Next Article in Journal
Healthy Diets from Sustainable Food Systems: Calculating the WISH Scores for Women in Rural East Africa
Next Article in Special Issue
High Fat Diet and Polycystic Ovary Syndrome (PCOS) in Adolescence: An Overview of Nutritional Strategies
Previous Article in Journal
The Association between Serum Vitamin D Levels and Urinary Tract Infection Risk in Children: A Systematic Review and Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Maternal High-Fat Diet Controls Offspring Kidney Health and Disease

1
Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
2
Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
3
School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
4
Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
5
College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2023, 15(12), 2698; https://doi.org/10.3390/nu15122698
Submission received: 5 May 2023 / Revised: 4 June 2023 / Accepted: 8 June 2023 / Published: 9 June 2023
(This article belongs to the Special Issue High-Fat Diet in High-Risk Population)

Abstract

:
A balanced diet during gestation is critical for fetal development, and excessive intake of saturated fats during gestation and lactation is related to an increased risk of offspring kidney disease. Emerging evidence indicates that a maternal high-fat diet influences kidney health and disease of the offspring via so-called renal programming. This review summarizes preclinical research documenting the connection between a maternal high-fat diet during gestation and lactation and offspring kidney disease, as well as the molecular mechanisms behind renal programming, and early-life interventions to offset adverse programming processes. Animal models indicate that offspring kidney health can be improved via perinatal polyunsaturated fatty acid supplementation, gut microbiota changes, and modulation of nutrient-sensing signals. These findings reinforce the significance of a balanced maternal diet for the kidney health of offspring.

1. Introduction

The public health debate on dietary fat and health has been continuing for more than half a century. Most epidemiological studies have linked high consumption of fats, especially saturated fats, to an increased risk of cardiometabolic disorder [1]. Nevertheless, direct evidence of the benefits of lipid-lowering by altering dietary fat composition is lacking. Although dietary advice recommends lowering the total fat content [2], the types of fats must be taken into consideration.
The rising incidence of kidney disease is a global public health challenge that influences all age groups [3,4]. As adult kidney disease can originate in early life [5,6], prevention of kidney disease must begin as early as in the fetal or childhood stage [7]. During development, the kidneys can adapt to environmental stimuli through structural or functional alterations, i.e., developmental origins of health and disease (DOHaD) or developmental programming [8,9].
The most well-known structural alteration is a low nephron number. As the basic functional unit of the kidney, a reduction in nephrons can have a major impact on renal programming. A low nephron endowment formed in utero can lead to glomerular hyperfiltration and compensatory glomerular hypertrophy, consequently initiating kidney dysfunction and adult kidney disease [10]. Likewise, renal programming of tubular function may also result in renal dysfunction later in life [11].
Recently, several factors have been reported to contribute to renal programming, including maternal diseases, improper nutrition, medication use, toxic substance exposure, exogenous stress, and infection [6,8,9]. Maternal nutrition is the key modifiable factor that may be targeted for controlling kidney disease [12,13,14]. Today, excessive dietary saturated fat intake has increased attention directed toward discovering how a high-fat diet increases the risk of developing kidney disease [15,16,17].
Considering that precisely monitoring food intake and manipulating the diet of pregnant women is challenging in human research, animal models offer an invaluable tool to control dietary fat composition and discover the molecular pathways participating in developmental programming [18,19,20,21]. Many animal studies have associated maternal high-fat diets with alterations to structure and function in fetal tissues/organs giving rise to various adult diseases in later life [18,19,20,21]. These phenotypes cover hypertension, adipocyte hypertrophy, dyslipidemia, obesity, increased visceral fat mass, hepatic steatosis, and insulin resistance [6,7,8,9,10]. However, kidney disease has received relatively less attention.
Controlling maternal diet can also be advantageous for renal programming [22]. Multiple reports have revealed that developmental programming of adult disease can be reversible through nutritional interventions during pregnancy and lactation by reprogramming [22,23,24]. Although polyunsaturated fatty acids (PUFAs) are shown to have a broad spectrum of health benefits against several diseases including kidney disease [25,26], how maternal PUFA supplementation can help avert offspring kidney disease remains largely unclear.
Therefore, this review aims to provide an overview of the roles played by maternal exposure to a high-fat diet in offspring kidney health and disease (Figure 1). Our literature review was performed by searching the databases MEDLINE, Cochrane Library, and Embase using keywords related to a maternal high-fat diet, DOHaD, and kidney disease to survey and establish the evidence in this regard. We used the following search terms: “high-fat diet”, “cholesterol”, “triglyceride”, “fatty acid”, “lipid’, “polyunsaturated fatty acid”, “DOHaD”, “reprogramming”, “developmental programming”, “kidney disease”, “mother”, “pregnancy”, “gestation”, “lactation”, “offspring”, “progeny”, and “hypertension”. We also examined reference lists of articles to detect any extra references that would be related to this review. The last search was made on 30 April 2023.

2. Fats in Pregnancy and Kidney Disease

2.1. Dietary Fats

Dietary fats are mostly triglycerides [1]. In general, we call the triglycerides in our food “fats” and “oils”. Fats are solid lipids, whereas oils are liquid at room temperature. Fats belong to the triglycerides group, which is a subclass of lipids. The main difference between lipids and fats is that lipids are a broad group of biomolecules, while fats are a type of lipids.
Fatty acids and glycerol are the building blocks of triglycerides. Dietary fatty acids are categorized into four common types: saturated, monounsaturated, polyunsaturated, and trans fats (Figure 1). Based on carbon chain length (6–24 carbon units) and degree of saturation, fatty acids differ from each other. Saturated fatty acids are saturated with hydrogen with only single bonds, whereas unsaturated fatty acid chains have one (i.e., monounsaturated) or more double bonds (i.e., polyunsaturated) in their carbon chains. The double bonds can be in a cis (same side) or trans (opposite side) position. Naturally occurring fatty acids usually have a cis configuration. By contrast, trans fats are a type of unsaturated fat that originates from artificial or natural sources. Natural trans fats derived from ruminant animals are safe in moderation, but artificial ones may lead to health issues. In addition, polyunsaturated fatty acids (PUFAs) with double bonds that are three carbon atoms (n-3; e.g., eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)) or six carbons (n-6; e.g., arachidonic acid) from the N-terminal end of the fatty acid are considered essential fatty acids.
Various fatty acids have individual biochemical properties and, thus, are able to produce physiological functions. In general, saturated fatty acids and trans fats are linked to an increased risk of cardiovascular disease (CVD). Monounsaturated and polyunsaturated fatty acids are connected with a decreased risk of CVD [27].
In the body, fats confer significant properties on the cell membrane and are mediators of intra- and inter-cellular signaling [16]. In the gut, dietary fats are absorbed, where fatty acids can be esterified with glycerol to produce triglycerides. Cholesteryl esters are generated via the esterification of long-chain fatty acids with cholesterol.
As cholesterol and triglycerides are insoluble in water, these lipids must be transported by lipoproteins. Plasma lipoproteins can be divided into different classes based on size, lipid composition, and apolipoproteins. Chylomicrons are large triglyceride-rich particles made by the intestine. The removal of triglyceride from chylomicrons by peripheral tissues results in smaller particles known as chylomicron remnants. Very low-density lipoproteins (VLDL) are triglyceride-rich particles made in the liver. VLDL can be further metabolized to low-density lipoprotein (LDL). LDL, when enriched with cholesterol, transports cholesterol to the liver for removal from the organism, whereas the accumulation of oxidatively modified LDL can initiate pathological processes in peripheral tissues. Conversely, high-density lipoprotein (HDL) has cardioprotective properties that operate via reverse cholesterol transport, by which the body removes excess cholesterol from peripheral tissues and delivers them to the liver [28]. HDL particles are enriched with cholesterol.

2.2. Fats and Kidney Health

Circulating triglycerides and cholesterol are transported within lipoprotein particles, whereas free fatty acids require albumin as their transporter. Lipids present in the kidneys include triglycerides, cholesterol, free fatty acids, and phospholipids [16]. Renal tubular cells take up circulating free fatty acids disassociated from albumin through specific membrane proteins, for example, fatty-acid-binding protein and fatty acid translocase [29]. These lipids enter the mitochondria, where they are metabolized to yield ATP, thus sustaining energy balance in the tubules [30]. Although lipid metabolism in renal tubular cells protects the kidney against damage under physiological conditions, excess lipid accumulation may cause kidney damage in the tubule cells [17].
Prior work has indicated that the risk of the development of chronic kidney disease (CKD) increases with high levels of triglycerides [31], LDL cholesterol (LDL-C) [32], and total cholesterol [33], together with low levels of HDL cholesterol (HDL-C) [34]. Lipid overload and impaired fatty acid β-oxidation (FAO) are able to trigger oxidative stress, inflammation, and renal fibrosis [16]. However, different fatty acids may differentially affect mitochondrial function and kidney health. The saturated fatty acid palmitate has been reported to induce mitochondrial stress and kidney damage, whereas the monounsaturated fatty acid (MUFA) oleate increases FAO, which can protect against saturated fatty-acid-induced kidney damage [35].
Fat accumulation in the kidneys can reduce kidney function in several ways, including impaired renal hemodynamics, increased sodium reabsorption and renin secretion, and activation of the renin-angiotensin-aldosterone system (RAAS) [36]. Increased volumes of perirenal fat might compress the loop of Henle and the vasa recta of the renal medulla, leading to a reduction in tubular flow rate [37]. A reduction in NaCl concentration in the macula densa cells can stimulate renin secretion [38]. Activation of the RAAS, beginning with renin secretion, can further stimulate renal tubular sodium reabsorption. Indeed, fats have a great influence on kidney health and disease [39]. We will not attempt a detailed discussion here, as these observations have been reviewed elsewhere [15,36,37].

2.3. Fats and Fetal Development

Maternal diet might alter lipid uptake, lipid transport, and lipid-sensing signals in the developing fetal kidneys, resulting in renal programming (Figure 1). During pregnancy, the fetus requires a significant number of fatty acids and cholesterol. For structural purposes, the fetus needs 1.5 mg of cholesterol per gram of tissue [40]. Fatty acids are required as structural components of tissues, as a source of energy, and as activators of transcription factors [40]. In gestational diabetes, maternal plasma fatty acid levels correlate with fetal lipids, fetal growth, and fat mass [41]. In addition, impaired placental transfer of lipophilic compounds has been shown to be related to intrauterine growth restriction [42]. These observations suggest that lipid metabolism during pregnancy has a role in fetal growth and development [42].
Lipid metabolism involves the uptake of lipids in the gut, the synthesis and degradation of lipids in cells, and transport to compartments such as mitochondria. Phosphoinositides are regulators of key sub-cellular processes including cytoskeletal function, membrane transport, and plasma membrane signaling. The kidney relies on phosphoinositides for physiological processes, such as filtration, solute reabsorption, cell polarization, and signal transduction [43]. It is known that mutations of the genes encoding the phosphoinositide system in the kidney very often result in human genetic kidney diseases, such as Joubert syndrome and Lowe syndrome [43]. Nevertheless, no information is available regarding their impact on renal programming.
Several lipid-sensing nuclear receptors, including peroxisome-proliferator-activated receptors (PPARs), liver X receptors (LXRs), and PPARγ coactivator-1α (PGC-1α), influence all aspects of lipid metabolism [44]. Previously, our data demonstrated that several PPAR target genes are involved in renal programming and hypertension, such as Ren, Nrf2, Sod2, Nos2, Nos3, Sirt7, and Sgk1 [45]. Since PPARs play a critical role in the pathophysiology of kidney disease [46], it is possible that dysregulated lipid sensing induced by a maternal high-fat diet, such as through the dysregulation of PPARs, may have a close link to renal programming.
Several lines of evidence support the hypothesis that a maternal high-fat diet might be involved in the pathogenesis of renal programming. First, a previous study revealed that high-fat-intake-induced renal injury is related to a decrease in renal Pax2 expression [47]. Our prior work indicated that several nephrogenesis genes related to reduced nephron numbers are PPAR target genes—for example, Pax2 [45]. Second, PPARγ was reported to directly regulate a vast array of genes involved in oxidative stress, including Nos2, Nos3, Sod2, and Nrf2 [48]. Emerging evidence supports the hypothesis that oxidative stress has a critical role in renal programming [6,8,9]; we will discuss this in detail later. Third, it has been observed that several PPAR target genes are RAAS components or sodium transporters. PPARγ has been reported to stimulate renin gene expression [49] and to increase sodium hydrogen exchanger-3 (NHE3) [50].
Free fatty acids are ligands for G-protein-coupled receptors (GPR), which are also referred to as free fatty acid receptors (FFAR) [51]. Short-chain fatty acids (SCFAs) are generated from dietary fiber through fermentation via gut microbes and mainly contain acetate, butyrate, and propionate [51]. SCFAs are capable of activating GPR41 and GPR43, whereas long-chain fatty acids can activate GPR40 and GPR120. SCFAs and their receptors play an important role in maternal metabolism and fetal programming [52]. As lipid signaling has been related to fetal programming, it is increasingly important to better identify the actions of maternal exposure to a high-fat diet on lipid signaling and to have the ability to identify mechanisms underlying renal programming.
Although previous studies reported alterations in kidney structure, i.e., reduced nephron numbers, in offspring exposed to nutritional imbalance during pregnancy and lactation [6], current literature offers little or no understanding of this mechanism in a maternal high-fat diet. However, prior investigations support the hypothesis that a maternal high-fat diet affects the offspring’s renal transcriptome. Aberrant gene expression of several molecular pathways (e.g., PPARs) in the developing kidney may contribute to nephron deficit, dysregulated RAAS, increased sodium transporters, and increased BP [12,13]. All of these mechanisms underlying renal programming are deleterious to future kidney health.

