[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (219)

Search Parameters:
Keywords = DOHaD

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 5475 KiB  
Article
Maternal Diet during Pregnancy Alters the Metabolites in Relation to Metabolic and Neurodegenerative Diseases in Young Adult Offspring
by Soo-Min Kim, Songjin Oh, Sang Suk Lee, Sunwha Park, Young-Min Hur, AbuZar Ansari, Gain Lee, Man-Jeong Paik, Young-Ah You and Young Ju Kim
Int. J. Mol. Sci. 2024, 25(20), 11046; https://doi.org/10.3390/ijms252011046 (registering DOI) - 14 Oct 2024
Viewed by 318
Abstract
Maternal nutrition during the critical period of pregnancy increases the susceptibility of offspring to the development of diseases later in life. This study aimed to analyze metabolite profiles to investigate the effect of maternal diet during pregnancy on changes in offspring plasma metabolites [...] Read more.
Maternal nutrition during the critical period of pregnancy increases the susceptibility of offspring to the development of diseases later in life. This study aimed to analyze metabolite profiles to investigate the effect of maternal diet during pregnancy on changes in offspring plasma metabolites and to identify correlations with metabolic parameters. Pregnant Sprague-Dawley rats were exposed to under- and overnutrition compared to controls, and their offspring were fed a standard diet after birth. Plasma metabolism was profiled in offspring at 16 weeks of age using liquid chromatography–mass spectrometry (LC-MS/MS) and gas chromatography–tandem mass spectrometry (GC-MS/MS). We analyzed 80 metabolites to identify distinct metabolites and metabolic and neurodegenerative disease-associated metabolites that were sex-differentially altered in each group compared to controls (p < 0.05, VIP score > 1.0). Specifically, changes in 3-indolepropionic acid, anthranilic acid, linoleic acid, and arachidonic acid, which are involved in tryptophan and linoleic acid metabolism, were observed in male offspring and correlated with plasma leptin levels in male offspring. Our results suggest that fatty acids involved in tryptophan and linoleic acid metabolism, which are altered by the maternal diet during pregnancy, may lead to an increased risk of metabolic and neurodegenerative diseases in the early life of male offspring. Full article
(This article belongs to the Section Molecular Endocrinology and Metabolism)
Show Figures

