fructose, followed by HF consumption in adulthood, fructose-induced hepatic lipid accretion was suppressed by UA compared to the control groups [87]. Similarly, 60 mg/kg of OA administered to suckling Sprague-Dawley rats counteracted the adverse effects of an HF diet by significantly decreasing the hepatic lipid load [101]. In HF-fed adult Sprague-Dawley rats of both sexes, curcumin, at 500 mg/kg body mass administered during suckling, conferred protection against NASH and lowered hepatic lipid accumulation in female, but not in the male, rats [100]. In another study, neonatal administration of genistein for 5 days post-natal protected against HFD-induced hepatic steatosis and NASH in adult rats [86]. Converging evidence has apparently demonstrated the potential of phytochemicals as prophylaxis against the development of NAFLD and, as such, may serve as a natural strategic intervention in abating NAFLD in the general populace.


#

Knowledge gaps and recommendations

This review provides an overview of the medicinal herbs and phytochemicals with potential for use as reprogramming agents to offset developmentally programmed MetS. Even though attempts have been made to provide plausible explanations on how plant extracts and phytochemicals could prevent programmed MetS, there is little research about the interaction between these natural products and epigenetic marks in programming models. Epigenetic changes, given its importance in metabolic programming, should be investigated to further understand the mechanism of action of plants and phytochemicals in the prevention of programmed MetS. There is also a need to assess toxicity and evaluate safety doses of herbal medicines and phytochemicals in humans. On the same basis, robust clinical trials with the aim of developing natural product-enriched supplements to ascertain effective interventions during pregnancy and the early postnatal period for the prevention of MetS and other chronic diseases are urgently needed to juxtapose the claims from experimental animal studies.


#

Conclusions

Ample experimental evidence indicates that medicinal plants and phytochemicals confer a substantial degree of protection against developmentally programmed MetS. It can be deduced that the majority of these agents are potentially strong candidates for mitigating the incidence of MetS and related NAFLD. Given their unparalleled health benefits and negligible side effects, medicinal plants and phytochemicals are on the path to transforming protective reprogramming against the development of MetS in individuals exposed to suboptimal conditions in the fetal and neonatal periods.


#
#

Contributorsʼ Statement

Conceptual framework: K. G. I. and K. A. A.; scientific assessment and approval of review proposal: M. U. I., M. B. A. M. B. B., I. M., and B. A.; drafting the manuscript: K. A. A., K. G. I., I. M., M. B. B., B. A., and M. U. I.; concept and preparation of figure: K. A. A., K. G. I., and I. M.; critical revision of the manuscript: M. B. A., M. U. I., I. M., M. B. B., and B. A. All authors read and agree on the final version of the manuscript.


#
#

Conflict of Interest

The authors declare that they have no conflict of interest.

Acknowledgements

The scholars and staff of the Centre for Advanced Medical Research and Training (CAMRET) are highly appreciated for their various contributions to the success of this paper. Kehinde Ahmad Adeshina is a recipient of a CAMRET research funded scholarship (CAMRET/2019/MSc/SCH002). This study was supported by the Institution-Based Research (IBR) grant of the Tertiary Education Trust Fund (TETFUND) of Nigeria awarded to Usmanu Danfodiyo University Sokoto (TETFUND/DR&D/CE/UNIV/SOKOTO/RP/VOL.1).