3. Renal Programming: The Impact of a Maternal High-Fat Diet

Currently, little reliable information is available regarding whether high fat intake during gestation and lactation leads to adulthood kidney disease in humans. Most pooled epidemiological studies that recruit diverse participants, along with investigating different types of fats from various dietary sources, carry a high possibility of diluting any real findings. Therefore, animal models provide a means to understand the underlying mechanisms of maternal high-fat-diet-induced programming effects.
Although a high-fat diet has long been recognized as a cause of obesity and related disorders in animal models [18,19,20,21] and the term is in frequent usage, it lacks a precise definition. Different high-fat diets with fat proportions ranging from 20 to 60% energy as fat have been developed in animal models. Additionally, the fat component can vary from animal-derived fats (e.g., butter or lard) to plant oils (e.g., corn or coconut oil) [18,19,20,21,53,54]. Moreover, diets rich in saturated or unsaturated fats might have different impacts on health risks [1]. As this dietary intervention is not standardized, phenotypes induced by a maternal high-fat diet may differ markedly among various animal studies [53,54].
Several animal species have been used to elucidate the effect of a maternal high-fat diet on the progeny, covering non-human primates [55], pigs [56], rabbits [57], rats [20], and mice [20]. As we and others reviewed elsewhere [16,19,31,32], offspring exposed to a maternal high-fat diet may have altered feeding habits, effects on their body composition, reduced cognitive development, increased risk of type 2 diabetes, obesity, insulin resistance, liver steatosis, dyslipidemia, hypertension, etc.
Although a growing number of animal studies have been reported to determine the impact of a maternal high-fat diet on offspring outcomes [20,21,58,59], only a few studies evaluated renal programming. In this review, we only considered studies restricting exposure to cover the period of nephrogenesis. In rodents, kidney development is roughly from mid-pregnancy to mid-lactation. Table 1 summarizes preclinical studies recording offsprings’ renal outcomes in which maternal high-fat diets were applied during gestation and lactation [60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75]. Although maternal obesity is frequently studied using rodents on high-fat diets, it is clear that the programming effects of maternal obesity and high fat consumption on offspring outcomes are different [76]. Studies in which high-fat diets were fed to rodents to induce maternal obesity usually started the diets at 4–9 weeks before pregnancy.
Table 1 shows maternal high-fat diets with different fat proportions ranging from 20 to 58% energy as fat, which is in good agreement with previous studies [18]. However, the high-fat diets used most frequently with rodents did not closely match Western diets, as the latter are lower in fats and protein [76]. In addition to purified high-fat diets, the utilization of a human Western diet, a Western-style diet, or a cafeteria diet has been conducted for metabolic diseases [77,78]. However, none has been applied to study renal programming in this regard.
Rats and mice are the most frequently used species. Adverse renal outcomes in offspring are mainly induced by a maternal diet enriched with saturated fat, such as lard, palm oil, and coconut oil. As presented in Table 1, the influence of a maternal high-fat diet on rat offspring was evaluated from the age of 9 weeks to 6 months. The rodent ages in Table 1 correspond to human ages from adolescence to young adulthood [79]. These renal-programming-related phenotypes cover tubular dysfunction [60,62,65], renal hypertrophy [65], renal function impairment [67,69], proteinuria [67,68,69], renal fibrosis [68,69], and hypertension [70,71,72,73,74,75]. Notably, maternal high-fat-diet-induced renal phenotypes vary, mostly according to age, species, and varied fatty acid fractions and compositions.
Of note is that kidney disease can be attributed to multiple “hits” [80]. As reported in the DOHaD research, lifelong health can be adversely affected by a series of “hits” experienced at critical developmental periods and across the lifespan [81]. “First hits” are adverse insults experienced by the mother that make the offspring more vulnerable to adult disease. Postnatal insults then present “second hits”, through which prenatally primed vulnerability can be triggered or exacerbated. In some studies, a maternal high-fat diet was applied as the first hit, followed by a second hit to induce kidney disease in later life. For instance, animal models of a maternal and postnatal high-fat diet [82] and a combined maternal high-fat, high-sucrose, and high-salt diet [83] have been used to study renal programming. Another hit may trigger the same programming mechanisms and amplify adverse actions culminating in a disease state. Together, animal models with various types of maternal high-fat diets support the hypothesis that such diets have programming effects on the kidneys of the offspring.

4. Mechanisms Linking Maternal High-Fat Diets to Renal Programming

To date, several hypothetical mechanisms have been reported to be bound up with renal programming [6,8,9,12]. Among them, oxidative stress, deficient nitric oxide (NO), aberrant activation of the RAAS, disrupted nutrient-sensing signals, dysbiotic gut microbiota, inflammation, and dysregulated hydrogen sulfide (H2S) signaling are interrelated with maternal exposure to a high-fat diet and will be discussed in turn (Figure 2).

4.1. Oxidative Stress

Oxidative stress, an imbalance between pro- and antioxidant capacity, has been implicated in renal programming [84]. During pregnancy, the developing kidney is vulnerable to overproduction of reactive oxygen species (ROS) under suboptimal intrauterine conditions owing to the deficient antioxidant capacity in the fetus [85]. As we reviewed elsewhere [84], multiple animal models indicated various maternal insults can induce oxidative-stress-related renal programming.
The mechanistic linking of oxidative stress to renal programming induced by various types of maternal insults covers increased production of ROS [86], decreased antioxidant capabilities [87], increased lipid peroxidation [88], and increased oxidative damage [89]. Conversely, natural and synthetic antioxidants can serve as reprogramming therapies for kidney diseases of developmental origins [90,91,92].
Table 1 demonstrates that maternal-high-fat-diet-primed renal programming is associated with reduced antioxidant activity [63], increased lipid peroxidation [61,63], and increased oxidative DNA damage [61,68,71,72]. A commonly used marker of DNA damage, 8-hydroxydeoxyguanosine (8-OHdG), has been utilized to identify oxidative damage and has revealed such damage to be augmented in the kidneys of adult rat progeny from dams fed on a diet rich in saturated fats [61,68,71,72]. Although various antioxidants show a potential role for reducing oxidative stress in preventing kidney disease [93], their effects on maternal-high-fat-diet-primed renal programming roles are still largely unclear.

4.2. Deficient NO

In the kidney, NO carries out important physiological and signaling functions, whereas deficient NO implicates the pathogenesis of kidney diseases [94,95]. During pregnancy, NO has a significant role in the regulation of fetoplacental circulation and fetal development [96]. Deficient NO is one of the mechanistic pathways behind renal programming, whereas perinatal use of NO-based interventions has shown benefits which protect against the developmental programming of kidney disease [81]. Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of NO synthase, which competes with l-arginine to decrease NO production [97]. ADMA can cause a nephron deficit in cultured rat embryonic kidneys and alter renal transcriptome [98]. Whether ADMA–NO imbalance contributes to the developmental programming of kidney disease remains to be investigated further [98].
Deficient NO in kidneys [71,72,75] is related to maternal-high-fat-diet-primed renal programming. High fat intake during gestation and lactation results in decreases in plasma l-arginine concentrations and in the l-arginine-to-ADMA ratio, an index of NO bioavailability [75]. One study demonstrated that maternal bisphenol A exposure (BPA) exacerbates maternal high-fat diet-primed hypertension in adult male offspring, which is associated with increased ADMA concentration and a decreased ratio of l-arginine-to-ADMA [99]. In addition, there was a synergistic effect of maternal high-fat diet and BPA exposure on inducing oxidative damage in offspring kidneys. Conversely, resveratrol, a polyphenolic antioxidant, protected adult offspring from a maternal high-fat diet as well as from BPA-induced hypertension and oxidative damage. The protective action of resveratrol is related to the restoration of NO bioavailability [99] Another study using a dexamethasone and high-fat diet two-hit model demonstrated that adult offspring developed hypertension and kidney oxidative damage that coincided with increases in plasma ADMA and decreases in plasma l-arginine-to-ADMA ratios [100]. Maternal antioxidant therapy by N-acetylcysteine could prevent adult offsprings’ hypertension and oxidative damage via restoration of the ADMA–NO balance [100].
As several currently available prescription drugs have the ability to restore the balance of the ADMA–NO pathway [97], additional work is needed to understand the reprogramming actions of NO-based intervention in maternal-high-fat-diet-primed renal programming.

4.3. Aberrant RAAS

The RAAS is a key hormone cascade regulating BP and the renal system [101]. There are two RAAS pathways: the classic and the non-classic systems. The classic RAAS is composed of angiotensin-converting enzyme (ACE), Ang II, and Ang type 1 receptor (AT1R). On the other hand, the ACE2–angiotensin (1–7)–Mas receptor pathway is a counter-regulatory RAAS system to offset the harmful effects of Ang II signaling. One such example is that administration of ACE2 activator or ANG-(1–7) during pregnancy has been reported to attenuate hypertension and kidney fibrosis in adult SHR offspring [102].
Activation of the classic RAAS through high fat intake can lead to vasoconstriction, oxidative stress, and inflammation, resulting in kidney disease [103,104]. Hypertension in maternal-high-fat-diet-primed offspring coincides with aberrant activation of the classic RAAS, represented by increases in the renal protein level of AT1R and mRNA expression of Agt and Ace [64].
Glucose transporter 4 (GLUT4) mediates the uptake of glucose [105]. Ang II can mediate GLUT4, which has a role in insulin resistance and in the development of diabetic kidney disease [106]. Prior research revealed that GLUT4 heterozygous (GLTU4 +/−) mice exhibited insulin resistance [107]. In this GLUT4 +/- mice model, maternal high-fat-diet-induced hypertension in offspring was accompanied by increased renal gene expression of renin and the AT1R [66]. Likewise, another study showed maternal high-fat diet increased renal protein levels of AT1R, as well as mRNA expression of Agt and Ace, in adult rat offspring at 16 weeks of age [108]. Moreover, the non-classic RAAS also participates in renal programming. Another study reported that 16-week-old male rats that are perinatally exposed to a high-fat diet have low Ang-(1–7) levels [109]. ACE2-deficient mice, with low Ang-(1–7) levels, developed hypertension and kidney injury [110]. In the context of experimental kidney diseases, most studies have proposed that the ACE2–angiotensin (1–7)–Mas axis has a protective role [111]. Whether a maternal high-fat diet downregulating the ACE2–angiotensin (1–7)–Mas axis contributes to kidney disease later in life awaits further investigation.
Emerging evidence supports the hypothesis that there is a transient biphasic response with the downregulation of the classic RAAS system in the neonatal period that returns to normal with age [112,113]. A maternal high-fat diet may disrupt this normalization in the adult offspring; thereafter, the classic RAAS system is abnormally activated, whereas the non-classic RAAS axis is downregulated. Considering that renal programming induced by a maternal high-fat diet coincides with aberrant RAAS, it is interesting to elucidate whether targeting RAAS could serve as a reprogramming approach in this regard.