Figure 1

Figure 1
<p>PLS-DA plots showing that metabolites are differentially categorized between male and female groups. Cross-validation and overfitting of PLS-DA were confirmed using parameter accuracy, correlation coefficient (R2), and cross-validation correlation coefficient (Q2). The VIP scores of PLS-DA in the male and female offspring of the FR, HF, and OB groups are presented in <a href="#ijms-25-11046-t002" class="html-table">Table 2</a>. A Hierarchical clustering heatmap was performed with metabolites (<span class="html-italic">p</span> &lt; 0.05), which is a data visualization technique combining hierarchical clustering and grouping similar data points with a heatmap, a graphical representation of data values (<a href="#app1-ijms-25-11046" class="html-app">Figure S4</a>). Control vs. each group in males (<b>A</b>) and in females (<b>B</b>).</p>
Full article ">Figure 1 Cont.
<p>PLS-DA plots showing that metabolites are differentially categorized between male and female groups. Cross-validation and overfitting of PLS-DA were confirmed using parameter accuracy, correlation coefficient (R2), and cross-validation correlation coefficient (Q2). The VIP scores of PLS-DA in the male and female offspring of the FR, HF, and OB groups are presented in <a href="#ijms-25-11046-t002" class="html-table">Table 2</a>. A Hierarchical clustering heatmap was performed with metabolites (<span class="html-italic">p</span> &lt; 0.05), which is a data visualization technique combining hierarchical clustering and grouping similar data points with a heatmap, a graphical representation of data values (<a href="#app1-ijms-25-11046" class="html-app">Figure S4</a>). Control vs. each group in males (<b>A</b>) and in females (<b>B</b>).</p>
Full article ">Figure 2
<p>Metabolic pathway analysis of offspring groups. Metabolic pathway analysis was performed using the 80 metabolites, comparing the control group and the FR, HF, and OB groups, respectively. The statistical significance of pathway analysis was indicated by a <span class="html-italic">p</span> &lt; 0.05 and an impact score &gt; 0.1.</p>
Full article ">Figure 3
<p>Correlation with metabolites related to metabolic pathway and plasma leptin and GLP-1 levels in the offspring. Hierarchical cluster correlation heatmap analysis and Spearman rank correlation coefficient (R value) were computed to investigate the correlation between the selected metabolites (<span class="html-italic">p</span> &lt; 0.05, VIP score &gt; 1.0, AUC &gt; 0.8) and hormones (leptin and GLP-1) in male and female offspring of the FR, HF, and OB groups. The correlation heatmaps are shown in red (positive correlation) and blue (negative correlation). IPA, 3-Indolepropionic acid; 2HB, 2-Hydroxybutyric acid; 3HB, 3-Hydroxybutyric acid; AABA: Alpha-aminobutyric acid; ALA: Alpha-linolenic acid; QA, Quinolinic acid.</p>
Full article ">Figure 3 Cont.
<p>Correlation with metabolites related to metabolic pathway and plasma leptin and GLP-1 levels in the offspring. Hierarchical cluster correlation heatmap analysis and Spearman rank correlation coefficient (R value) were computed to investigate the correlation between the selected metabolites (<span class="html-italic">p</span> &lt; 0.05, VIP score &gt; 1.0, AUC &gt; 0.8) and hormones (leptin and GLP-1) in male and female offspring of the FR, HF, and OB groups. The correlation heatmaps are shown in red (positive correlation) and blue (negative correlation). IPA, 3-Indolepropionic acid; 2HB, 2-Hydroxybutyric acid; 3HB, 3-Hydroxybutyric acid; AABA: Alpha-aminobutyric acid; ALA: Alpha-linolenic acid; QA, Quinolinic acid.</p>
Full article ">
22 pages, 1596 KiB  
Review
Preterm Birth and Kidney Health: From the Womb to the Rest of Life
by You-Lin Tain and Chien-Ning Hsu
Children 2024, 11(10), 1213; https://doi.org/10.3390/children11101213 - 2 Oct 2024
Viewed by 381
Abstract
Chronic kidney disease (CKD) is a widespread condition often resulting from multiple factors, including maternal influences. These risk factors not only heighten the likelihood of developing CKD but increase the risk of a preterm birth. Adverse events during nephrogenesis can disrupt kidney development, [...] Read more.
Chronic kidney disease (CKD) is a widespread condition often resulting from multiple factors, including maternal influences. These risk factors not only heighten the likelihood of developing CKD but increase the risk of a preterm birth. Adverse events during nephrogenesis can disrupt kidney development, leading to a reduced number of nephrons. As survival rates for preterm infants improve, more individuals are living into adulthood, thereby elevating their risk of CKD later in life. This review aims to explore the connections between preterm birth, kidney development, and the increased risk of CKD, while proposing practical solutions for the future through a multidisciplinary approach. We examine human studies linking preterm birth to negative kidney outcomes, summarize animal models demonstrating kidney programming and reduced nephron numbers, and consolidate knowledge on common mechanisms driving kidney programming. Additionally, we discuss factors in the postnatal care environment that may act as secondary insults contributing to CKD risk, such as acute kidney injury (AKI), the use of nephrotoxic drugs, preterm nutrition, and catch-up growth. Finally, we outline recommendations for action, emphasizing the importance of avoiding modifiable risk factors and implementing early CKD screening for children born preterm. Together, we can ensure that advancements in kidney health keep pace with improvements in preterm care. Full article
(This article belongs to the Special Issue Renal and Cardiovascular Consequences of Prematurity)
Show Figures