  • References

  • 1 Ramírez-Alarcón K, Sánchez-Agurto Á, Lamperti L, Martorell M. Epigenetics, maternal diet and metabolic programming. Open Biol J 2019; 7: 45-51
  • 2 Godfrey KM, Costello PM, Lillycrop KA. Development, Epigenetics and metabolic Programming. In: Fewtrell MS, Haschke F, Prescott SL. eds. Preventive Aspect of early Nutrition. Basel: Karger; 2016: 71-80
  • 3 Grundy SM. Metabolic syndrome update. Trends Cardiovasc Med 2016; 26: 364-373
  • 4 Lee HS. Maternal Nutrition, epigenetic Programming and metabolic Syndrome. In: Ferguson BS. ed. Nutritional Epigenomics. Amsterdam: Elsevier; 2019: 153-166
  • 5 Grundy SM, Brewer jr. HB, Cleeman JI, Smith jr. SC, Lenfant C. National Heart, Lung, and Blood Institute, American Heart Association. Definition of metabolic syndrome: report of the National Heart, Lung, and Blood Institute/American Heart Association conference on scientific issues related to definition. Arterioscler Thromb Vasc Biol 2004; 24: e13-e18
  • 6 Zarocostas J. Need to increase focus on non-communicable diseases in global health, says WHO. BMJ 2010; 341: c7065
  • 7 Gregory JW. Prevention of obesity and metabolic syndrome in children. Front Endocrinol (Lausanne) 2019; 10: 669
  • 8 Furukawa S, Fujita T, Shimabukuro M, Iwaki M, Yamada Y, Nakajima Y, Nakayama O, Makishima M, Matsuda M, Shimomura I. Increased oxidative stress in obesity and its impact on metabolic syndrome. J Clin Invest 2004; 114: 1752-1761
  • 9 World Health Organization. Facts about overweight and obesity. World Health Organization 2020. Accessed April 3, 2020 at: https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight
  • 10 Saklayen MG. The global epidemic of the metabolic syndrome. Curr Hypertens Rep 2018; 20: 12
  • 11 Gillman MW, Barker D, Bier D, Cagampang F, Challis J, Fall C, Godfrey K, Gluckman P, Hanson M, Kuh D, Nathanielsz P, Nestel P, Thornburg KL. Meeting report on the 3rd international congress on Developmental Origins of Health and Disease (DOHaD). Pediatr Res 2007; 61: 625-629
  • 12 Suzuki K. The developing world of DOHaD. J Dev Orig Health Dis 2018; 9: 266-269
  • 13 Tain YL, Joles JA. Reprogramming: A preventive strategy in hypertension focusing on the kidney. Int J Mol Sci 2015; 17: 23
  • 14 Zou T, Chen D, Yang Q, Wang B, Zhu MJ, Nathanielsz PW, Du M. Resveratrol supplementation of high-fat diet-fed pregnant mice promotes brown and beige adipocyte development and prevents obesity in male offspring. J Physiol 2017; 595: 1547-1562
  • 15 Nyakudya TT, Mukwevho E, Erlwanger KH. The protective effect of neonatal oral administration of oleanolic acid against the subsequent development of fructose-induced metabolic dysfunction in male and female rats. Nutr Metab 2019; 15: 82
  • 16 Li S, Tse IMY, Li ETS. Maternal green tea extract supplementation to rats fed a high-fat diet ameliorates insulin resistance in adult male offspring. J Nutr Biochem 2012; 23: 1655-1660
  • 17 Resende AC, Emiliano AF, Cordeiro VSC, de Bem GF, de Cavalho LC, de Oliveira PR, Neto ML, Costa CA, Boaventura GT, de Moura RS. Grape skin extract protects against programmed changes in the adult rat offspring caused by maternal high-fat diet during lactation. J Nutr Biochem 2013; 24: 2119-2126
  • 18 Tain YL, Hsu CN. Developmental programming of the metabolic syndrome: Can we reprogram with resveratrol?. Int J Mol Sci 2018; 19: 2584
  • 19 de Gusmão Correia ML, Volpato AM, Águila MB, Mandarim-de-Lacerda CA. Developmental origins of health and disease: experimental and human evidence of fetal programming for metabolic syndrome. J Hum Hypertens 2012; 26: 405-419
  • 20 Vaiserman AM. Early-life nutritional programming of type 2 diabetes: Experimental and quasi-experimental evidence. Nutrients 2017; 9: 236
  • 21 Lumey LH, Stein AD, Susser E. Prenatal famine and adult health. Annu Rev Public Health 2011; 32: 237-262
  • 22 Heijmans BT, Tobi EW, Lumey LH, Slagboom PE. The epigenome: archive of the prenatal environment. Epigenetics 2009; 4: 526-531
  • 23 Roseboom TJ, Painter RC, van Abeelen AF, Veenendaal MV, de Rooij SR. Hungry in the womb: What are the consequences? Lessons from the Dutch famine. Maturitas 2011; 70: 141-145
  • 24 Thurner S, Klimek P, Szell M, Duftschmid G, Endel G, Kautzky-Willer A, Kasper DC. Quantification of excess risk for diabetes for those born in times of hunger, in an entire population of a nation, across a century. Proc Natl Acad Sci U S A 2013; 110: 4703-4707
  • 25 Lumey LH, Khalangot MD, Vaiserman AM. Association between type 2 diabetes and prenatal exposure to the Ukraine famine of 1932–33: a retrospective cohort study. Lancet Diabetes Endocrinol 2015; 3: 787-794
  • 26 Koupil I, Shestov DB, Sparén P, Plavinskaja S, Parfenova N, Vågerö D. Blood pressure, hypertension and mortality from circulatory disease in men and women who survived the siege of Leningrad. Eur J Epidemiol 2007; 22: 223-234
  • 27 Wang N, Wang X, Han B, Li Q, Chen Y, Zhu C, Chen Y, Xia F, Cang Z, Zhu C, Lu M, Meng Y, Chen C, Lin D, Wang B, Jensen MD, Lu Y. Is exposure to famine in childhood and economic development in adulthood associated with diabetes?. J Clin Endocrinol Metab 2015; 100: 4514-4523
  • 28 Hult M, Tornhammar P, Ueda P, Chima C, Bonamy AK, Ozumba B, Norman M. Hypertension, diabetes and overweight: Looming legacies of the biafran famine. PLoS One 2010; 5: e13582
  • 29 Keinan-Boker L, Shasha-Lavsky H, Eilat-Zanani S, Edri-Shur A, Shasha SM. Chronic health conditions in Jewish Holocaust survivors born during world war II. Isr Med Assoc J 2015; 17: 206-212
  • 30 Banegas JR, Rodríguez-Artalejo F, de la Cruz JJ, Graciani A, Villar F, del Rey-Calero J. Adult men born in spring have lower blood pressure. J Hypertens 2000; 18: 1763-1766
  • 31 Panchal SK, Brown L. Rodent models for metabolic syndrome research. J Biomed Biotechnol 2011; 2011: 351982
  • 32 Armitage JA, Khan IY, Taylor PD, Nathanielsz PW, Poston L. Developmental programming of the metabolic syndrome by maternal nutritional imbalance: how strong is the evidence from experimental models in mammals?. J Physiol 2004; 561: 355-377
  • 33 Srinivasan M, Katewa SD, Palaniyappan A, Pandya JD, Patel MS. Maternal high-fat diet consumption results in fetal malprogramming predisposing to the onset of metabolic syndrome-like phenotype in adulthood. Am J Physiol Endocrinol Metab 2006; 291: E792-E799
  • 34 Samuelsson AM, Matthews PA, Argenton M, Christie MR, McConnell JM, Jansen EH, Piersma AH, Ozanne SE, Twinn DF, Remacle C, Rowlerson A, Poston L, Taylor PD. Diet-induced obesity in female mice leads to offspring hyperphagia, adiposity, hypertension, and insulin resistance: a novel murine model of developmental programming. Hypertension 2008; 51: 383-392
  • 35 Torrens C, Ethirajan P, Bruce KD, Cagampang FR, Siow RC, Hanson MA, Byrne CD, Mann GE, Clough GF. Interaction between maternal and offspring diet to impair vascular function and oxidative balance in high fat fed male mice. PLoS One 2012; 7: e50671
  • 36 Kelishadi R, Haghdoost AA, Jamshidi F, Aliramezany M, Moosazadeh M. Low birthweight or rapid catch-up growth: which is more associated with cardiovascular disease and its risk factors in later life? A systematic review and cryptanalysis. Paediatr Int Child Health 2015; 35: 110-123
  • 37 Kitsiou-Tzeli S, Tzetis M. Maternal epigenetics and fetal and neonatal growth. Curr Opin Endocrinol Diabetes Obes 2017; 24: 43-46
  • 38 Desai M, Jellyman JK, Ross MG. Epigenomics, gestational programming and risk of metabolic syndrome. Int J Obes 2015; 39: 633-641