4.4. Disrupted Nutrient-Sensing Signals

Accumulating evidence demonstrates that dietary fat modulates nutrient-sensing signals that are responsible for lipid detection, satiation signals, food intake, and weight gain [114,115]. These nutrient-sensing signals include AMP-activated protein kinase (AMPK) [116], sirtuin-1 (SIRT1) [117], PPAR, and PPARγ coactivator-1α (PGC-1α) [118].
In pregnancy, the maternal diet can regulate fetal metabolism and development via nutrient-sensing signals [119]. Accordingly, an imbalanced diet during gestation could disrupt nutrient-sensing signals, having a decisive impact on adult diseases of developmental origins [120,121].
Maternal-high-fat-diet-primed hypertension is associated with the inhibitory AMPK/SIRT1/PGC-1α pathway in an offspring’s kidneys [71,75]. AMPK can phosphorylate PGC-1α and regulate its downstream PPARγ signaling. Prior work indicated that specific sets of PPAR target genes participate in renal programming [120]. Although several natural and synthetic PPAR agonists have been studied in kidney-related disorders [46,122,123,124], whether PPAR modulators have protective actions against maternal-high-fat-diet-induced adverse renal outcomes in offspring is awaiting further elucidation.

4.5. Gut Microbiota Dysbiosis

The gut microbiome is highly diverse and harbors trillions of microorganisms coexisting with the host, which in turn can determine human health and disease [125]. The shaping and multiplication of gut microbiota start at birth, but the modification of their composition depends on nutritional and environmental factors. Accordingly, maternal dietary nutrients play a key role in the modulation of an offspring’s gut microbiome composition [126].
Current evidence suggests that high saturated fat can lower microbiota richness and diversity [127,128]. Similarly, reduced α-diversity in gut microbiota was noted in adult rat offspring from dams fed on a diet rich in fat [129]. In addition, a maternal high-fat diet inducing hypertension in offspring has been linked to an increased Firmicutes-to-Bacteroidetes (F/B) ratio, which is considered to be a microbial marker for hypertension [130]. Moreover, the reduction in beneficial microbes, a feature of dysbiotic gut microbiota such as Lactobacillus and Akkermansia [131,132], was reduced in the maternal high-fat diet model [70,74].
Microbial metabolites, such as tryptophan-derived metabolites, SCFAs, trimethylamine (TMA), and trimethylamine N-oxide (TMAO), are also involved in the pathogenesis of renal programming [133,134,135]. One study indicated that maternal exposure to a high-fat diet could reduce fecal propionate concentration, an SCFA, in 3-week-old rat progeny [74]. Conversely, perinatal propionate supplementation was shown to protect adult offspring born to mother rats with CKD against hypertension [136]. In addition, a maternal high-fat diet increased TMA concentrations and decreased the TMAO-to-TMA ratio [74]. As the inhibition of microbiota-derived metabolites TMA and TMAO is able to attenuate kidney disease [137], targeting the TMA/TMAO pathway as a reprogramming strategy has been studied in different animal models of renal programming [138,139]. Inhibition of TMA formation by 3,3-Dimethyl-1-butanol (DMB) was reported to reduce plasma TMAO levels in mice fed on a Western diet [140]. Our previous research revealed that maternal DMB treatment prevented high-fructose-diet-induced hypertension in adult offspring via regulating the TMA–TMAO metabolic pathway and reshaping the gut microbiome [138]. Another study demonstrated that maternal CKD led to hypertension and renal hypertrophy in 12-week-old male offspring. These adverse renal programming effects can be prevented by maternal iodomethylcholine (an inhibitor of TMA formation) treatment, which coincides with a reduction in TMAO [139]. However, whether high-fat-diet-primed renal programming can be averted in this regard warrants further investigation.
In experimental and human CKD, the increases in tryptophan-derived uremic toxins from indole and kynurenine pathways participate in the progression of CKD [141,142]. These tryptophan-derived microbial metabolites are endogenous ligands for aryl hydrocarbon receptor (AhR) [143], which can trigger renal inflammation and fibrosis. As high fat intake can activate AhR signaling [144] and that AhR antagonist resveratrol has been associated with the protection of offspring from renal programming [145], more research on the interconnection between a high-fat diet and AhR is required, as they may be a potential reprogramming approach. Together, the findings above suggest that dysbiotic gut microbiota and their derived metabolites might be a probable reason contributing to maternal-high-fat-diet-primed renal programming.

4.6. Inflammation

Inflammation has a role in compromised pregnancies and associated complications [146]. The accumulation of T cells, monocytes/macrophages, and T-cell-derived cytokines are involved in the pathogenesis of hypertension [146].
Activated T cells are able to secrete cytokines, such as tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ), which have been linked to kidney damage and hypertension in pre-clinical models [147].
In CKD, the interplay between inflammation and an imbalance of T regulatory cells (Treg) and T helper 17 (TH17) cells has also been related to hypertension [148]. As several tryptophan-derived uremic toxins are ligands for aryl hydrocarbon receptor (AhR) [149], activation of AhR signaling can initiate inflammation through increasing monocyte adhesion, upregulating proinflammatory gene expression, reducing NO bioavailability, and inducing the expression of endothelial adhesion molecules [150].
High-fat diets increase free fatty acid uptake and overexpression of fatty acid uptake systems such as the CD36 scavenger receptor, promoting renal inflammation and kidney injury [151]. One study showed that adult rat progeny born to dams exposed to TCDD developed hypertension, which is related to the activation of AhR signaling and induction of TH17-dependent renal inflammation [152]. Although activation of AhR contributes to high-fat-diet-induced vascular dysfunction [153], more research is required to gain a comprehensive insight into whether maternal-high-fat diet-induced renal programming is attributed to the induction of TH17- and AhR-mediated inflammation.

4.7. Others

Considering that maternal-high-fat-diet-related offspring phenotypes are a complex phenomenon, there might be other mechanistic pathways behind renal programming—for example, epigenetic regulation and dysregulation of H2S and sodium transporters. We previously found that maternal high-fat diet considerably altered transcriptome in 1-week-old rat offsprings’ kidneys, with females being more sensitive than males [72]. There were 154 upregulated and 97 downregulated genes identified in the kidneys of female offspring. In addition to effects on the kidney, a maternal high-fat diet also causes significant changes in gene expression in the brain [145], placenta [154], and heart [155] in progeny. Whether organ-specific epigenetic regulation may be involved in maternal-high-fat-diet-primed renal programming deserves to be explored further.
Hydrogen sulfide (H2S) is a member of the growing family of gasotransmitters and has emerged as an important signaling molecule in kidney function [156]. Lower H2S levels are observed in many renal pathologies, whereas H2S-related interventions could be used as a reprogramming approach for DOHaD-related disease [157]. A maternal high-fat diet caused low plasma H2S concentrations and renal H2S-releasing activity in male rat offspring [73]. Conversely, therapy with perinatal garlic oil, an H2S donor, protected offspring from hypertension that was programmed by a maternal high-fat diet, which was connected to the restoration of the H2S signaling pathway.
In addition, high maternal fat consumption increased the protein level or activity of sodium transporter in an offspring’s kidney [60,62,72]. Considering that increased expression/activity of sodium transporters participates in the development of kidney disease and hypertension in various models [158,159], whether maternal-high-fat-diet-induced renal programming can be attributed to inappropriate expression/activity of sodium transporters deserves further clarification.

5. Reprogramming Interventions

With a deeper insight into the mechanisms behind maternal-high-fat-diet-induced renal programming, the advances in developing mechanism-targeted strategies hold the potential for reprogramming. Up to now, reprogramming interventions to offset mechanisms governing the developmental programming of kidney disease have covered avoidance of risk factors, lifestyle modification, nutritional supplementation, and pharmacological therapies [160,161].
Because of the adverse effects of a maternal diet rich in saturated fats, a universal approach is required to avoid excessive intake of saturated fats during gestation and lactation and avert kidney disease in offspring. However, dietary supplementation with unsaturated fatty acids during gestation and lactation may have beneficial effects on an offspring’s kidney health.
Feeding pregnant SHRs with a diet enriched with PUFAs during the last week of pregnancy and lactation attenuated the development of hypertension in their male offspring at 16 weeks of age [162]. Another study reported that perinatal omega-3 PUFA supplementation attenuated maternal-high-fat-diet-induced kidney injury and renal programming in female adult offspring [163]. Similarly, supplementing linoleic acid, an omega-6 PUFA, during gestation and lactation can avert offspring hypertension programmed by a maternal high-fat diet [164]. Our previous research also indicated that targeting omega-6 PUFA arachidonic acid can avert maternal-high-fructose-diet-primed renal programming and offspring hypertension [165].
Several other early-life interventions have been utilized as reprogramming approaches to prevent maternal-high-fat-diet-primed renal programming (as listed in Table 1), covering Limosilactobacillus fermentum [64], a SIRT1 activator [68], hydralazine [69], long-chain inulin [70], Lactobacillus casei [70], resveratrol [71], garlic oil [73], and an AMPK activator [75].
Importantly, the gut microbiome is an emerging target for most reprogramming interventions for improving maternal-high-fat-diet-primed renal programming. Probiotics and prebiotics are the most frequently studied gut microbiota-targeted tools. Both have long been acknowledged for their benefits to human health [166,167] and in treating kidney disease [168,169]. Probiotic treatment with Lactobacillus casei [70] and Limosilactobacillus fermentum [64] during gestation and lactation averts offspring hypertension programmed by high maternal fat consumption. Additionally, prebiotic treatment with long-chain inulin protected offspring from maternal-high-fat-diet-induced renal programming and was related to an increased abundance of beneficial microbe Lactobacillus species, increased fecal SCAF concentrations, and reduced plasma TMAO levels [70]. Another study revealed that garlic, a natural prebiotic, offered protection from hypertension to maternal-high-fat-diet-primed offspring, accompanied by increases in α-diversity and abundance of beneficial bacteria Lactobacillus and Bifidobacterium and plasma SCFA concentrations [73].
As a reprogramming strategy, resveratrol has been utilized to avert maternal-high-fat-diet-primed hypertension in offspring by restoration of the SIRT1/AMPK/PGC1-α pathway [71]. Likewise, the use of SIRT1 activator SRT1720 [68] or direct AMPK activator 5-aminoimidazole-4-carboxamide riboside showed beneficial effects in this regard.
Moreover, a previous study demonstrated that low-dose hydrazine treatment can be beneficial in protecting against renal programming [69]. Hydralazine, a BP-lowering agent with DNA demethylating activities [170], was shown to improve kidney fibrosis at low doses independently of BP [171]. In the maternal high-fat diet model, low-dose hydralazine administration during pregnancy can attenuate albuminuria and glomerulosclerosis, and the increased serum concentration of creatinine in adult offspring is possibly due to epigenetic regulation [69].
Of note is that several interventions targeting mechanistic pathways underlying renal programming have been proven to be effective in averting adult-onset kidney diseases with the utilization of different animal models [6,8,9,12]. These interventions consist of l-cysteine [172], l-citrulline [173], N-acetylcysteine [174], melatonin [175], epigallocatechin gallate [176], quabain [177], green tea polyphenol [178], l-carnitine [179], and acetate [180]. Although several early-life interventions have highlighted their potential as an attractive approach to improving kidney health, their efficiency in maternal-high-fat-diet-related renal programming warrants further study.

6. Concluding Remarks

Fats in the maternal diet are like a double-edged sword. Data from preclinical research demonstrate that maternal exposure to a diet enriched with saturated fats is associated with renal programming in adult offspring, including renal function impairment, proteinuria, tubular dysfunction, renal hypertrophy, kidney fibrosis, and hypertension. However, offspring kidney health can be improved via perinatal PUFA supplementation. These findings highlight the importance of a balanced diet during gestation and lactation in determining offsprings’ susceptibility to kidney disease later in life.
The Kidney Diseases Global Outcomes (KDIGO) 2012 guidelines have not provided specific dietary recommendations for fat intake in patients with CKD [181]. Currently, there is still limited direct evidence linking specific high fat intake in pregnant women and kidney disease in their children. Nevertheless, animal models, such as those described above, provide significant insight into the molecular mechanisms behind maternal-high-fat-diet-primed renal programming. We are fully aware that the presented mechanisms in the present review might not cover the whole picture of the programming actions of high fat consumption. Considering that fats can affect various tissues/organs, consequently leading to different phenotypes in adult offspring, additional research into their organ-specific programming effects is a pressing need. In addition, what is missing from the literature is the comparison of diets with different fat levels and contents on the severity of offspring kidney disease. Hence, it remains difficult to draw a definite conclusion from the available literature based on the wide variations in experimental “high-fat diets” at this time.
Regardless of recent advances in developing potential reprogramming approaches targeting gut microbiota and nutrient-sensing signals for renal programming, almost all of them have not been translated into human trials. In summary, fats in the maternal diet tightly control offspring kidney health and disease. After a greater understanding of maternal-high-fat-induced renal programming, we expect that translating preclinical results into optimal clinical practice is a valuable strategy that could reduce the global burden of kidney disease.