Figure 1

Figure 1
<p>Primary and secondary prevention to improve kidney health in preterm infants: outline of key recommended actions.</p>
Full article ">Figure 2
<p>The consequences of preterm birth on kidney health and disease throughout the life course. An outline of the early-life (first hit) and postnatal (second hit) events that contribute to low nephron numbers, kidney programming, and the development of chronic kidney disease (CKD) later in life.</p>
Full article ">
22 pages, 1569 KiB  
Review
Maternal Polyphenols and Offspring Cardiovascular–Kidney–Metabolic Health
by You-Lin Tain and Chien-Ning Hsu
Nutrients 2024, 16(18), 3168; https://doi.org/10.3390/nu16183168 - 19 Sep 2024
Viewed by 757
Abstract
Background: The convergence of cardiovascular, kidney, and metabolic disorders at the pathophysiological level has led to the recognition of cardiovascular–kidney–metabolic (CKM) syndrome, which represents a significant global health challenge. Polyphenols, a group of phytochemicals, have demonstrated potential health-promoting effects. Methods: This review highlights [...] Read more.
Background: The convergence of cardiovascular, kidney, and metabolic disorders at the pathophysiological level has led to the recognition of cardiovascular–kidney–metabolic (CKM) syndrome, which represents a significant global health challenge. Polyphenols, a group of phytochemicals, have demonstrated potential health-promoting effects. Methods: This review highlights the impact of maternal polyphenol supplementation on the CKM health of offspring. Results: Initially, we summarize the interconnections between polyphenols and each aspect of CKM syndrome. We then discuss in vivo studies that have investigated the use of polyphenols during pregnancy and breastfeeding, focusing on their role in preventing CKM syndrome in offspring. Additionally, we explore the common mechanisms underlying the protective effects of maternal polyphenol supplementation. Conclusions: Overall, this review underscores the potential of early-life polyphenol interventions in safeguarding against CKM syndrome in offspring. It emphasizes the importance of continued research to advance our understanding and facilitate the clinical translation of these interventions. Full article
(This article belongs to the Special Issue Polyphenols: Exploring the Potential Health Benefits and Beyond)
Show Figures

Figure 1

Figure 1
<p>Polyphenol classes and main compounds.</p>
Full article ">Figure 2
<p>Metabolic fates of dietary polyphenols in the body involve several stages: phase I and II metabolism in the gut and liver, microbial metabolism, absorption into systemic circulation, interaction with target organs, and eventual elimination through feces and urine.</p>
Full article ">Figure 3
<p>Schema outlining the protective mechanisms of polyphenols behind cardiovascular–kidney–metabolic syndrome with developmental origins.</p>
Full article ">
14 pages, 2717 KiB  
Article
Effect of Purified Resveratrol Butyrate Ester Monomers against Hypertension after Maternal High-Fructose Intake in Adult Offspring
by You-Lin Tain, Chih-Yao Hou, Hong-Tai Tzeng, Shu-Fen Lin, Guo-Ping Chang-Chien, Wei-Chia Lee, Kay L. H. Wu, Hong-Ren Yu, Julie Y. H. Chan and Chien-Ning Hsu
Nutrients 2024, 16(18), 3132; https://doi.org/10.3390/nu16183132 - 17 Sep 2024
Viewed by 639
Abstract
Background: Offspring hypertension arising from adverse maternal conditions can be mitigated through dietary nutritional supplementation, including resveratrol. Previously, we identified derivatives of resveratrol butyrate ester (RBE), specifically 3,4′-di-O-butanoylresveratrol (ED2) and 3-O-butanoylresveratrol (ED4), demonstrating their superior antioxidant capabilities compared to RBE itself. This study [...] Read more.
Background: Offspring hypertension arising from adverse maternal conditions can be mitigated through dietary nutritional supplementation, including resveratrol. Previously, we identified derivatives of resveratrol butyrate ester (RBE), specifically 3,4′-di-O-butanoylresveratrol (ED2) and 3-O-butanoylresveratrol (ED4), demonstrating their superior antioxidant capabilities compared to RBE itself. This study sought to assess the protective impact of maternal supplementation with ED2 or ED4 on offspring hypertension in a rat model subjected to a high-fructose (HF) diet during pregnancy and lactation. Methods: Female Sprague–Dawley rats were distributed into distinct dietary groups throughout pregnancy and lactation: (1) standard chow; (2) HF diet (60%); (3) HF diet supplemented with ED2 (25 mg/L); and (4) HF diet supplemented with ED4 (25 mg/L). Male offspring were euthanized at the age of 12 weeks. Results: The maternal HF diet induced hypertension in the offspring, which was mitigated by perinatal supplementation with either ED2 or ED4. These protective effects were attributed to the antioxidant properties of ED2 and ED4, resulting in an increased availability of nitric oxide (NO). Additionally, supplementation with ED2 was connected to an increased abundance of Bifidobacterium and Clostridium genera, which was accompanied by a decrease in Angelakisella and Christensenella. On the other hand, ED4 supplementation shielded rat offspring from hypertension by elevating concentrations of short-chain fatty acids (SCFAs) and their receptors while reducing trimethylamine-N-oxide (TMAO) levels. Conclusions: These findings highlight the potential of purified RBE monomers, ED2 and ED4, as preventive measures against hypertension resulting from a maternal high-fructose diet. Further research is warranted to explore their clinical applications based on these promising results. Full article
Show Figures