  • 39 Erdélyi K, Pacher P, Virág L, Szabó C. Role of poly(ADP-ribosyl)ation in a ‘two-hit’ model of hypoxia and oxidative stress in human A549 epithelial cells in vitro . Int J Mol Med 2013; 32: 339-346
  • 40 Morris CF, Tahir M, Arshid S, Castro MS, Fontes W. Reconciling the IPC and two-hit models: Dissecting the underlying cellular and molecular mechanisms of two seemingly opposing frameworks. J Immunol Res 2015; 2015: 697193
  • 41 Bayer TA, Falkai P, Maier W. Genetic and non-genetic vulnerability factors in schizophrenia: the basis of the “two hit hypothesis”. J Psychiatr Res 1999; 33: 543-548
  • 42 Lahiri DK, Maloney B, Basha MR, Ge YW, Zawia NH. How and when environmental agents and dietary factors affect the course of Alzheimerʼs Disease: the “LEARn” model (latent early-life associated regulation) may explain the triggering of AD. Curr Alzheimer Res 2007; 4: 219-228
  • 43 Lahiri DK, Maloney B, Zawia NH. The LEARn model: an epigenetic explanation for idiopathic neurobiological diseases. Mol Psychiatry 2009; 14: 992-1003
  • 44 Heindel JJ, Balbus J, Birnbaum L, Brune-Drisse MN, Grandjean P, Gray K, Landrigan PJ, Sly PD, Suk W, Cory Slechta D, Thompson C, Hanson M. Developmental origins of health and disease: Integrating environmental influences. Endocrinology 2015; 156: 3416-3421
  • 45 Tsukamoto H, Machida K, Dynnyk A, Mkrtchyan H. “Second hit” models of alcoholic liver disease. Semin Liver Dis 2009; 29: 178-187
  • 46 Howard J. The cytokine hypothesis: a neurodevelopmental explanation for the emergence of schizophrenia later in life. Adv Biosci Biotechnol 2013; 04: 81-88
  • 47 Zhang P, Torres K, Liu X, Liu CG, Pollock RE. An overview of chromatin-regulating proteins in cells. Curr Protein Pept Sci 2016; 17: 401-410
  • 48 Gao J, Cahill CM, Huang X, Roffman JL, Lamon-Fava S, Fava M, Mischoulon D, Rogers JT. S-adenosyl methionine and transmethylation pathways in neuropsychiatric diseases throughout life. Neurotherapeutics 2018; 15: 156-175
  • 49 Curtin K, Samowitz WS, Ulrich CM, Wolff RK, Herrick JS, Caan BJ, Slattery ML. Nutrients in folate-mediated, one-carbon metabolism and the risk of rectal tumors in men and women. Nutr Cancer 2011; 63: 357-366
  • 50 Hardy TM, Tollefsbol TO. Epigenetic diet: impact on the epigenome and cancer. Epigenomics 2011; 3: 503-518
  • 51 Williams SR, Yang Q, Chen F, Liu X, Keene KL, Jacques P, Chen WM, Weinstein G, Hsu FC, Beiser A, Wang L, Bookman E, Doheny KF, Wolf PA, Zilka M, Selhub J, Nelson S, Gogarten SM, Worrall BB, Seshadri S. Sale MM; Genomics and Randomized Trials Network, Framingham Heart Study. Correction to genome-wide meta-analysis of homocysteine and methionine metabolism identifies five one carbon metabolism loci and a novel association of ALDH1L1 with ischemic stroke. PLoS Genet 2014; 10: e1004214
  • 52 Zhu Z, Cao F, Li X. Epigenetic programming and fetal metabolic programming. Front Endocrinol (Lausanne) 2019; 10: 764
  • 53 Jablonka E, Lamb MJ. The inheritance of acquired epigenetic variations. Int J Epidemiol 2015; 44: 1094-1103
  • 54 Lee HS. Impact of maternal diet on the epigenome during in utero life and the developmental programming of diseases in childhood and adulthood. Nutrients 2015; 7: 9492-9507
  • 55 Lee PS, Chiou YS, Ho CT, Pan MH. Chemoprevention by resveratrol and pterostilbene: Targeting on epigenetic regulation. Biofactors 2018; 44: 26-35
  • 56 Chango A, Pogribny IP. Considering maternal dietary modulators for epigenetic regulation and programming of the fetal epigenome. Nutrients 2015; 7: 2748-2770
  • 57 Vahid F, Zand H, Nosrat-Mirshekarlou E, Najafi R, Hekmatdoost A. The role dietary of bioactive compounds on the regulation of histone acetylases and deacetylases: A review. Gene 2015; 562: 8-15
  • 58 Sun Y, Mukai Y, Tanaka M, Saito T, Sato S, Kurasaki M. Green tea extract increases mRNA expression of enzymes which influence epigenetic marks in newborn female offspring from undernourished pregnant mother. PLoS One 2013; 8: e74559
  • 59 Lou XD, Wang HD, Xia SJ, Skog S, Sun J. Effects of resveratrol on the expression and DNA methylation of cytokine genes in diabetic rat aortas. Arch Immunol Ther Exp (Warsz) 2014; 62: 329-340
  • 60 Yun JM, Jialal I, Devaraj S. Epigenetic regulation of high glucose-induced proinflammatory cytokine production in monocytes by curcumin. J Nutr Biochem 2011; 22: 450-458
  • 61 Joven J, Espinel E, Rull A, Aragonès G, Rodríguez-Gallego E, Camps J, Micol V, Herranz-López M, Menéndez JA, Borrás I, Segura-Carretero A, Alonso-Villaverde C, Beltrán-Debón R. Plant-derived polyphenols regulate expression of miRNA paralogs miR-103/107 and miR-122 and prevent diet-induced fatty liver disease in hyperlipidemic mice. Biochim Biophys Acta 2012; 1820: 894-899
  • 62 Ross SA, Davis CD. The emerging role of microRNAs and nutrition in modulating health and disease. Annu Rev Nutr 2014; 34: 305-336
  • 63 Fennell CW, Lindsey KL, McGaw LJ, Sparg SG, Stafford GI, Elgorashi EE, Grace OM, van Staden J. Assessing African medicinal plants for efficacy and safety: pharmacological screening and toxicology. J Ethnopharmacol 2004; 94: 205-217
  • 64 Harborne JB. Classes and Functions of secondary Products from Plants. In: Walton JN, Brown DE. eds. Chemicals from Plants: Perspectives on Plant Secondary. New Jersey: World Scientific; 1999: 1-25
  • 65 Saxena M, Saxena J, Nema R, Singh D, Gupta A. Phytochemistry of medicinal plants. J Pharmacogn Phytochem 2013; 1: 168-182
  • 66 Harborne JB, Baxter HE. Phytochemical Dictionary: A Handbook of Bioactive Compounds from Plants. London: Taylor & Francis; 1993
  • 67 Dillard CJ, German BJ. Phytochemicals: nutraceuticals and human health. J Sci Food Agric 2000; 80: 1744-1756
  • 68 Kopin L, Lowenstein C. In the clinic. Dyslipidemia. Ann Intern Med 2017; 167: ITC81-ITC96
  • 69 Hachul ACL, Boldarine VT, Neto NIP, Moreno MF, Ribeiro EB, do Nascimento CMO, Oyama LM. Maternal consumption of green tea extract during pregnancy and lactation alters offspringʼs metabolism in rats. PLoS One 2018; 13: e0199969
  • 70 Zhou L, Xiao X, Zhang Q, Zheng J, Deng M. Maternal genistein intake mitigates the deleterious effects of high-fat diet on glucose and lipid metabolism and modulates gut microbiota in adult life of male mice. Front Physiol 2019; 10: 985
  • 71 Liu J, Iqbal A, Raslawsky A, Browne RW, Patel MS, Rideout TC. Influence of maternal hypercholesterolemia and phytosterol intervention during gestation and lactation on dyslipidemia and hepatic lipid metabolism in offspring of Syrian golden hamsters. Mol Nutr Food Res 2016; 60: 2151-2160
  • 72 Ibrahim KG, Chivandi E, Mojiminiyi FBO, Erlwanger KH. The response of male and female rats to a high-fructose diet during adolescence following early administration of Hibiscus sabdariffa aqueous calyx extracts. J Dev Orig Health Dis 2017; 8: 628-637
  • 73 Oikari S, Ahtialansaari T, Heinonen MV, Mauriala T, Auriola S, Kiehne K, Fölsch UR, Jänne J, Alhonen L, Herzig KH. Downregulation of PPARs and SREBP by acyl-CoA-binding protein overexpression in transgenic rats. Pflugers Arch 2008; 456: 369-377
  • 74 Wang Y, Viscarra J, Kim SJ, Sul HS. Transcriptional regulation of hepatic lipogenesis. Nat Rev Mol Cell Biol 2015; 16: 678-689
  • 75 Supruniuk E, Miklosz A, Chabowski A. The implication of PGC-1α on fatty acid transport across plasma and mitochondrial membranes in the insulin sensitive tissues. Front Physiol 2017; 8: 923
  • 76 Planavila A, Redondo I, Hondares E, Vinciguerra M, Munts C, Iglesias R, Gabrielli LA, Sitges M, Giralt M, van Bilsen M, Villarroya F. Fibroblast growth factor 21 protects against cardiac hypertrophy in mice. Nat Commun 2013; 4: 2019