Author Contributions

Data curation, C.-N.H., H.-Y.L., C.-H.L. and Y.-L.T.; funding acquisition, Y.-L.T.; writing—original draft, C.-N.H., H.-Y.L., C.-H.L. and Y.-L.T.; conceptualization, H.-Y.L., C.-H.L., C.-N.H. and Y.-L.T.; writing—review and editing, H.-Y.L., C.-H.L., C.-N.H. and Y.-L.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received grants CMRPG8N0171, CMRPG8M1371, CORPG8N0091, and CORPG8N0121 from Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Forouhi, N.G.; Krauss, R.M.; Taubes, G.; Willett, W. Dietary fat and cardiometabolic health: Evidence, controversies, and consensus for guidance. Bmj 2018, 361, k2139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Nettleton, J.A.; Lovegrove, J.A.; Mensink, R.P.; Schwab, U. Dietary Fatty Acids: Is it Time to Change the Recommendations? Ann. Nutr. Metab. 2016, 68, 249–257. [Google Scholar] [CrossRef] [PubMed]
  3. Luyckx, V.A.; Tonelli, M.; Stanifer, J.W. The global burden of kidney disease and the sustainable development goals. Bull. World Health Organ. 2018, 96, 414–422. [Google Scholar] [CrossRef] [PubMed]
  4. Lozano, R.; Naghavi, M.; Foreman, K.; Lim, S.; Shibuya, K.; Aboyans, V.; Abraham, J.; Adair, T.; Aggarwal, R.; Ahn, S.Y.; et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: A systematic analysis for the Global Burden of Disease Study 2010. Lancet 2012, 380, 2095–2128. [Google Scholar] [CrossRef]
  5. Luyckx, V.A.; Bertram, J.F.; Brenner, B.M.; Fall, C.; Hoy, W.E.; Ozanne, S.E.; Vikse, B.E. Effect of fetal and child health on kidney development and long-term risk of hypertension and kidney disease. Lancet 2013, 382, 273–283. [Google Scholar] [CrossRef] [Green Version]
  6. Tain, Y.L.; Hsu, C.N. Developmental Origins of Chronic Kidney Disease: Should We Focus on Early Life? Int. J. Mol. Sci. 2017, 18, 381. [Google Scholar] [CrossRef] [Green Version]
  7. Ingelfinger, J.R.; Kalantar-Zadeh, K.; Schaefer, F.; World Kidney Day Steering Committee. World Kidney Day 2016: Averting the legacy of kidney disease-focus in childhood. Pediatr. Nephrol. 2016, 31, 343–348. [Google Scholar] [CrossRef] [Green Version]
  8. Chong, E.; Yosypiv, I.V. Developmental programming of hypertension and kidney disease. Int. J. Nephrol. 2012, 2012, 760580. [Google Scholar] [CrossRef] [Green Version]
  9. Kett, M.M.; Denton, K.M. Renal programming: Cause for concern? Am. J. Physiol. Regul. Integr. Comp. Physiol. 2011, 300, R791–R803. [Google Scholar] [CrossRef]
  10. Bertram, J.F.; Douglas-Denton, R.N.; Diouf, B.; Hughson, M.D.; Hoy, W.E. Human nephron number: Implications for health and disease. Pediatr. Nephrol. 2011, 26, 1529–1533. [Google Scholar] [CrossRef]
  11. Lumbers, E.R.; Kandasamy, Y.; Delforce, S.J.; Boyce, A.C.; Gibson, K.J.; Pringle, K.G. Programming of Renal Development and Chronic Disease in Adult Life. Front. Physiol. 2020, 11, 757. [Google Scholar] [CrossRef] [PubMed]
  12. Hsu, C.N.; Tain, Y.L. The First Thousand Days: Kidney Health and Beyond. Healthcare 2021, 9, 1332. [Google Scholar] [CrossRef] [PubMed]
  13. Wood-Bradley, R.J.; Barrand, S.; Giot, A.; Armitage, J. Understanding the role of maternal diet on kidney development; an opportunity to improve cardiovascular and renal health for future generations. Nutrients 2015, 7, 1881–1905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Lee, Y.Q.; Collins, C.E.; Gordon, A.; Rae, K.M.; Pringle, K.G. the relationship between maternal nutrition during pregnancy and offspring kidney structure and function in humans: A systematic review. Nutrients 2018, 10, 241. [Google Scholar] [CrossRef] [Green Version]
  15. Gai, Z.; Wang, T.; Visentin, M.; Kullak-Ublick, G.A.; Fu, X.; Wang, Z. Lipid Accumulation and Chronic Kidney Disease. Nutrients 2019, 11, 722. [Google Scholar] [CrossRef] [Green Version]
  16. Noels, H.; Lehrke, M.; Vanholder, R.; Jankowski, J. Lipoproteins and fatty acids in chronic kidney disease: Molecular and metabolic alterations. Nat. Rev. Nephrol. 2021, 17, 528–542. [Google Scholar] [CrossRef]
  17. Chen, S.; Chen, J.; Li, S.; Guo, F.; Li, A.; Wu, H.; Chen, J.; Pan, Q.; Liao, S.; Liu, H.; et al. High-Fat Diet-Induced Renal Proximal Tubular Inflammatory Injury: Emerging Risk Factor of Chronic Kidney Disease. Front. Physiol. 2021, 12, 786599. [Google Scholar]
  18. Ribaroff, G.A.; Wastnedge, E.; Drake, A.J.; Sharpe, R.M.; Chambers, T.J.G. Animal models of maternal high fat diet exposure and effects on metabolism in offspring: A meta-regression analysis. Obes. Rev. 2017, 18, 673–686. [Google Scholar] [CrossRef] [Green Version]
  19. Seet, E.L.; Yee, J.K.; Jellyman, J.K.; Han, G.; Ross, M.G.; Desai, M. Maternal high-fat-diet programs rat offspring liver fatty acid metabolism. Lipids 2015, 50, 565–573. [Google Scholar] [CrossRef] [Green Version]
  20. Williams, L.; Seki, Y.; Vuguin, P.M.; Charron, M.J. Animal models of in utero exposure to a high fat diet: A review. Biochim. Biophys. Acta 2014, 1842, 507–519. [Google Scholar] [CrossRef] [Green Version]
  21. Tain, Y.L.; Hsu, C.N. Maternal High-Fat Diet and Offspring Hypertension. Int. J. Mol. Sci. 2022, 23, 8179. [Google Scholar] [CrossRef] [PubMed]
  22. Hsu, C.N.; Tain, Y.L. The Good, the Bad, and the Ugly of Pregnancy Nutrients and Developmental Programming of Adult Disease. Nutrients 2019, 11, 894. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Noyan-Ashraf, M.H.; Wu, L.; Wang, R.; Juurlink, B.H. Dietary approaches to positively influence fetal determinants of adult health. FASEB J. 2006, 20, 371–373. [Google Scholar] [CrossRef]
  24. Tain, Y.L.; Joles, J.A. Reprogramming: A Preventive Strategy in Hypertension Focusing on the Kidney. Int. J. Mol. Sci. 2015, 17, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Kapoor, B.; Kapoor, D.; Gautam, S.; Singh, R.; Bhardwaj, S. Dietary Polyunsaturated Fatty Acids (PUFAs): Uses and Potential Health Benefits. Curr. Nutr. Rep. 2021, 10, 232–242. [Google Scholar] [CrossRef]
  26. Syren, M.L.; Turolo, S.; Marangoni, F.; Milani, G.P.; Edefonti, A.; Montini, G.; Agostoni, C. The polyunsaturated fatty acid balance in kidney health and disease: A review. Clin. Nutr. 2018, 37, 1829–1839. [Google Scholar] [CrossRef]
  27. White, B. Dietary fatty acids. Am. Fam. Physician 2009, 80, 345–350. [Google Scholar]
  28. Marques, L.R.; Diniz, T.A.; Antunes, B.M.; Rossi, F.E.; Caperuto, E.C.; Lira, F.S.; Gonçalves, D.C. Reverse Cholesterol Transport: Molecular Mechanisms and the Non-medical Approach to Enhance HDL Cholesterol. Front. Physiol. 2018, 9, 526. [Google Scholar] [CrossRef] [Green Version]
  29. Stremmel, W.; Pohl, L.; Ring, A.; Herrmann, T. A new concept of cellular uptake and intracellular trafficking of long-chain fatty acids. Lipids 2001, 36, 981–989. [Google Scholar] [CrossRef]
  30. Brunskill, N.J.; Nahorski, S.; Walls, J. Characteristics of albumin binding to opossum kidney cells and identification of potential receptors. Pflugers. Arch. 1997, 433, 497–504. [Google Scholar] [CrossRef]
  31. Muntner, P.; Coresh, J.; Smith, J.C.; Eckfeldt, J.; Klag, M.J. Plasma lipids and risk of developing renal dysfunction: The atherosclerosis risk in communities study. Kidney Int. 2000, 58, 293–301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Kuma, A.; Uchino, B.; Ochiai, Y.; Kawashima, M.; Enta, K.; Tamura, M.; Otsuji, Y.; Kato, A. Impact of low- density lipoprotein cholesterol on decline in estimated glomerular filtration rate in apparently healthy young to middle- aged working men. Clin. Exp. Nephrol. 2018, 22, 15–27. [Google Scholar] [CrossRef] [PubMed]
  33. Schaeffner, E.S.; Kurth, T.; Curhan, G.C.; Glynn, R.J.; Rexrode, K.M.; Baigent, C.; Buring, J.E.; Gaziano, J.M. Cholesterol and the risk of renal dysfunction in apparently healthy men. J. Am. Soc. Nephrol. 2003, 14, 2084–2091. [Google Scholar] [PubMed]
  34. Fox, C.S.; Larson, M.G.; Leip, E.P.; Culleton, B.; Wilson, P.W.; Levy, D. Predictors of new- onset kidney disease in a community- based population. Jama 2004, 291, 844–850. [Google Scholar] [CrossRef] [Green Version]
  35. Soumura, M.; Kume, S.; Isshiki, K.; Takeda, N.; Araki, S.; Tanaka, Y.; Sugimoto, T.; Chin-Kanasaki, M.; Nishio, Y.; Haneda, M. Oleate and eicosapentaenoic acid attenuate palmitate- induced inflammation and apoptosis in renal proximal tubular cell. Biochem. Biophys. Res. Commun. 2010, 402, 265–271. [Google Scholar] [CrossRef]
  36. Hall, J.E.; do Carmo, J.M.; da Silva, A.A.; Wang, Z.; Hall, M.E. Obesity, kidney dysfunction and hypertension: Mechanistic links. Nat. Rev. Nephrol. 2019, 15, 367–385. [Google Scholar] [CrossRef]
  37. Hall, J.E.; Crook, E.D.; Jones, D.W.; Wofford, M.R.; Dubbert, P.M. Mechanisms of obesity-associated cardiovascular and renal disease. Am. J. Med. Sci. 2002, 324, 127–137. [Google Scholar] [CrossRef]
  38. Woods, L.L.; Mizelle, H.L.; Hall, J.E. Control of renal hemodynamics in hyperglycemia: Possible role of tubuloglomerular feedback. Am. J. Physiol. 1987, 252, F65–F73. [Google Scholar] [CrossRef]
  39. Hammoud, S.H.; AlZaim, I.; Al-Dhaheri, Y.; Eid, A.H.; El-Yazbi, A.F. Perirenal Adipose Tissue Inflammation: Novel Insights Linking Metabolic Dysfunction to Renal Diseases. Front. Endocrinol. 2021, 12, 707126. [Google Scholar] [CrossRef]
  40. Woollett, L.A. Fetal lipid metabolism. Front. Biosci. 2001, 6, D536–D545. [Google Scholar]
  41. Herrera, E.; Desoye, G. Maternal and fetal lipid metabolism under normal and gestational diabetic conditions. Horm. Mol. Biol. Clin. Investig. 2016, 26, 109–127. [Google Scholar] [CrossRef] [PubMed]
  42. Herrera, E.; Ortega-Senovilla, H. Lipid metabolism during pregnancy and its implications for fetal growth. Curr. Pharm. Biotechnol. 2014, 15, 24–31. [Google Scholar] [PubMed]
  43. Staiano, L.; De Matteis, M.A. Phosphoinositides in the kidney. J. Lipid Res. 2019, 60, 287–298. [Google Scholar] [PubMed] [Green Version]
  44. Alaynick, W.A. Nuclear receptors, mitochondria and lipid metabolism. Mitochondrion 2008, 8, 329–337. [Google Scholar]