Figure 1

Figure 1
<p>Effects of ED2 and ED4 on systolic blood pressures in offspring from week 3 to 12. Statistical analysis by a one-way ANOVA with Tukey’s post hoc test. * <span class="html-italic">p</span> &lt; 0.05 vs. ND; # <span class="html-italic">p</span> &lt; 0.05 vs. HF.</p>
Full article ">Figure 2
<p>Concentrations of (<b>A</b>) arginine, (<b>B</b>) asymmetric dimethylarginine (ADMA), (<b>C</b>) symmetric dimethylarginine (SDMA), and (<b>D</b>) the ratio of arginine to ADMA (AAR) in the plasma. Statistical analysis by a one-way ANOVA with Tukey’s post hoc test. * <span class="html-italic">p</span> &lt; 0.05 vs. ND; # <span class="html-italic">p</span> &lt; 0.05 vs. HF.</p>
Full article ">Figure 3
<p>The assessment of gut microbial community α-diversity is demonstrated in (<b>A</b>) Faith’s phylogenetic diversity (PD) index and (<b>B</b>) the Shannon index. (<b>C</b>) Principal coordinate analysis (PCoA) plots were used to visualize beta diversity, with each data point representing one sample and each color corresponding to a different group. # <span class="html-italic">p</span> &lt; 0.05 vs. HF.</p>
Full article ">Figure 4
<p>Linear discriminant analysis effect size (LEfSe) was employed to identify taxa that were significantly differentially abundant between groups. Taxa with a linear discriminant analysis (LDA) score greater than 3 were primarily highlighted. Statistical analysis by the Wilcoxon test.</p>
Full article ">Figure 5
<p>Genus-based comparison between the HF and HFED2 group showing relative abundance of (<b>A</b>) <span class="html-italic">Bifidobacterium</span>, (<b>B</b>) <span class="html-italic">Clostridium</span>, (<b>C</b>) <span class="html-italic">Angelakisella</span>, and (<b>D</b>) <span class="html-italic">Christensenella</span>. Genus-based comparison between the HF and HFED4 group showing relative abundance of (<b>E</b>) <span class="html-italic">Angelakisella</span> and (<b>F</b>) <span class="html-italic">Christensenella</span>. The dots represent the outliers. Statistical analysis by using <span class="html-italic">p</span>-values adjusted for multiple comparisons with the false discovery rate (FDR) method. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.005.</p>
Full article ">Figure 6
<p>Plasma concentrations of (<b>A</b>) acetate, (<b>B</b>) propionate, (<b>C</b>) butyrate, and (<b>D</b>) mRNA expression of their receptors in rat kidneys, including G protein-coupled receptor 41 (GPR41), GPR43, GPR109A, and olfactory receptor 78 (Oflr78). Statistical analysis by a one-way ANOVA with Tukey’s post hoc test. * <span class="html-italic">p</span> &lt; 0.05 vs. ND; # <span class="html-italic">p</span> &lt; 0.05 vs. HF.</p>
Full article ">
21 pages, 796 KiB  
Review
Maternal Dietary Strategies for Improving Offspring Cardiovascular–Kidney–Metabolic Health: A Scoping Review
by You-Lin Tain and Chien-Ning Hsu
Int. J. Mol. Sci. 2024, 25(18), 9788; https://doi.org/10.3390/ijms25189788 - 10 Sep 2024
Viewed by 681
Abstract
Dietary regulation has been recognized for its profound impact on human health. The convergence of cardiovascular, kidney, and metabolic disorders at the pathophysiological level has given rise to cardiovascular–kidney–metabolic (CKM) syndrome, which constitutes a significant global health burden. Maternal dietary nutrients play a [...] Read more.
Dietary regulation has been recognized for its profound impact on human health. The convergence of cardiovascular, kidney, and metabolic disorders at the pathophysiological level has given rise to cardiovascular–kidney–metabolic (CKM) syndrome, which constitutes a significant global health burden. Maternal dietary nutrients play a crucial role in fetal development, influencing various programmed processes. This review emphasizes the effects of different types of dietary interventions on each component of CKM syndrome in both preclinical and clinical settings. We also provide an overview of potential maternal dietary strategies, including amino acid supplementation, lipid-associated diets, micronutrients, gut microbiota-targeted diets, and plant polyphenols, aimed at preventing CKM syndrome in offspring. Additionally, we discuss the mechanisms mediated by nutrient-sensing signals that contribute to CKM programming. Altogether, we underscore the interaction between maternal dietary interventions and the risk of CKM syndrome in offspring, emphasizing the need for continued research to facilitate their clinical translation. Full article
Show Figures