  • 77 Ching RH, Yeung LO, Tse IM, Sit WH, Li ET. Supplementation of bitter melon to rats fed a high-fructose diet during gestation and lactation ameliorates fructose-induced dyslipidemia and hepatic oxidative stress in male offspring. J Nutr 2011; 141: 1664-1672
  • 78 Leff T. AMP-activated protein kinase regulates gene expression by direct phosphorylation of nuclear proteins. Biochem Soc Trans 2003; 31: 224-227
  • 79 Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, Wu M, Ventre J, Doebber T, Fujii N, Musi N, Hirshman MF, Goodyear LJ, Moller DE. Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest 2001; 108: 1167-1174
  • 80 Matumba MG, Ayeleso AO, Nyakudya T, Erlwanger K, Chegou NN, Mukwevho E. Long-term impact of neonatal intake of oleanolic acid on the expression of AMP-activated protein kinase, adiponectin and inflammatory cytokines in rats fed with a high fructose diet. Nutrients 2019; 11: 226
  • 81 Schwiertz A, Taras D, Schäfer K, Beijer S, Bos NA, Donus C, Hardt PD. Microbiota and SCFA in lean and overweight healthy subjects. Obesity (Silver Spring) 2010; 18: 190-195
  • 82 Gomez-Arango LF, Barrett HL, McIntyre HD, Callaway LK, Morrison M, Dekker Nitert M. SPRING Trial Group. Connections between the gut microbiome and metabolic hormones in early pregnancy in overweight and obese women. Diabetes 2016; 65: 2214-2223
  • 83 Jung MJ, Lee J, Shin NR, Kim MS, Hyun DW, Yun JH, Kim PS, Whon TW, Bae JW. Chronic repression of mTOR complex 2 induces changes in the gut microbiota of diet-induced obese mice. Sci Rep 2016; 6: 30887
  • 84 Canfora EE, Jocken JW, Blaak EE. Short-chain fatty acids in control of body weight and insulin sensitivity. Nat Rev Endocrinol 2015; 11: 577-591
  • 85 Tremaroli V, Bäckhed F. Functional interactions between the gut microbiota and host metabolism. Nature 2012; 489: 242-249
  • 86 Huang C, Qiao X, Dong B. Neonatal exposure to genistein ameliorates high-fat diet-induced non-alcoholic steatohepatitis in rats. Br J Nutr 2011; 106: 105-113
  • 87 Mukonowenzou NC, Dangarembizi R, Chivandi E, Nkomozepi P, Erlwanger KH. Administration of ursolic acid to new-born pups prevents dietary fructose-induced non-alcoholic fatty liver disease in Sprague Dawley rats. J Dev Orig Health Dis 2021; 12: 101-112
  • 88 Tran NT, Alexandre-Gouabau MC, Pagniez A, Ouguerram K, Boquien CY, Winer N, Darmaun D. Neonatal citrulline supplementation and later exposure to a high fructose diet in rats born with a low birth weight: A preliminary report. Nutrients 2017; 9: 375
  • 89 Jegatheesan P, Beutheu S, Ventura G, Nubret E, Sarfati G, Bergheim I, De Bandt JP. Citrulline and nonessential amino acids prevent fructose-induced nonalcoholic fatty liver disease in rats. J Nutr 2015; 145: 2273-2279
  • 90 Jegatheesan P, Beutheu S, Ventura G, Sarfati G, Nubret E, Kapel N, Waligora-Dupriet AJ, Bergheim I, Cynober L, De-Bandt JP. Effect of specific amino acids on hepatic lipid metabolism in fructose-induced non-alcoholic fatty liver disease. Clin Nutr 2016; 35: 175-182
  • 91 Petersen MC, Shulman GI. Mechanisms of insulin action and insulin resistance. Physiol Rev 2018; 98: 2133-2223
  • 92 Kahn SE. The relative contributions of insulin resistance and beta-cell dysfunction to the pathophysiology of Type 2 diabetes. Diabetologia 2003; 46: 1707
  • 93 Czech MP. Insulin action and resistance in obesity and type 2 diabetes. Nat Med 2017; 23: 804-814
  • 94 Gayoso-Diz P, Otero-González A, Rodriguez-Alvarez MX, Gude F, García F, De Francisco A, Quintela AG. Insulin resistance (HOMA-IR) cut-off values and the metabolic syndrome in a general adult population: effect of gender and age: EPIRCE cross-sectional study. BMC Endocr Disord 2013; 13: 47
  • 95 Wu Z, Zhao J, Xu H, Lyv Y, Feng X, Fang Y, Xu Y. Maternal quercetin administration during gestation and lactation decrease endoplasmic reticulum stress and related inflammation in the adult offspring of obese female rats. Eur J Nutr 2014; 1669-1683
  • 96 Ros P, Díaz F, Freire-Regatillo A, Argente-Arizón P, Barrios V, Argente J, Chowen JA. Resveratrol intake during pregnancy and lactation modulates the early metabolic effects of maternal nutrition differently in male and female offspring. Endocrinology 2018; 159: 810-825
  • 97 Lembede BW, Erlwanger KH, Nkomozepi P, Chivandi E. Effect of neonatal orally administered S-allyl cysteine in high-fructose diet fed Wistar rats. J Dev Orig Health Dis 2018; 9: 160-171
  • 98 Lembede BW, Joubert J, Nkomozepi P, Erlwanger KH, Chivandi E. Insulinotropic effect of S-Allyl cysteine in rat pups. Prev Nutr Food Sci 2018; 23: 15-21
  • 99 Clancy B, Darlington RB, Finlay BL. Translating developmental time across mammalian species. Neuroscience 2001; 105: 7-17
  • 100 Ibrahim KG, Chivandi E, Nkomozepi P, Matumba MG, Mukwevho E, Erlwanger KH. The long-term protective effects of neonatal administration of curcumin against nonalcoholic steatohepatitis in high-fructose-fed adolescent rats. Physiol Rep 2019; 7: e14032
  • 101 Nyakudya TT, Mukwevho E, Nkomozepi P, Erlwanger KH. Neonatal intake of oleanolic acid attenuates the subsequent development of high fructose diet-induced non-alcoholic fatty liver disease in rats. J Dev Orig Health Dis 2018; 9: 500-510
  • 102 Hanson M, Gluckman P. Developmental origins of noncommunicable disease: population and public health implications. Am J Clin Nutr 2011; 94: 1754S-1758S
  • 103 Kett MM, Denton KM. Renal programming: cause for concern?. Am J Physiol Regul Integr Comp Physiol 2011; 300: R791-R803
  • 104 Williams L, Seki Y, Vuguin PM, Charron MJ. Animal models of in utero exposure to a high fat diet: a review. Biochim Biophys Acta 2014; 1842: 507-519
  • 105 Tain YL, Hsu CN. Interplay between oxidative stress and nutrient sensing signaling in the developmental origins of cardiovascular disease. Int J Mol Sci 2017; 18: 841
  • 106 Tain YL, Hsu CN. Targeting on asymmetric dimethylarginine-related nitric oxide-reactive oxygen species imbalance to reprogram the development of hypertension. Int J Mol Sci 2016; 17: 2020
  • 107 Hsu CN, Lin YJ, Tain YL. Maternal exposure to bisphenol A combined with high-fat diet-induced programmed hypertension in adult male rat offspring: Effects of resveratrol. Int J Mol Sci 2019; 20: 4382
  • 108 Chen HE, Lin YJ, Lin IC, Yu HR, Sheen JM, Tsai CC, Huang LT, Tain YL. Resveratrol prevents combined prenatal NG-nitro-L-arginine-methyl ester (L-NAME) treatment plus postnatal high-fat diet induced programmed hypertension in adult rat offspring: interplay between nutrient-sensing signals, oxidative stress and gut microbiota. J Nutr Biochem 2019; 70: 28-37
  • 109 de Bem GF, da Costa CA, de Oliveira PR, Cordeiro VS, Santos IB, de Carvalho LC, Souza MA, Ognibene DT, Daleprane JB, Sousa PJ, Resende AC, de Moura RS. Protective effect of Euterpe oleracea Mart (açaí) extract on programmed changes in the adult rat offspring caused by maternal protein restriction during pregnancy. J Pharm Pharmacol 2014; 66: 1328-1338
  • 110 Liang W, Menke AL, Driessen A, Koek GH, Lindeman JH, Stoop R, Havekes LM, Kleemann R, van den Hoek AM. Establishment of a general NAFLD scoring system for rodent models and comparison to human liver pathology. PLoS One 2014; 9: e115922
  • 111 Castro GS, Cardoso JF, Vannucchi H, Zucoloto S, Jordão AA. Fructose and NAFLD: metabolic implications and models of induction in rats. Acta Cir Bras 2011; 26: 45-50
  • 112 Dietrich P, Hellerbrand C. Non-alcoholic fatty liver disease, obesity and the metabolic syndrome. Best Pract Res Clin Gastroenterol 2014; 28: 637-653