  45. Tain, Y.L.; Hsu, C.N.; Chan, J.Y. PPARs link early life nutritional insults to later programmed hypertension and metabolic syndrome. Int. J. Mol. Sci. 2015, 17, 20. [Google Scholar]
  46. Gao, J.; Gu, Z. The Role of Peroxisome Proliferator-Activated Receptors in Kidney Diseases. Front. Pharmacol. 2022, 13, 832732. [Google Scholar]
  47. Luo, Y.; Wu, M.Y.; Deng, B.Q.; Huang, J.; Hwang, S.H.; Li, M.Y.; Zhou, C.Y.; Zhang, Q.Y.; Yu, H.B.; Zhao, D.K.; et al. Inhibition of soluble epoxide hydrolase attenuates a high-fat diet-mediated renal injury by activating PAX2 and AMPK. Proc. Natl. Acad. Sci. USA 2019, 116, 5154–5159. [Google Scholar]
  48. Polvani, S.; Tarocchi, M.; Galli, A. PPARγ and Oxidative Stress: Con(β) Catenating NRF2 and FOXO. PPAR Res. 2012, 2012, 641087. [Google Scholar]
  49. Todorov, V.T.; Desch, M.; Schmitt-Nilson, N.; Todorova, A.; Kurtz, A. Peroxisome proliferator-activated receptor-γ is involved in the control of renin gene expression. Hypertension 2007, 50, 939–944. [Google Scholar]
  50. Saad, S.; Agapiou, D.J.; Chen, X.M.; Stevens, V.; Pollock, C.A. The role of Sgk-1 in the upregulation of transport proteins by PPAR-γ agonists in human proximal tubule cells. Nephrol. Dial. Transplant. 2009, 24, 1130–1141. [Google Scholar] [CrossRef] [Green Version]
  51. Pluznick, J.L. Microbial short-chain fatty acids and blood pressure regulation. Curr. Hypertens. Rep. 2017, 19, 25. [Google Scholar] [CrossRef] [Green Version]
  52. Ziętek, M.; Celewicz, Z.; Szczuko, M. Short-Chain Fatty Acids, Maternal Microbiota and Metabolism in Pregnancy. Nutrients 2021, 13, 1244. [Google Scholar] [CrossRef] [PubMed]
  53. Buettner, R.; Parhofer, K.G.; Woenckhaus, M.; Wrede, C.E.; Kunz-Schughart, L.A.; Schölmerich, J.; Bollheimer, L.C. Defining high-fat-diet rat models: Metabolic and molecular effects of different fat types. J. Mol. Endocrinol. 2006, 36, 485–501. [Google Scholar] [CrossRef] [Green Version]
  54. Buettner, R.; Schölmerich, J.; Bollheimer, L.C. High-fat diets: Modeling the metabolic disorders of human obesity in rodents. Obesity 2007, 15, 798–808. [Google Scholar] [CrossRef]
  55. Salati, J.A.; Roberts, V.H.J.; Schabel, M.C.; Lo, J.O.; Kroenke, C.D.; Lewandowski, K.S.; Lindner, J.R.; Grove, K.L.; Frias, A.E. Maternal high-fat diet reversal improves placental hemodynamics in a nonhuman primate model of diet-induced obesity. Int. J. Obes. 2019, 43, 906–916. [Google Scholar] [CrossRef]
  56. Sanguinetti, E.; Liistro, T.; Mainardi, M.; Pardini, S.; Salvadori, P.A.; Vannucci, A.; Burchielli, S.; Iozzo, P. Maternal high-fat feeding leads to alterations of brain glucose metabolism in the offspring: Positron emission tomography study in a porcine model. Diabetologia 2016, 59, 813–821. [Google Scholar] [CrossRef] [Green Version]
  57. Lim, K.; Burke, S.L.; Marques, F.Z.; Jackson, K.L.; Gueguen, C.; Sata, Y.; Armitage, J.A.; Head, G.A. Leptin and Melanocortin Signaling Mediates Hypertension in Offspring from Female Rabbits Fed a High-Fat Diet During Gestation and Lactation. Front. Physiol. 2021, 12, 693157. [Google Scholar] [CrossRef]
  58. Chaves, W.F.; Pinheiro, I.L.; da Silva, J.M.; Manhães-de-Castro, R.; da Silva Aragão, R. Repercussions of maternal exposure to high-fat diet on offspring feeding behavior and body composition: A systematic review. J. Dev. Orig. Health Dis. 2021, 12, 220–228. [Google Scholar] [CrossRef]
  59. Ainge, H.; Thompson, C.; Ozanne, S.E.; Rooney, K.B. A systematic review on animal models of maternal high fat feeding and offspring glycaemic control. Int. J. Obes. 2011, 35, 325–335. [Google Scholar] [CrossRef] [Green Version]
  60. Armitage, J.A.; Lakasing, L.; Taylor, P.D.; Balachandran, A.A.; Jensen, R.I.; Dekou, V.; Ashton, N.; Nyengaard, J.R.; Poston, L. Developmental programming of aortic and renal structure in offspring of rats fed fat-rich diets in pregnancy. J. Physiol. 2005, 565, 171–184. [Google Scholar] [CrossRef]
  61. Glastras, S.J.; Chen, H.; McGrath, R.T.; Zaky, A.A.; Gill, A.J.; Pollock, C.A.; Saad, S. Effect of GLP-1 Receptor Activation on Offspring Kidney Health in a Rat Model of Maternal Obesity. Sci. Rep. 2016, 6, 23525. [Google Scholar] [CrossRef] [Green Version]
  62. Armitage, J.A.; Gupta, S.; Wood, C.; Jensen, R.I.; Samuelsson, A.M.; Fuller, W.; Shattock, M.J.; Poston, L.; Taylor, P.D. Maternal dietary supplementation with saturated, but not monounsaturated or polyunsaturated fatty acids, leads to tissue-specific inhibition of offspring Na+,K+-ATPase. J. Physiol. 2008, 586, 5013–5022. [Google Scholar] [CrossRef]
  63. Khan, I.Y.; Taylor, P.D.; Dekou, V.; Seed, P.T.; Lakasing, L.; Graham, D.; Dominiczak, A.F.; Hanson, M.A.; Poston, L. Gender-linked hypertension in offspring of lard-fed pregnant rats. Hypertension 2003, 41, 168–175. [Google Scholar] [CrossRef] [Green Version]
  64. Do Nascimento, L.C.P.; de Souza, E.L.; de Luna Freire, M.O.; de Andrade Braga, V.; de Albuqeurque, T.M.R.; Lagranha, C.J.; de Brito Alves, J.L. Limosilactobacillus fermentum prevent gut-kidney oxidative damage and the rise in blood pressure in male rat offspring exposed to a maternal high-fat diet. J. Dev. Orig. Health Dis. 2022, 19, 719–726. [Google Scholar] [CrossRef]
  65. Kasper, P.; Vohlen, C.; Dinger, K.; Mohr, J.; Hucklenbruch-Rother, E.; Janoschek, R.; Köth, J.; Matthes, J.; Appel, S.; Dötsch, J.; et al. Renal Metabolic Programming Is Linked to the Dynamic Regulation of a Leptin-Klf15 Axis and Akt/AMPKα Signaling in Male Offspring of Obese Dams. Endocrinology 2017, 158, 3399–3415. [Google Scholar] [CrossRef] [Green Version]
  66. Kruse, M.; Fiallo, A.; Tao, J.; Susztak, K.; Amann, K.; Katz, E.B.; Charron, M.J. A High Fat Diet During Pregnancy and Lactation Induces Cardiac and Renal Abnormalities in GLUT4 +/- Male Mice. Kidney Blood Press. Res. 2017, 42, 468–482. [Google Scholar] [CrossRef]
  67. Busnardo de Oliveira, F.; Fortunato Silva, J.; Prado, H.S.D.; Ferreira-Neto, M.L.; Balbi, A.P.C. Maternal high-fat diet consumption during pregnancy and lactation predisposes offspring to renal and metabolic injury later in life: Comparative study of diets with different lipid contents. J. Dev. Orig. Health Dis. 2023, 14, 33–41. [Google Scholar] [CrossRef]
  68. Nguyen, L.T.; Mak, C.H.; Chen, H.; Zaky, A.A.; Wong, M.G.; Pollock, C.A.; Saad, S. SIRT1 Attenuates Kidney Disorders in Male Offspring Due to Maternal High-Fat Diet. Nutrients 2019, 11, 146. [Google Scholar]
  69. Larkin, B.P.; Nguyen, L.T.; Hou, M.; Glastras, S.J.; Chen, H.; Wang, R.; Pollock, C.A.; Saad, S. Novel Role of Gestational Hydralazine in Limiting Maternal and Dietary Obesity-Related Chronic Kidney Disease. Front. Cell Dev. Biol. 2021, 9, 705263. [Google Scholar] [CrossRef]
  70. Hsu, C.N.; Hou, C.Y.; Chan, J.Y.H.; Lee, C.T.; Tain, Y.L. Hypertension Programmed by Perinatal High-Fat Diet: Effect of Maternal Gut Microbiota-Targeted Therapy. Nutrients 2019, 11, 2908. [Google Scholar] [CrossRef] [Green Version]
  71. Tain, Y.L.; Lin, Y.J.; Sheen, J.M.; Lin, I.C.; Yu, H.R.; Huang, L.T.; Hsu, C.N. Resveratrol prevents the combined maternal plus postweaning high-fat-diets-induced hypertension in male offspring. J. Nutr. Biochem. 2017, 48, 120–127. [Google Scholar] [CrossRef]
  72. Tain, Y.L.; Lin, Y.J.; Sheen, J.M.; Yu, H.R.; Tiao, M.M.; Chen, C.C.; Tsai, C.C.; Huang, L.T.; Hsu, C.N. High Fat Diets Sex-Specifically Affect the Renal Transcriptome and Program Obesity, Kidney Injury, and Hypertension in the Offspring. Nutrients 2017, 9, 357. [Google Scholar] [CrossRef] [Green Version]
  73. Hsu, C.N.; Hou, C.Y.; Chang-Chien, G.P.; Lin, S.; Tain, Y.L. Maternal Garlic Oil Supplementation Prevents High-Fat Diet-Induced Hypertension in Adult Rat Offspring: Implications of H2S-Generating Pathway in the Gut and Kidneys. Mol. Nutr. Food Res. 2021, 65, e2001116. [Google Scholar] [CrossRef]
  74. Hsu, C.N.; Hou, C.Y.; Lee, C.T.; Chan, J.Y.H.; Tain, Y.L. The Interplay between Maternal and Post-Weaning High-Fat Diet and Gut Microbiota in the Developmental Programming of Hypertension. Nutrients 2019, 11, 1982. [Google Scholar] [CrossRef] [Green Version]
  75. Tsai, W.L.; Hsu, C.N.; Tain, Y.L. Whether AICAR in Pregnancy or Lactation Prevents Hypertension Programmed by High Saturated Fat Diet: A Pilot Study. Nutrients 2020, 12, 448. [Google Scholar] [CrossRef] [Green Version]
  76. Christians, J.K.; Lennie, K.I.; Wild, L.K.; Garcha, R. Effects of high-fat diets on fetal growth in rodents: A systematic review. Reprod. Biol. Endocrinol. 2019, 17, 39. [Google Scholar] [CrossRef] [Green Version]
  77. Bortolin, R.C.; Vargas, A.R.; Gasparotto, J.; Chaves, P.R.; Schnorr, C.E.; Martinello, K.B.; Silveira, A.K.; Rabelo, T.K.; Gelain, D.P.; Moreira, J.C.F. A new animal diet based on human Western diet is a robust diet-induced obesity model: Comparison to high-fat and cafeteria diets in term of metabolic and gut microbiota disruption. Int. J. Obes. 2018, 42, 525–534. [Google Scholar] [CrossRef]
  78. de la Garza, A.L.; Martínez-Tamez, A.M.; Mellado-Negrete, A.; Arjonilla-Becerra, S.; Peña-Vázquez, G.I.; Marín-Obispo, L.M.; Hernández-Brenes, C. Characterization of the Cafeteria Diet as Simulation of the Human Western Diet and Its Impact on the Lipidomic Profile and Gut Microbiota in Obese Rats. Nutrients 2022, 15, 86. [Google Scholar] [CrossRef]
  79. Sengupta, P. The Laboratory Rat: Relating Its Age with Human’s. Int. J. Prev. Med. 2013, 4, 624–630. [Google Scholar]
  80. Nenov, V.D.; Taal, M.W.; Sakharova, O.V.; Brenner, B.M. Multi-hit nature of chronic renal disease. Curr. Opin. Nephrol. Hypertens. 2000, 9, 85–97. [Google Scholar] [CrossRef]
  81. Winett, L.; Wallack, L.; Richardson, D.; Boone-Heinonen, J.; Messer, L. A Framework to Address Challenges in Communicating the Developmental Origins of Health and Disease. Curr. Environ. Health Rep. 2016, 3, 169–177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Yu, H.R.; Sheen, J.M.; Tiao, M.M.; Tain, Y.L.; Chen, C.C.; Lin, I.C.; Lai, Y.J.; Tsai, C.C.; Lin, Y.J.; Tsai, C.C.; et al. Resveratrol Treatment Ameliorates Leptin Resistance and Adiposity Programed by the Combined Effect of Maternal and Post-Weaning High-Fat Diet. Mol. Nutr. Food Res. 2019, 63, e1801385. [Google Scholar] [CrossRef] [PubMed]