Correspondence

Kasimu Ghandi Ibrahim, PhD
Department of Physiology
Faculty of Basic Medical Sciences
College of Health Sciences
Usmanu Danfodiyo University
Hospital Road, P. M. B. 2254
840232 Sokoto
Nigeria   
Phone: + 23 4 80 35 04 61 18   

Publication History

Received: 22 December 2020

Accepted after revision: 13 April 2021

Article published online:
17 May 2021

© 2021. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References

  • 1 Ramírez-Alarcón K, Sánchez-Agurto Á, Lamperti L, Martorell M. Epigenetics, maternal diet and metabolic programming. Open Biol J 2019; 7: 45-51
  • 2 Godfrey KM, Costello PM, Lillycrop KA. Development, Epigenetics and metabolic Programming. In: Fewtrell MS, Haschke F, Prescott SL. eds. Preventive Aspect of early Nutrition. Basel: Karger; 2016: 71-80
  • 3 Grundy SM. Metabolic syndrome update. Trends Cardiovasc Med 2016; 26: 364-373
  • 4 Lee HS. Maternal Nutrition, epigenetic Programming and metabolic Syndrome. In: Ferguson BS. ed. Nutritional Epigenomics. Amsterdam: Elsevier; 2019: 153-166
  • 5 Grundy SM, Brewer jr. HB, Cleeman JI, Smith jr. SC, Lenfant C. National Heart, Lung, and Blood Institute, American Heart Association. Definition of metabolic syndrome: report of the National Heart, Lung, and Blood Institute/American Heart Association conference on scientific issues related to definition. Arterioscler Thromb Vasc Biol 2004; 24: e13-e18
  • 6 Zarocostas J. Need to increase focus on non-communicable diseases in global health, says WHO. BMJ 2010; 341: c7065
  • 7 Gregory JW. Prevention of obesity and metabolic syndrome in children. Front Endocrinol (Lausanne) 2019; 10: 669
  • 8 Furukawa S, Fujita T, Shimabukuro M, Iwaki M, Yamada Y, Nakajima Y, Nakayama O, Makishima M, Matsuda M, Shimomura I. Increased oxidative stress in obesity and its impact on metabolic syndrome. J Clin Invest 2004; 114: 1752-1761
  • 9 World Health Organization. Facts about overweight and obesity. World Health Organization 2020. Accessed April 3, 2020 at: https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight
  • 10 Saklayen MG. The global epidemic of the metabolic syndrome. Curr Hypertens Rep 2018; 20: 12
  • 11 Gillman MW, Barker D, Bier D, Cagampang F, Challis J, Fall C, Godfrey K, Gluckman P, Hanson M, Kuh D, Nathanielsz P, Nestel P, Thornburg KL. Meeting report on the 3rd international congress on Developmental Origins of Health and Disease (DOHaD). Pediatr Res 2007; 61: 625-629
  • 12 Suzuki K. The developing world of DOHaD. J Dev Orig Health Dis 2018; 9: 266-269
  • 13 Tain YL, Joles JA. Reprogramming: A preventive strategy in hypertension focusing on the kidney. Int J Mol Sci 2015; 17: 23
  • 14 Zou T, Chen D, Yang Q, Wang B, Zhu MJ, Nathanielsz PW, Du M. Resveratrol supplementation of high-fat diet-fed pregnant mice promotes brown and beige adipocyte development and prevents obesity in male offspring. J Physiol 2017; 595: 1547-1562
  • 15 Nyakudya TT, Mukwevho E, Erlwanger KH. The protective effect of neonatal oral administration of oleanolic acid against the subsequent development of fructose-induced metabolic dysfunction in male and female rats. Nutr Metab 2019; 15: 82
  • 16 Li S, Tse IMY, Li ETS. Maternal green tea extract supplementation to rats fed a high-fat diet ameliorates insulin resistance in adult male offspring. J Nutr Biochem 2012; 23: 1655-1660
  • 17 Resende AC, Emiliano AF, Cordeiro VSC, de Bem GF, de Cavalho LC, de Oliveira PR, Neto ML, Costa CA, Boaventura GT, de Moura RS. Grape skin extract protects against programmed changes in the adult rat offspring caused by maternal high-fat diet during lactation. J Nutr Biochem 2013; 24: 2119-2126
  • 18 Tain YL, Hsu CN. Developmental programming of the metabolic syndrome: Can we reprogram with resveratrol?. Int J Mol Sci 2018; 19: 2584
  • 19 de Gusmão Correia ML, Volpato AM, Águila MB, Mandarim-de-Lacerda CA. Developmental origins of health and disease: experimental and human evidence of fetal programming for metabolic syndrome. J Hum Hypertens 2012; 26: 405-419
  • 20 Vaiserman AM. Early-life nutritional programming of type 2 diabetes: Experimental and quasi-experimental evidence. Nutrients 2017; 9: 236
  • 21 Lumey LH, Stein AD, Susser E. Prenatal famine and adult health. Annu Rev Public Health 2011; 32: 237-262
  • 22 Heijmans BT, Tobi EW, Lumey LH, Slagboom PE. The epigenome: archive of the prenatal environment. Epigenetics 2009; 4: 526-531
  • 23 Roseboom TJ, Painter RC, van Abeelen AF, Veenendaal MV, de Rooij SR. Hungry in the womb: What are the consequences? Lessons from the Dutch famine. Maturitas 2011; 70: 141-145
  • 24 Thurner S, Klimek P, Szell M, Duftschmid G, Endel G, Kautzky-Willer A, Kasper DC. Quantification of excess risk for diabetes for those born in times of hunger, in an entire population of a nation, across a century. Proc Natl Acad Sci U S A 2013; 110: 4703-4707
  • 25 Lumey LH, Khalangot MD, Vaiserman AM. Association between type 2 diabetes and prenatal exposure to the Ukraine famine of 1932–33: a retrospective cohort study. Lancet Diabetes Endocrinol 2015; 3: 787-794
  • 26 Koupil I, Shestov DB, Sparén P, Plavinskaja S, Parfenova N, Vågerö D. Blood pressure, hypertension and mortality from circulatory disease in men and women who survived the siege of Leningrad. Eur J Epidemiol 2007; 22: 223-234
  • 27 Wang N, Wang X, Han B, Li Q, Chen Y, Zhu C, Chen Y, Xia F, Cang Z, Zhu C, Lu M, Meng Y, Chen C, Lin D, Wang B, Jensen MD, Lu Y. Is exposure to famine in childhood and economic development in adulthood associated with diabetes?. J Clin Endocrinol Metab 2015; 100: 4514-4523
  • 28 Hult M, Tornhammar P, Ueda P, Chima C, Bonamy AK, Ozumba B, Norman M. Hypertension, diabetes and overweight: Looming legacies of the biafran famine. PLoS One 2010; 5: e13582
  • 29 Keinan-Boker L, Shasha-Lavsky H, Eilat-Zanani S, Edri-Shur A, Shasha SM. Chronic health conditions in Jewish Holocaust survivors born during world war II. Isr Med Assoc J 2015; 17: 206-212
  • 30 Banegas JR, Rodríguez-Artalejo F, de la Cruz JJ, Graciani A, Villar F, del Rey-Calero J. Adult men born in spring have lower blood pressure. J Hypertens 2000; 18: 1763-1766
  • 31 Panchal SK, Brown L. Rodent models for metabolic syndrome research. J Biomed Biotechnol 2011; 2011: 351982
  • 32 Armitage JA, Khan IY, Taylor PD, Nathanielsz PW, Poston L. Developmental programming of the metabolic syndrome by maternal nutritional imbalance: how strong is the evidence from experimental models in mammals?. J Physiol 2004; 561: 355-377
  • 33 Srinivasan M, Katewa SD, Palaniyappan A, Pandya JD, Patel MS. Maternal high-fat diet consumption results in fetal malprogramming predisposing to the onset of metabolic syndrome-like phenotype in adulthood. Am J Physiol Endocrinol Metab 2006; 291: E792-E799
  • 34 Samuelsson AM, Matthews PA, Argenton M, Christie MR, McConnell JM, Jansen EH, Piersma AH, Ozanne SE, Twinn DF, Remacle C, Rowlerson A, Poston L, Taylor PD. Diet-induced obesity in female mice leads to offspring hyperphagia, adiposity, hypertension, and insulin resistance: a novel murine model of developmental programming. Hypertension 2008; 51: 383-392
  • 35 Torrens C, Ethirajan P, Bruce KD, Cagampang FR, Siow RC, Hanson MA, Byrne CD, Mann GE, Clough GF. Interaction between maternal and offspring diet to impair vascular function and oxidative balance in high fat fed male mice. PLoS One 2012; 7: e50671
  • 36 Kelishadi R, Haghdoost AA, Jamshidi F, Aliramezany M, Moosazadeh M. Low birthweight or rapid catch-up growth: which is more associated with cardiovascular disease and its risk factors in later life? A systematic review and cryptanalysis. Paediatr Int Child Health 2015; 35: 110-123
  • 37 Kitsiou-Tzeli S, Tzetis M. Maternal epigenetics and fetal and neonatal growth. Curr Opin Endocrinol Diabetes Obes 2017; 24: 43-46
  • 38 Desai M, Jellyman JK, Ross MG. Epigenomics, gestational programming and risk of metabolic syndrome. Int J Obes 2015; 39: 633-641
  • 39 Erdélyi K, Pacher P, Virág L, Szabó C. Role of poly(ADP-ribosyl)ation in a ‘two-hit’ model of hypoxia and oxidative stress in human A549 epithelial cells in vitro . Int J Mol Med 2013; 32: 339-346
  • 40 Morris CF, Tahir M, Arshid S, Castro MS, Fontes W. Reconciling the IPC and two-hit models: Dissecting the underlying cellular and molecular mechanisms of two seemingly opposing frameworks. J Immunol Res 2015; 2015: 697193
  • 41 Bayer TA, Falkai P, Maier W. Genetic and non-genetic vulnerability factors in schizophrenia: the basis of the “two hit hypothesis”. J Psychiatr Res 1999; 33: 543-548
  • 42 Lahiri DK, Maloney B, Basha MR, Ge YW, Zawia NH. How and when environmental agents and dietary factors affect the course of Alzheimerʼs Disease: the “LEARn” model (latent early-life associated regulation) may explain the triggering of AD. Curr Alzheimer Res 2007; 4: 219-228
  • 43 Lahiri DK, Maloney B, Zawia NH. The LEARn model: an epigenetic explanation for idiopathic neurobiological diseases. Mol Psychiatry 2009; 14: 992-1003
  • 44 Heindel JJ, Balbus J, Birnbaum L, Brune-Drisse MN, Grandjean P, Gray K, Landrigan PJ, Sly PD, Suk W, Cory Slechta D, Thompson C, Hanson M. Developmental origins of health and disease: Integrating environmental influences. Endocrinology 2015; 156: 3416-3421
  • 45 Tsukamoto H, Machida K, Dynnyk A, Mkrtchyan H. “Second hit” models of alcoholic liver disease. Semin Liver Dis 2009; 29: 178-187
  • 46 Howard J. The cytokine hypothesis: a neurodevelopmental explanation for the emergence of schizophrenia later in life. Adv Biosci Biotechnol 2013; 04: 81-88
  • 47 Zhang P, Torres K, Liu X, Liu CG, Pollock RE. An overview of chromatin-regulating proteins in cells. Curr Protein Pept Sci 2016; 17: 401-410
  • 48 Gao J, Cahill CM, Huang X, Roffman JL, Lamon-Fava S, Fava M, Mischoulon D, Rogers JT. S-adenosyl methionine and transmethylation pathways in neuropsychiatric diseases throughout life. Neurotherapeutics 2018; 15: 156-175
  • 49 Curtin K, Samowitz WS, Ulrich CM, Wolff RK, Herrick JS, Caan BJ, Slattery ML. Nutrients in folate-mediated, one-carbon metabolism and the risk of rectal tumors in men and women. Nutr Cancer 2011; 63: 357-366
  • 50 Hardy TM, Tollefsbol TO. Epigenetic diet: impact on the epigenome and cancer. Epigenomics 2011; 3: 503-518
  • 51 Williams SR, Yang Q, Chen F, Liu X, Keene KL, Jacques P, Chen WM, Weinstein G, Hsu FC, Beiser A, Wang L, Bookman E, Doheny KF, Wolf PA, Zilka M, Selhub J, Nelson S, Gogarten SM, Worrall BB, Seshadri S. Sale MM; Genomics and Randomized Trials Network, Framingham Heart Study. Correction to genome-wide meta-analysis of homocysteine and methionine metabolism identifies five one carbon metabolism loci and a novel association of ALDH1L1 with ischemic stroke. PLoS Genet 2014; 10: e1004214
  • 52 Zhu Z, Cao F, Li X. Epigenetic programming and fetal metabolic programming. Front Endocrinol (Lausanne) 2019; 10: 764
  • 53 Jablonka E, Lamb MJ. The inheritance of acquired epigenetic variations. Int J Epidemiol 2015; 44: 1094-1103
  • 54 Lee HS. Impact of maternal diet on the epigenome during in utero life and the developmental programming of diseases in childhood and adulthood. Nutrients 2015; 7: 9492-9507
  • 55 Lee PS, Chiou YS, Ho CT, Pan MH. Chemoprevention by resveratrol and pterostilbene: Targeting on epigenetic regulation. Biofactors 2018; 44: 26-35
  • 56 Chango A, Pogribny IP. Considering maternal dietary modulators for epigenetic regulation and programming of the fetal epigenome. Nutrients 2015; 7: 2748-2770
  • 57 Vahid F, Zand H, Nosrat-Mirshekarlou E, Najafi R, Hekmatdoost A. The role dietary of bioactive compounds on the regulation of histone acetylases and deacetylases: A review. Gene 2015; 562: 8-15