  83. Zhang, X.; Hasan, A.A.; Wu, H.; Gaballa, M.M.S.; Zeng, S.; Liu, L.; Xie, L.; Jung, T.; Grune, T.; Krämer, B.K.; et al. High-fat, sucrose and salt-rich diet during rat spermatogenesis lead to the development of chronic kidney disease in the female offspring of the F2 generation. FASEB J. 2022, 36, e22259. [Google Scholar] [CrossRef] [PubMed]
  84. Hsu, C.N.; Tain, Y.L. Developmental Origins of Kidney Disease: Why Oxidative Stress Matters? Antioxidants 2021, 10, 33. [Google Scholar] [CrossRef] [PubMed]
  85. Thompson, L.P.; Al-Hasan, Y. Impact of oxidative stress in fetal programming. J. Pregnancy 2012, 2012, 582748. [Google Scholar] [CrossRef]
  86. Ojeda, N.B.; Hennington, B.S.; Williamson, D.T.; Hill, M.L.; Betson, N.E.; Sartori-Valinotti, J.C.; Reckelhoff, J.F.; Royals, T.P.; Alexander, B.T. Oxidative stress contributes to sex differences in blood pressure in adult growth-restricted offspring. Hypertension 2012, 60, 114–122. [Google Scholar] [CrossRef] [Green Version]
  87. Cambonie, G.; Comte, B.; Yzydorczyk, C.; Ntimbane, T.; Germain, N.; Lê, N.L.; Pladys, P.; Gauthier, C.; Lahaie, I.; Abran, D.; et al. Antenatal antioxidant prevents adult hypertension, vascular dysfunction, and microvascular rarefaction associated with in utero exposure to a low-protein diet. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2007, 292, R1236–R1245. [Google Scholar] [CrossRef]
  88. Vieira, L.D.; Farias, J.S.; de Queiroz, D.B.; Cabral, E.V.; Lima-Filho, M.M.; Sant’Helena, B.R.M.; Aires, R.S.; Ribeiro, V.S.; SantosRocha, J.; Xavier, F.E.; et al. Oxidative stress induced by prenatal LPS leads to endothelial dysfunction and renal haemodynamic changes through angiotensin II/NADPH oxidase pathway: Prevention by early treatment with α-tocopherol. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 3577–3587. [Google Scholar] [CrossRef]
  89. Tain, Y.L.; Lee, W.C.; Wu, K.L.H.; Leu, S.; Chan, J.Y.H. Resveratrol Prevents the Development of Hypertension Programmed by Maternal Plus Post-Weaning High-Fructose Consumption through Modulation of Oxidative Stress, Nutrient-Sensing Signals, and Gut Microbiota. Mol. Nutr. Food Res. 2018, 30, e1800066. [Google Scholar] [CrossRef]
  90. Chen, H.E.; Lin, Y.J.; Lin, I.C.; Yu, H.R.; Sheen, J.M.; Tsai, C.C.; Huang, L.T.; Tain, Y.L. 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. [Google Scholar] [CrossRef]
  91. Koeners, M.P.; van Faassen, E.E.; Wesseling, S.; Sain-van der Velden, M.; Koomans, H.A.; Braam, B.; Joles, J.A. Maternal supplementation with citrulline increases renal nitric oxide in young spontaneously hypertensive rats and has long-term antihypertensive effects. Hypertension 2007, 50, 1077–1084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Tain, Y.L.; Lee, C.T.; Chan, J.Y.; Hsu, C.N. Maternal melatonin or N-acetylcysteine therapy regulates hydrogen sulfide-generating pathway and renal transcriptome to prevent prenatal N(G)-Nitro-L-arginine-methyl ester (L-NAME)-induced fetal programming of hypertension in adult male offspring. Am. J. Obstet. Gynecol. 2016, 215, 636. [Google Scholar] [CrossRef] [PubMed]
  93. Daenen, K.; Andries, A.; Mekahli, D.; Van Schepdael, A.; Jouret, F.; Bammens, B. Oxidative stress in chronic kidney disease. Pediatr. Nephrol. 2019, 34, 975–991. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Baylis, C. Nitric oxide synthase derangements and hypertension in kidney disease. Curr. Opin. Nephrol. Hypertens. 2012, 21, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Wilcox, C.S. Oxidative stress and nitric oxide deficiency in the kidney: A critical link to hypertension? Am. J. Physiol. Regul. Integr. Comp. Physiol. 2005, 289, R913–R935. [Google Scholar] [CrossRef] [PubMed]
  96. Rosselli, M.; Keller, P.J.; Dubey, R.K. Role of nitric oxide in the biology, physiology and pathophysiology of reproduction. Hum. Reprod. Update 1998, 4, 3–24. [Google Scholar] [CrossRef] [Green Version]
  97. Hsu, C.N.; Tain, Y.L. Regulation of Nitric Oxide Production in the Developmental Programming of Hypertension and Kidney Disease. Int. J. Mol. Sci. 2019, 20, 681. [Google Scholar] [CrossRef] [Green Version]
  98. Tain, Y.Y.; Lee, W.C.; Hsu, C.N.; Lee, W.C.; Huang, L.T.; Lee, C.T.; Lin, C.Y. Asymmetric dimethylarginine is associated with developmental programming of adult kidney disease and hypertension in offspring of streptozotocin-treated mothers. PLoS ONE 2013, 8, e55420. [Google Scholar] [CrossRef]
  99. Hsu, C.N.; Lin, Y.J.; Tain, Y.L. 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. [Google Scholar] [CrossRef] [Green Version]
  100. Tai, I.H.; Sheen, J.M.; Lin, Y.J.; Yu, H.R.; Tiao, M.M.; Chen, C.C.; Huang, L.T.; Tain, Y.L. Maternal N-acetylcysteine therapy regulates hydrogen sulfide-generating pathway and prevents programmed hypertension in male offspring exposed to prenatal dexamethasone and postnatal high-fat diet. Nitric Oxide 2016, 53, 6–12. [Google Scholar] [CrossRef]
  101. Yang, T.; Xu, C. Physiology and Pathophysiology of the Intrarenal Renin-Angiotensin System: An Update. J. Am. Soc. Nephrol. 2017, 28, 1040–1049. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Bessa, A.S.M.; Jesus, É.F.; Nunes, A.D.C.; Pontes, C.N.R.; Lacerda, I.S.; Costa, J.M.; Souza, E.J.; Lino-Júnior, R.S.; Biancardi, M.F.; Dos Santos, F.C.A.; et al. Stimulation of the ACE2/Ang-(1-7)/Mas axis in hypertensive pregnant rats attenuates cardiovascular dysfunction in adult male offspring. Hypertens. Res. 2019, 42, 1883–1893. [Google Scholar] [CrossRef] [PubMed]
  103. Li, C.; Culver, S.A.; Quadri, S.; Ledford, K.L.; Al-Share, Q.Y.; Ghadieh, H.E.; Najjar, S.M.; Siragy, H.M. High-fat diet amplifies renal renin angiotensin system expression, blood pressure elevation, and renal dysfunction caused by Ceacam1 null deletion. Am. J. Physiol. Endocrinol. Metab. 2015, 309, E802–E810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Li, C.; Lin, Y.; Luo, R.; Chen, S.; Wang, F.; Zheng, P.; Levi, M.; Yang, T.; Wang, W. Intrarenal renin-angiotensin system mediates fatty acid-induced ER stress in the kidney. Am. J. Physiol. Ren. Physiol. 2016, 310, F351–F363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Klip, A.; McGraw, T.E.; James, D.E. Thirty sweet years of GLUT4. J. Biol. Chem. 2019, 294, 11369–11381. [Google Scholar] [CrossRef] [Green Version]
  106. Olivares-Reyes, J.A.; Arellano-Plancarte, A.; Castillo-Hernandez, J.R. Angiotensin II and the development of insulin resistance: Implications for diabetes. Mol. Cell Endocrinol. 2009, 302, 128–139. [Google Scholar] [CrossRef]
  107. Stenbit, A.E.; Tsao, T.S.; Li, J.; Burcelin, R.; Geenen, D.L.; Factor, S.M.; Houseknecht, K.; Katz, E.B.; Charron, M.J. GLUT4 heterozygous knockout mice develop muscle insulin resistance and diabetes. Nat. Med. 1997, 3, 1096–1101. [Google Scholar] [CrossRef]
  108. Mennitti, L.V.; Oliveira, J.L.; Morais, C.A.; Estadella, D.; Oyama, L.M.; Oller do Nascimento, C.M.; Pisani, L.P. Type of fatty acids in maternal diets during pregnancy and/or lactation and metabolic consequences of the offspring. J. Nutr. Biochem. 2015, 26, 99–111. [Google Scholar] [CrossRef]
  109. Soppert, J.; Lehrke, M.; Marx, N.; Jankowski, J.; Noels, H. Lipoproteins and lipids in cardiovascular disease: From mechanistic insights to therapeutic targeting. Adv. Drug Deliv. Rev. 2020, 159, 4–33. [Google Scholar] [CrossRef]
  110. Liu, Z.; Huang, X.R.; Chen, H.Y.; Fung, E.; Liu, J.; Lan, H.Y. Deletion of Angiotensin- Converting Enzyme-2 Promotes Hypertensive Nephropathy by Targeting Smad7 for Ubiquitin Degradation. Hypertension 2017, 70, 822–830. [Google Scholar] [CrossRef]
  111. Simões e Silva, A.C.; Silveira, K.D.; Ferreira, A.J.; Teixeira, M.M. ACE2, angiotensin-(1-7) and Mas receptor axis in inflammation and fibrosis. Br. J. Pharmacol. 2013, 169, 477–492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Hsu, C.N.; Tain, Y.L. Targeting the renin–angiotensin–aldosterone system to prevent hypertension and kidney disease of developmental origins. Int. J. Mol. Sci. 2021, 22, 2298. [Google Scholar] [CrossRef] [PubMed]
  113. Gubler, M.C.; Antignac, C. Renin-angiotensin system in kidney development: Renal tubular dysgenesis. Kidney Int. 2010, 77, 400–406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Efeyan, A.; Comb, W.C.; Sabatini, D.M. Nutrient-sensing mechanisms and pathways. Nature 2015, 517, 302–310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Duca, F.A.; Sakar, Y.; Covasa, M. The modulatory role of high fat feeding on gastrointestinal signals in obesity. J. Nutr. Biochem. 2013, 24, 1663–1677. [Google Scholar] [CrossRef]
  116. Mount, P.; Davies, M.; Choy, S.W.; Cook, N.; Power, D. Obesity-Related Chronic Kidney Disease-The Role of Lipid Metabolism. Metabolites 2015, 5, 720–732. [Google Scholar] [CrossRef]
  117. Lomb, D.J.; Laurent, G.; Haigis, M.C. Sirtuins regulate key aspects of lipid metabolism. Biochim. Biophys. Acta 2010, 1804, 1652–1657. [Google Scholar] [CrossRef]
  118. Grygiel-Górniak, B. Peroxisome proliferator-activated receptors and their ligands: Nutritional and clinical implications-a review. Nutr. J. 2014, 13, 17. [Google Scholar] [CrossRef] [Green Version]
  119. Jansson, T.; Powell, T. Role of Placental Nutrient Sensing in Developmental Programming. Clin. Obstet. Gynecol. 2013, 56, 591–601. [Google Scholar] [CrossRef] [Green Version]
  120. Tain, Y.L.; Hsu, C.N. The Impact of Nutrient Intake and Metabolic Wastes during Pregnancy on Offspring Hypertension: Challenges and Future Opportunities. Metabolites 2023, 13, 418. [Google Scholar] [CrossRef]