  • 58 Sun Y, Mukai Y, Tanaka M, Saito T, Sato S, Kurasaki M. Green tea extract increases mRNA expression of enzymes which influence epigenetic marks in newborn female offspring from undernourished pregnant mother. PLoS One 2013; 8: e74559
  • 59 Lou XD, Wang HD, Xia SJ, Skog S, Sun J. Effects of resveratrol on the expression and DNA methylation of cytokine genes in diabetic rat aortas. Arch Immunol Ther Exp (Warsz) 2014; 62: 329-340
  • 60 Yun JM, Jialal I, Devaraj S. Epigenetic regulation of high glucose-induced proinflammatory cytokine production in monocytes by curcumin. J Nutr Biochem 2011; 22: 450-458
  • 61 Joven J, Espinel E, Rull A, Aragonès G, Rodríguez-Gallego E, Camps J, Micol V, Herranz-López M, Menéndez JA, Borrás I, Segura-Carretero A, Alonso-Villaverde C, Beltrán-Debón R. Plant-derived polyphenols regulate expression of miRNA paralogs miR-103/107 and miR-122 and prevent diet-induced fatty liver disease in hyperlipidemic mice. Biochim Biophys Acta 2012; 1820: 894-899
  • 62 Ross SA, Davis CD. The emerging role of microRNAs and nutrition in modulating health and disease. Annu Rev Nutr 2014; 34: 305-336
  • 63 Fennell CW, Lindsey KL, McGaw LJ, Sparg SG, Stafford GI, Elgorashi EE, Grace OM, van Staden J. Assessing African medicinal plants for efficacy and safety: pharmacological screening and toxicology. J Ethnopharmacol 2004; 94: 205-217
  • 64 Harborne JB. Classes and Functions of secondary Products from Plants. In: Walton JN, Brown DE. eds. Chemicals from Plants: Perspectives on Plant Secondary. New Jersey: World Scientific; 1999: 1-25
  • 65 Saxena M, Saxena J, Nema R, Singh D, Gupta A. Phytochemistry of medicinal plants. J Pharmacogn Phytochem 2013; 1: 168-182
  • 66 Harborne JB, Baxter HE. Phytochemical Dictionary: A Handbook of Bioactive Compounds from Plants. London: Taylor & Francis; 1993
  • 67 Dillard CJ, German BJ. Phytochemicals: nutraceuticals and human health. J Sci Food Agric 2000; 80: 1744-1756
  • 68 Kopin L, Lowenstein C. In the clinic. Dyslipidemia. Ann Intern Med 2017; 167: ITC81-ITC96
  • 69 Hachul ACL, Boldarine VT, Neto NIP, Moreno MF, Ribeiro EB, do Nascimento CMO, Oyama LM. Maternal consumption of green tea extract during pregnancy and lactation alters offspringʼs metabolism in rats. PLoS One 2018; 13: e0199969
  • 70 Zhou L, Xiao X, Zhang Q, Zheng J, Deng M. Maternal genistein intake mitigates the deleterious effects of high-fat diet on glucose and lipid metabolism and modulates gut microbiota in adult life of male mice. Front Physiol 2019; 10: 985
  • 71 Liu J, Iqbal A, Raslawsky A, Browne RW, Patel MS, Rideout TC. Influence of maternal hypercholesterolemia and phytosterol intervention during gestation and lactation on dyslipidemia and hepatic lipid metabolism in offspring of Syrian golden hamsters. Mol Nutr Food Res 2016; 60: 2151-2160
  • 72 Ibrahim KG, Chivandi E, Mojiminiyi FBO, Erlwanger KH. The response of male and female rats to a high-fructose diet during adolescence following early administration of Hibiscus sabdariffa aqueous calyx extracts. J Dev Orig Health Dis 2017; 8: 628-637
  • 73 Oikari S, Ahtialansaari T, Heinonen MV, Mauriala T, Auriola S, Kiehne K, Fölsch UR, Jänne J, Alhonen L, Herzig KH. Downregulation of PPARs and SREBP by acyl-CoA-binding protein overexpression in transgenic rats. Pflugers Arch 2008; 456: 369-377
  • 74 Wang Y, Viscarra J, Kim SJ, Sul HS. Transcriptional regulation of hepatic lipogenesis. Nat Rev Mol Cell Biol 2015; 16: 678-689
  • 75 Supruniuk E, Miklosz A, Chabowski A. The implication of PGC-1α on fatty acid transport across plasma and mitochondrial membranes in the insulin sensitive tissues. Front Physiol 2017; 8: 923
  • 76 Planavila A, Redondo I, Hondares E, Vinciguerra M, Munts C, Iglesias R, Gabrielli LA, Sitges M, Giralt M, van Bilsen M, Villarroya F. Fibroblast growth factor 21 protects against cardiac hypertrophy in mice. Nat Commun 2013; 4: 2019
  • 77 Ching RH, Yeung LO, Tse IM, Sit WH, Li ET. Supplementation of bitter melon to rats fed a high-fructose diet during gestation and lactation ameliorates fructose-induced dyslipidemia and hepatic oxidative stress in male offspring. J Nutr 2011; 141: 1664-1672
  • 78 Leff T. AMP-activated protein kinase regulates gene expression by direct phosphorylation of nuclear proteins. Biochem Soc Trans 2003; 31: 224-227
  • 79 Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, Wu M, Ventre J, Doebber T, Fujii N, Musi N, Hirshman MF, Goodyear LJ, Moller DE. Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest 2001; 108: 1167-1174
  • 80 Matumba MG, Ayeleso AO, Nyakudya T, Erlwanger K, Chegou NN, Mukwevho E. Long-term impact of neonatal intake of oleanolic acid on the expression of AMP-activated protein kinase, adiponectin and inflammatory cytokines in rats fed with a high fructose diet. Nutrients 2019; 11: 226
  • 81 Schwiertz A, Taras D, Schäfer K, Beijer S, Bos NA, Donus C, Hardt PD. Microbiota and SCFA in lean and overweight healthy subjects. Obesity (Silver Spring) 2010; 18: 190-195
  • 82 Gomez-Arango LF, Barrett HL, McIntyre HD, Callaway LK, Morrison M, Dekker Nitert M. SPRING Trial Group. Connections between the gut microbiome and metabolic hormones in early pregnancy in overweight and obese women. Diabetes 2016; 65: 2214-2223
  • 83 Jung MJ, Lee J, Shin NR, Kim MS, Hyun DW, Yun JH, Kim PS, Whon TW, Bae JW. Chronic repression of mTOR complex 2 induces changes in the gut microbiota of diet-induced obese mice. Sci Rep 2016; 6: 30887
  • 84 Canfora EE, Jocken JW, Blaak EE. Short-chain fatty acids in control of body weight and insulin sensitivity. Nat Rev Endocrinol 2015; 11: 577-591
  • 85 Tremaroli V, Bäckhed F. Functional interactions between the gut microbiota and host metabolism. Nature 2012; 489: 242-249
  • 86 Huang C, Qiao X, Dong B. Neonatal exposure to genistein ameliorates high-fat diet-induced non-alcoholic steatohepatitis in rats. Br J Nutr 2011; 106: 105-113