  121. Tain, Y.L.; Hsu, C.N. AMP-Activated Protein Kinase as a Reprogramming Strategy for Hypertension and Kidney Disease of Developmental Origin. Int. J. Mol. Sci. 2018, 19, 1744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Ruan, X.; Zheng, F.; Guan, Y. PPARs and the kidney in metabolic syndrome. Am. J. Physiol. Ren. Physiol. 2008, 294, F1032–F1047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Azhar, S. Peroxisome proliferator-activated receptors, metabolic syndrome and cardiovascular disease. Future Cardiol. 2010, 6, 657–691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Monsalve, F.A.; Pyarasani, R.D.; Delgado-Lopez, F.; Moore-Carrasco, R. Peroxisome proliferator-activated receptor targets for the treatment of metabolic diseases. Mediat. Inflamm. 2013, 2013, 549627. [Google Scholar] [CrossRef] [Green Version]
  125. Zmora, N.; Suez, J.; Elinav, E. You are what you eat: Diet, health and the gut microbiota. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 35–56. [Google Scholar] [CrossRef] [Green Version]
  126. Al Rubaye, H.; Adamson, C.C.; Jadavji, N.M. The role of maternal diet on offspring gut microbiota development: A review. J. Neurosci. Res. 2021, 99, 284–293. [Google Scholar] [CrossRef]
  127. Wolters, M.; Ahrens, J.; Romaní-Pérez, M.; Watkins, C.; Sanz, Y.; Benítez-Páez, A.; Stanton, C.; Günther, K. Dietary fat, the gut microbiota, and metabolic health—A systematic review conducted within the MyNewGut project. Clin. Nutr. 2019, 38, 2504–2520. [Google Scholar] [CrossRef] [Green Version]
  128. Mosca, A.; Leclerc, M.; Hugot, J.P. Gut Microbiota Diversity and Human Diseases: Should We Reintroduce Key Predators in Our Ecosystem? Front. Microbiol. 2016, 7, 455. [Google Scholar] [CrossRef] [Green Version]
  129. Guimarães, K.S.L.; Braga, V.A.; Noronha, S.I.S.R.; Costa, W.K.A.D.; Makki, K.; Cruz, J.C.; Brandão, L.R.; Chianca Junior, D.A.; Meugnier, E.; Leulier, F.; et al. Lactiplantibacillus plantarum WJL administration during pregnancy and lactation improves lipid profile, insulin sensitivity and gut microbiota diversity in dyslipidemic dams and protects male offspring against cardiovascular dysfunction in later life. Food Funct. 2020, 11, 8939–8950. [Google Scholar] [CrossRef]
  130. Yang, T.; Richards, E.M.; Pepine, C.J.; Raizada, M.K. The gut microbiota and the brain-gut-kidney axis in hypertension and chronic kidney disease. Nat. Rev. Nephrol. 2018, 14, 442–456. [Google Scholar] [CrossRef]
  131. Un-Nisa, A.; Khan, A.; Zakria, M.; Siraj, S.; Ullah, S.; Tipu, M.K.; Ikram, M.; Kim, M.O. Updates on the Role of Probiotics against Different Health Issues: Focus on Lactobacillus. Int. J. Mol. Sci. 2022, 24, 142. [Google Scholar] [CrossRef] [PubMed]
  132. Cani, P.D.; de Vos, W.M. Next-Generation Beneficial Microbes: The Case of Akkermansia muciniphila. Front. Microbiol. 2017, 8, 1765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Zixin, Y.; Lulu, C.; Xiangchang, Z.; Qing, F.; Binjie, Z.; Chunyang, L.; Tai, R.; Dongsheng, O. TMAO as a potential biomarker and therapeutic target for chronic kidney disease: A review. Front. Pharmacol. 2022, 13, 929262. [Google Scholar] [CrossRef] [PubMed]
  134. Li, L.; Ma, L.; Fu, P. Gut microbiota-derived short-chain fatty acids and kidney diseases. Drug Des. Devel. Ther. 2017, 11, 3531–3542. [Google Scholar] [CrossRef] [Green Version]
  135. Liu, J.R.; Miao, H.; Deng, D.Q.; Vaziri, N.D.; Li, P.; Zhao, Y.Y. Gut microbiota-derived tryptophan metabolism mediates renal fibrosis by aryl hydrocarbon receptor signaling activation. Cell Mol. Life Sci. 2021, 78, 909–922. [Google Scholar] [CrossRef]
  136. Tain, Y.L.; Hou, C.Y.; Chang-Chien, G.P.; Lin, S.F.; Hsu, C.N. Perinatal Propionate Supplementation Protects Adult Male Offspring from Maternal Chronic Kidney Disease-Induced Hypertension. Nutrients 2022, 14, 3435. [Google Scholar] [CrossRef]
  137. Zhang, W.; Miikeda, A.; Zuckerman, J.; Jia, X.; Charugundla, S.; Zhou, Z.; Kaczor-Urbanowicz, K.E.; Magyar, C.; Guo, F.; Wang, Z.; et al. Inhibition of microbiota-dependent TMAO production attenuates chronic kidney disease in mice. Sci. Rep. 2021, 11, 518. [Google Scholar] [CrossRef]
  138. Hsu, C.N.; Chang-Chien, G.P.; Lin, S.; Hou, C.Y.; Tain, Y.L. Targeting on Gut Microbial Metabolite Trimethylamine-N-Oxide and Short-Chain Fatty Acid to Prevent Maternal High-Fructose-Diet-Induced Developmental Programming of Hypertension in Adult Male Offspring. Mol. Nutr. Food Res. 2019, 63, e1900073. [Google Scholar] [CrossRef]
  139. Tain, Y.L.; Chang-Chien, G.P.; Lin, S.; Hou, C.Y.; Hsu, C.N. Iodomethylcholine Inhibits Trimethylamine-N-Oxide Production and Averts Maternal Chronic Kidney Disease-Programmed Offspring Hypertension. Int. J. Mol. Sci. 2023, 24, 1284. [Google Scholar] [CrossRef]
  140. Chen, K.; Zheng, X.; Feng, M.; Li, D.; Zhang, H. Gut Microbiota-Dependent Metabolite Trimethylamine N-Oxide Contributes to Cardiac Dysfunction in Western Diet-Induced Obese Mice. Front. Physiol. 2017, 8, 139. [Google Scholar] [CrossRef] [Green Version]
  141. Addi, T.; Dou, L.; Burtey, S. Tryptophan-Derived Uremic Toxins and Thrombosis in Chronic Kidney Disease. Toxins 2018, 10, 412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Hubbard, T.D.; Murray, I.A.; Perdew, G.H. Indole and Tryptophan Metabolism: Endogenous and Dietary Routes to Ah Receptor Activation. Drug Metab. Dispos. 2015, 43, 1522–1535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Zhang, N. The role of endogenous aryl hydrocarbon receptor signaling in cardiovascular physiology. J. Cardiovasc. Dis. Res. 2011, 2, 91–95. [Google Scholar] [CrossRef] [Green Version]
  144. Jaeger, C.; Xu, C.; Sun, M.; Krager, S.; Tischkau, S.A. Aryl hydrocarbon receptor-deficient mice are protected from high fat diet-induced changes in metabolic rhythms. Chronobiol. Int. 2017, 34, 318–336. [Google Scholar] [CrossRef] [PubMed]
  145. Gawlinska, K.; Gawli′nski, D.; Borczyk, M.; Korosty′nski, M.; Przegali′nski, E.; Filip, M. A Maternal High-Fat Diet during Early Development Provokes Molecular Changes Related to Autism Spectrum Disorder in the Rat Offspring Brain. Nutrients 2021, 13, 3212. [Google Scholar] [CrossRef] [PubMed]
  146. Challis, J.R.; Lockwood, C.J.; Myatt, L.; Norman, J.E.; Strauss, J.F.; Petraglia, F. Inflammation and pregnancy. Reprod. Sci. 2009, 16, 206–215. [Google Scholar] [CrossRef] [PubMed]
  147. Lu, X.; Crowley, S.D. Inflammation in Salt-Sensitive Hypertension and Renal Damage. Curr. Hypertens. Rep. 2018, 20, 103. [Google Scholar] [CrossRef]
  148. Zhang, J.; Hua, G.; Zhang, X.; Tong, R.; Du, X.; Li, Z. Regulatory T cells/T-helper cell 17 functional imbalance in uraemic patients on maintenance haemodialysis: A pivotal link between microinflammation and adverse cardiovascular events. Nephrology 2010, 15, 33–41. [Google Scholar] [CrossRef]
  149. Stevens, E.A.; Mezrich, J.D.; Bradfield, C.A. The aryl hydrocarbon receptor: A perspective on potential roles in the immune system. Immunology 2009, 127, 299–311. [Google Scholar] [CrossRef]
  150. Sallée, M.; Dou, L.; Cerini, C.; Poitevin, S.; Brunet, P.; Burtey, S. The aryl hydrocarbon receptor-activating effect of uremic toxins from tryptophan metabolism: A new concept to understand cardiovascular complications of chronic kidney disease. Toxins 2014, 6, 934–949. [Google Scholar] [CrossRef]
  151. Yang, P.; Xiao, Y.; Luo, X.; Zhao, Y.; Zhao, L.; Wang, Y.; Wu, T.; Wei, L.; Chen, Y. Inflammatory stress promotes the development of obesity-related chronic kidney disease via CD36 in mice. J. Lipid Res. 2017, 58, 1417–1427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Hsu, C.N.; Hung, C.H.; Hou, C.Y.; Chang, C.I.; Tain, Y.L. Perinatal Resveratrol Therapy to Dioxin-Exposed Dams Prevents the Programming of Hypertension in Adult Rat Offspring. Antioxidants 2021, 10, 1393. [Google Scholar] [CrossRef] [PubMed]
  153. da Silva, J.F.; Bolsoni, J.A.; da Costa, R.M.; Alves, J.V.; Bressan, A.F.M.; Silva, L.E.V.; Costa, T.J.; Oliveira, A.E.R.; Manzato, C.P.; Aguiar, C.A.; et al. Aryl hydrocarbon receptor (AhR) activation contributes to high-fat diet-induced vascular dysfunction. Br. J. Pharmacol. 2022, 179, 2938–2952. [Google Scholar] [CrossRef]
  154. Lin, Y.J.; Huang, L.T.; Tsai, C.C.; Sheen, J.M.; Tiao, M.M.; Yu, H.R.; Lin, I.C.; Tain, Y.L. Maternal high-fat diet sex-specifically alters placental morphology and transcriptome in rats: Assessment by next-generation sequencing. Placenta 2019, 78, 44–53. [Google Scholar] [CrossRef] [PubMed]
  155. Preston, C.C.; Larsen, T.D.; Eclov, J.A.; Louwagie, E.J.; Gandy, T.C.T.; Faustino, R.S.; Baack, M.L. Maternal High Fat Diet and Diabetes Disrupts Transcriptomic Pathways That Regulate Cardiac Metabolism and Cell Fate in Newborn Rat Hearts. Front. Endocrinol. 2020, 11, 570846. [Google Scholar] [CrossRef]
  156. Peleli, M.; Zampas, P.; Papapetropoulos, A. Hydrogen Sulfide and the Kidney: Physiological Roles, Contribution to Pathophysiology, and Therapeutic Potential. Antioxid. Redox Signal. 2022, 36, 220–243. [Google Scholar] [CrossRef]
  157. Hsu, C.N.; Tain, Y.L. Preventing developmental origins of cardiovascular disease: Hydrogen sulfide as a potential target? Antioxidants 2021, 10, 247. [Google Scholar] [CrossRef]
  158. Paixão, A.D.; Alexander, B.T. How the kidney is impacted by the perinatal maternal environment to develop hypertension. Biol. Reprod. 2013, 89, 144. [Google Scholar] [CrossRef]
  159. Paauw, N.D.; van Rijn, B.B.; Lely, A.T.; Joles, J.A. Pregnancy as a critical window for blood pressure regulation in mother and child: Programming and reprogramming. Acta Physiol. 2017, 219, 241–259. [Google Scholar] [CrossRef]
  160. Li, P.K.T.; Garcia-Garcia, G.; Lui, S.F.; Andreoli, S.; Fung, W.W.-S.; Hradsky, A.; Kumaraswami, L.; Liakopoulos, V.; Rakhimova, Z.; Saadi, G.; et al. Kidney health for everyone everywhere—From prevention to detection and equitable access to care. Pediatr. Nephrol. 2020, 35, 1801–1810. [Google Scholar] [CrossRef]