  • 87 Mukonowenzou NC, Dangarembizi R, Chivandi E, Nkomozepi P, Erlwanger KH. Administration of ursolic acid to new-born pups prevents dietary fructose-induced non-alcoholic fatty liver disease in Sprague Dawley rats. J Dev Orig Health Dis 2021; 12: 101-112
  • 88 Tran NT, Alexandre-Gouabau MC, Pagniez A, Ouguerram K, Boquien CY, Winer N, Darmaun D. Neonatal citrulline supplementation and later exposure to a high fructose diet in rats born with a low birth weight: A preliminary report. Nutrients 2017; 9: 375
  • 89 Jegatheesan P, Beutheu S, Ventura G, Nubret E, Sarfati G, Bergheim I, De Bandt JP. Citrulline and nonessential amino acids prevent fructose-induced nonalcoholic fatty liver disease in rats. J Nutr 2015; 145: 2273-2279
  • 90 Jegatheesan P, Beutheu S, Ventura G, Sarfati G, Nubret E, Kapel N, Waligora-Dupriet AJ, Bergheim I, Cynober L, De-Bandt JP. Effect of specific amino acids on hepatic lipid metabolism in fructose-induced non-alcoholic fatty liver disease. Clin Nutr 2016; 35: 175-182
  • 91 Petersen MC, Shulman GI. Mechanisms of insulin action and insulin resistance. Physiol Rev 2018; 98: 2133-2223
  • 92 Kahn SE. The relative contributions of insulin resistance and beta-cell dysfunction to the pathophysiology of Type 2 diabetes. Diabetologia 2003; 46: 1707
  • 93 Czech MP. Insulin action and resistance in obesity and type 2 diabetes. Nat Med 2017; 23: 804-814
  • 94 Gayoso-Diz P, Otero-González A, Rodriguez-Alvarez MX, Gude F, García F, De Francisco A, Quintela AG. Insulin resistance (HOMA-IR) cut-off values and the metabolic syndrome in a general adult population: effect of gender and age: EPIRCE cross-sectional study. BMC Endocr Disord 2013; 13: 47
  • 95 Wu Z, Zhao J, Xu H, Lyv Y, Feng X, Fang Y, Xu Y. Maternal quercetin administration during gestation and lactation decrease endoplasmic reticulum stress and related inflammation in the adult offspring of obese female rats. Eur J Nutr 2014; 1669-1683
  • 96 Ros P, Díaz F, Freire-Regatillo A, Argente-Arizón P, Barrios V, Argente J, Chowen JA. Resveratrol intake during pregnancy and lactation modulates the early metabolic effects of maternal nutrition differently in male and female offspring. Endocrinology 2018; 159: 810-825
  • 97 Lembede BW, Erlwanger KH, Nkomozepi P, Chivandi E. Effect of neonatal orally administered S-allyl cysteine in high-fructose diet fed Wistar rats. J Dev Orig Health Dis 2018; 9: 160-171
  • 98 Lembede BW, Joubert J, Nkomozepi P, Erlwanger KH, Chivandi E. Insulinotropic effect of S-Allyl cysteine in rat pups. Prev Nutr Food Sci 2018; 23: 15-21
  • 99 Clancy B, Darlington RB, Finlay BL. Translating developmental time across mammalian species. Neuroscience 2001; 105: 7-17
  • 100 Ibrahim KG, Chivandi E, Nkomozepi P, Matumba MG, Mukwevho E, Erlwanger KH. The long-term protective effects of neonatal administration of curcumin against nonalcoholic steatohepatitis in high-fructose-fed adolescent rats. Physiol Rep 2019; 7: e14032
  • 101 Nyakudya TT, Mukwevho E, Nkomozepi P, Erlwanger KH. Neonatal intake of oleanolic acid attenuates the subsequent development of high fructose diet-induced non-alcoholic fatty liver disease in rats. J Dev Orig Health Dis 2018; 9: 500-510
  • 102 Hanson M, Gluckman P. Developmental origins of noncommunicable disease: population and public health implications. Am J Clin Nutr 2011; 94: 1754S-1758S
  • 103 Kett MM, Denton KM. Renal programming: cause for concern?. Am J Physiol Regul Integr Comp Physiol 2011; 300: R791-R803
  • 104 Williams L, Seki Y, Vuguin PM, Charron MJ. Animal models of in utero exposure to a high fat diet: a review. Biochim Biophys Acta 2014; 1842: 507-519
  • 105 Tain YL, Hsu CN. Interplay between oxidative stress and nutrient sensing signaling in the developmental origins of cardiovascular disease. Int J Mol Sci 2017; 18: 841
  • 106 Tain YL, Hsu CN. Targeting on asymmetric dimethylarginine-related nitric oxide-reactive oxygen species imbalance to reprogram the development of hypertension. Int J Mol Sci 2016; 17: 2020
  • 107 Hsu CN, Lin YJ, Tain YL. Maternal exposure to bisphenol A combined with high-fat diet-induced programmed hypertension in adult male rat offspring: Effects of resveratrol. Int J Mol Sci 2019; 20: 4382
  • 108 Chen HE, Lin YJ, Lin IC, Yu HR, Sheen JM, Tsai CC, Huang LT, Tain YL. Resveratrol prevents combined prenatal NG-nitro-L-arginine-methyl ester (L-NAME) treatment plus postnatal high-fat diet induced programmed hypertension in adult rat offspring: interplay between nutrient-sensing signals, oxidative stress and gut microbiota. J Nutr Biochem 2019; 70: 28-37
  • 109 de Bem GF, da Costa CA, de Oliveira PR, Cordeiro VS, Santos IB, de Carvalho LC, Souza MA, Ognibene DT, Daleprane JB, Sousa PJ, Resende AC, de Moura RS. Protective effect of Euterpe oleracea Mart (açaí) extract on programmed changes in the adult rat offspring caused by maternal protein restriction during pregnancy. J Pharm Pharmacol 2014; 66: 1328-1338
  • 110 Liang W, Menke AL, Driessen A, Koek GH, Lindeman JH, Stoop R, Havekes LM, Kleemann R, van den Hoek AM. Establishment of a general NAFLD scoring system for rodent models and comparison to human liver pathology. PLoS One 2014; 9: e115922
  • 111 Castro GS, Cardoso JF, Vannucchi H, Zucoloto S, Jordão AA. Fructose and NAFLD: metabolic implications and models of induction in rats. Acta Cir Bras 2011; 26: 45-50
  • 112 Dietrich P, Hellerbrand C. Non-alcoholic fatty liver disease, obesity and the metabolic syndrome. Best Pract Res Clin Gastroenterol 2014; 28: 637-653

Zoom Image
Fig. 1 Influence of medicinal plants and phytochemicals on metabolic programming. The figure illustrates the epigenetic mechanisms by which phytochemicals or extracts of medicinal plants may upregulate or downregulate the expression of genes associated with metabolism or its control, thereby preventing the development of MetS induced by a high-fructose or high-fat diet. HFRT = high fructose, HF = high fat, RNA = ribonucleic acid.