  161. Tain, Y.L.; Chan, S.H.H.; Chan, J.Y.H. Biochemical basis for pharmacological intervention as a reprogramming strategy against hypertension and kidney disease of developmental origin. Biochem. Pharmacol. 2018, 153, 82–90. [Google Scholar] [CrossRef] [PubMed]
  162. Hoffmann, P.; Taube, C.; Heinroth-Hoffmann, I.; Fahr, A.; Beitz, J.; Förster, W.; Poleshuk, W.S.; Markov, C.M. Antihypertensive action of dietary polyunsaturated fatty acids in spontaneously hypertensive rats. Arch Int. Pharmacodyn. Ther. 1985, 276, 222–235. [Google Scholar] [PubMed]
  163. Shamseldeen, A.M.; Ali Eshra, M.; Ahmed Rashed, L.; Fathy Amer, M.; Elham Fares, A.; Samir Kamar, S. Omega-3 attenuates high fat diet-induced kidney injury of female rats and renal programming of their offsprings. Arch. Physiol. Biochem. 2019, 125, 367–377. [Google Scholar] [CrossRef] [PubMed]
  164. Gray, C.; Vickers, M.H.; Segovia, S.A.; Zhang, X.D.; Reynolds, C.M. A maternal high fat diet programmes endothelial function and cardiovascular status in adult male offspring independent of body weight, which is reversed by maternal conjugated linoleic acid (CLA) supplementation. PLoS ONE 2015, 10, e0115994. [Google Scholar]
  165. Tain, Y.L.; Lee, W.C.; Wu, K.L.H.; Leu, S.; Chan, J.Y.H. Targeting arachidonic acid pathway to prevent programmed hypertension in maternal fructose-fed male adult rat offspring. J. Nutr. Biochem. 2016, 38, 86–92. [Google Scholar] [CrossRef]
  166. Harkins, C.P.; Kong, H.H.; Segre, J.A. Manipulating the Human Microbiome to Manage Disease. JAMA 2020, 323, 303–304. [Google Scholar] [CrossRef]
  167. Tang, W.H.W.; Bäckhed, F.; Landmesser, U.; Hazen, S.L. Intestinal Microbiota in Cardiovascular Health and Disease: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2019, 73, 2089–2105. [Google Scholar] [CrossRef]
  168. Vitetta, L.; Gobe, G. Uremia and chronic kidney disease: The role of the gut microflora and therapies with pro- and prebiotics. Mol. Nutr. Food Res. 2013, 57, 824–832. [Google Scholar] [CrossRef]
  169. Rukavina Mikusic, N.L.; Kouyoumdzian, N.M.; Choi, M.R. Gut microbiota and chronic kidney disease: Evidences and mechanisms that mediate a new communication in the gastrointestinal-renal axis. Pflugers. Arch. 2020, 472, 303–320. [Google Scholar] [CrossRef]
  170. Zeisberg, E.M.; Zeisberg, M.A. A Rationale for Epigenetic Repurposing of Hydralazine in Chronic Heart and Kidney Failure. J. Clin. Epigenet. 2016, 2016, 2. [Google Scholar]
  171. Tampe, B.; Tampe, D.; Zeisberg, E.M.; Müller, G.A.; Bechtel-Walz, W.; Koziolek, M.; Kalluri, R.; Zeisberg, M. Induction of Tet3-dependent Epigenetic Remodeling by Low-dose Hydralazine Attenuates Progression of Chronic Kidney Disease. EBioMed. 2015, 2, 19–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Hsu, C.N.; Hou, C.Y.; Chang-Chien, G.P.; Lin, S.; Tain, Y.L. Dietary Supplementation with Cysteine during Pregnancy Rescues Maternal Chronic Kidney Disease-Induced Hypertension in Male Rat Offspring: The Impact of Hydrogen Sulfide and Microbiota-Derived Tryptophan Metabolites. Antioxidants 2022, 11, 483. [Google Scholar] [CrossRef] [PubMed]
  173. Tain, Y.L.; Huang, L.T.; Lee, C.T.; Chan, J.Y.; Hsu, C.N. Maternal citrulline supplementation prevents prenatal N(G)-nitro-L-arginine-methyl ester (L-NAME)-induced programmed hypertension in rats. Biol. Reprod. 2015, 92, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Hsu, C.N.; Hou, C.Y.; Chang-Chien, G.P.; Lin, S.; Tain, Y.L. Maternal N-Acetylcysteine Therapy Prevents Hypertension in Spontaneously Hypertensive Rat Offspring: Implications of Hydrogen Sulfide-Generating Pathway and Gut Microbiota. Antioxidants 2020, 9, 856. [Google Scholar] [CrossRef]
  175. Tain, Y.L.; Sheen, J.M.; Yu, H.R.; Chen, C.C.; Tiao, M.M.; Hsu, C.N.; Lin, Y.J.; Kuo, K.C.; Huang, L.T. Maternal Melatonin Therapy Rescues Prenatal Dexamethasone and Postnatal High-Fat Diet Induced Programmed Hypertension in Male Rat Offspring. Front. Physiol. 2015, 6, 377. [Google Scholar] [CrossRef] [Green Version]
  176. Lamothe, J.; Khurana, S.; Tharmalingam, S.; Williamson, C.; Byrne, C.J.; Lees, S.J.; Khaper, N.; Kumar, A.; Tai, T.C. Oxidative Stress Mediates the Fetal Programming of Hypertension by Glucocorticoids. Antioxidants 2021, 10, 531. [Google Scholar] [CrossRef]
  177. Wang, Q.; Yue, J.; Zhou, X.; Zheng, M.; Cao, B.; Li, J. Ouabain regulates kidney metabolic profiling in rat offspring of intrauterine growth restriction induced by low-protein diet. Life Sci. 2020, 259, 118281. [Google Scholar] [CrossRef]
  178. Kataoka, S.; Norikura, T.; Sato, S. Maternal green tea polyphenol intake during lactation attenuates kidney injury in high-fat-diet-fed male offspring programmed by maternal protein restriction in rats. J. Nutr. Biochem. 2018, 56, 99–108. [Google Scholar] [CrossRef]
  179. Nguyen, L.T.; Stangenberg, S.; Chen, H.; Al-Odat, I.; Chan, Y.L.; Gosnell, M.E.; Anwer, A.G.; Goldys, E.M.; Pollock, C.A.; Saad, S. L-Carnitine reverses maternal cigarette smoke exposure-induced renal oxidative stress and mitochondrial dysfunction in mouse offspring. Am. J. Physiol. Renal Physiol. 2015, 308, F689–F696. [Google Scholar] [CrossRef] [Green Version]
  180. Hsu, C.N.; Yu, H.R.; Chan, J.Y.H.; Lee, W.C.; Wu, K.L.H.; Hou, C.Y.; Chang-Chien, G.P.; Lin, S.; Tain, Y.L. Maternal Acetate Supplementation Reverses Blood Pressure Increase in Male Offspring Induced by Exposure to Minocycline during Pregnancy and Lactation. Int. J. Mol. Sci. 2022, 23, 7924. [Google Scholar] [CrossRef]
  181. Kidney Disease: Improving Global Outcomes (KDIGO) CKD Work Group. KDIGO 2012 clinical practice guideline for the evaluation and management of chronic kidney disease. Kidney Int. Suppl. 2013, 3, 11–50. [Google Scholar]
Figure 1. The role of a maternal high-fat diet in offspring kidney health and disease. A maternal diet enriched in saturated fat, trans fats, monounsaturated fatty acids (MUFAs), or polyunsaturated fatty acids (PUFAs) can alter lipid uptake, lipid transport, and lipid-sensing signals in the developing fetus. These changes may cause renal programming, leading to an increased risk for kidney disease in adulthood.
Figure 1. The role of a maternal high-fat diet in offspring kidney health and disease. A maternal diet enriched in saturated fat, trans fats, monounsaturated fatty acids (MUFAs), or polyunsaturated fatty acids (PUFAs) can alter lipid uptake, lipid transport, and lipid-sensing signals in the developing fetus. These changes may cause renal programming, leading to an increased risk for kidney disease in adulthood.
Nutrients 15 02698 g001
Figure 2. Schematic diagram of the mechanistic links between a maternal high-fat diet and renal programming. NO = nitric oxide. RAAS = renin-angiotensin-aldosterone system. H2S = hydrogen sulfide.
Figure 2. Schematic diagram of the mechanistic links between a maternal high-fat diet and renal programming. NO = nitric oxide. RAAS = renin-angiotensin-aldosterone system. H2S = hydrogen sulfide.
Nutrients 15 02698 g002
Table 1. Animal models of maternal high-fat-diet-induced renal programming.
Table 1. Animal models of maternal high-fat-diet-induced renal programming.
Fat Fraction and
Component
Species/GenderAge at Measure (Weeks)Programming EffectsRefs.
20% (Lard)SD rat/M & F52Reduced renin and Na+,K+-ATPase activity in kidney[60]
23% (Saturated fats)SD rat/M9Increased markers of oxidative stress, fibrosis, and inflammation[61]
24% (Palm oil)SD rat/M+F26Reduced renal Na+,K+-ATPase activity[62]
31% (Palm oil)Wistar rat/M+F13Increased lipid peroxidation and reduced SOD activity in the kidneys[63]
31% (Lard)Wistar rat/M14Increased renal oxidative stress[64]
34% (mainly linolenic acid and oleic acid)C57BL/6 mice/M3Renal hypertrophy, decreased renal sodium excretion, and increased renal matrix deposition[65]
35.5% (Lard)CD-1 mice and GLUT4 heterozygous mice/M24Elevated BP and increased renal expression of renin and AT1R[66]
40% (Saturated fats)Wistar rat/M13Decreased GFR and increased proteinuria[67]
43% (Saturated fats)C57BL/6 mice/M9Increased renal triglyceride levels, increased renal oxidative stress, inflammatory, and fibrotic markers, as well as increased albuminuria[68]
43% (Saturated fats)C57BL/6 mice/M32Increased creatinine level, albuminuria, glomerulosclerosis, and renal fibrosis[69]
58% (Coconut oil)SD rat/M16Elevated BP, increased renal AT1R expression, and alterations in gut microbiota [70]
58% (Coconut oil)SD rat/M16Elevated BP, decreased urinary NO level, increased renal oxidative stress, and decreased renal Ang-(1–7) level [71]
58% (Coconut oil)SD rat/M+F26Increased kidney injury and altered renal transcriptome[72]
58% (Coconut oil)SD rat/M16Elevated BP, dysregulated H2S-generating pathway in the kidney, and shifts in gut microbiota composition[73]
58% (Coconut oil)SD rat/M16Elevated BP, dysregulated nutrient-sensing signals in the kidney, and alterations in gut microbiota composition[74]
58% (Coconut oil)SD rat/M16Elevated BP and impaired nutrient-sensing pathway in kidneys[75]
Studies tabulated based on fat fractions in the maternal diet and age at evaluation. SD = Sprague Dawley; GLUT4 = glucose transporter 4; M = male; F = female; NO = nitric oxide; SOD = superoxide dismutase; GFR = glomerular filtration rate; AT1R = angiotensin II type 1 receptor; H2S = hydrogen sulfide.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Liu, H.-Y.; Lee, C.-H.; Hsu, C.-N.; Tain, Y.-L. Maternal High-Fat Diet Controls Offspring Kidney Health and Disease. Nutrients 2023, 15, 2698. https://doi.org/10.3390/nu15122698

AMA Style

Liu H-Y, Lee C-H, Hsu C-N, Tain Y-L. Maternal High-Fat Diet Controls Offspring Kidney Health and Disease. Nutrients. 2023; 15(12):2698. https://doi.org/10.3390/nu15122698

Chicago/Turabian Style

Liu, Hsi-Yun, Chen-Hao Lee, Chien-Ning Hsu, and You-Lin Tain. 2023. "Maternal High-Fat Diet Controls Offspring Kidney Health and Disease" Nutrients 15, no. 12: 2698. https://doi.org/10.3390/nu15122698

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

Article Metrics

Back to TopTop