Figure 1

Figure 1
<p>A summary of the role of maternal dietary regulation in preventing the developmental programming of cardiovascular–kidney–metabolic (CKM) syndrome in offspring later in life.</p>
Full article ">Figure 2
<p>Flowchart of the literature search and selection.</p>
Full article ">
20 pages, 3613 KiB  
Review
Placenta Extracellular Vesicles: Messengers Connecting Maternal and Fetal Systems
by Cheryl S. Rosenfeld
Biomolecules 2024, 14(8), 995; https://doi.org/10.3390/biom14080995 - 13 Aug 2024
Viewed by 1078
Abstract
The placenta operates during gestation as the primary communication organ between the mother and fetus. It is essential for gas, nutrient exchange, and fetal waste transfer. The placenta also produces a wide range of hormones and other factors that influence maternal physiology, including [...] Read more.
The placenta operates during gestation as the primary communication organ between the mother and fetus. It is essential for gas, nutrient exchange, and fetal waste transfer. The placenta also produces a wide range of hormones and other factors that influence maternal physiology, including survival and activity of the corpus luteum of the ovary, but the means whereby the placenta shapes fetal development remain less clear, although the fetal brain is thought to be dependent upon the placenta for factors that play roles in its early differentiation and growth, giving rise to the term “placenta–brain axis”. Placental hormones transit via the maternal and fetal vasculature, but smaller placental molecules require protection from fetal and maternal metabolism. Such biomolecules include small RNA, mRNA, peptides, lipids, and catecholamines that include serotonin and dopamine. These compounds presumably shuttle to maternal and fetal systems via protective extracellular vesicles (EVs). Placental EVs (pEVs) and their components, in particular miRNA (miRs), are known to play important roles in regulating maternal systems, such as immune, cardiovascular, and reproductive functions. A scant amount is known about how pEVs affect fetal cells and tissues. The composition of pEVs can be influenced by gestational diseases. This review will provide critical insight into the roles of pEVs as the intermediary link between maternal and fetal systems, the impact of maternal pathologies on pEV cargo contents, and how an understanding of biomolecular changes within pEVs in health and disease might be utilized to design early diagnostic and mitigation strategies to prevent gestational diseases and later offspring disorders. Full article
(This article belongs to the Section Biological Factors)
Show Figures

Figure 1

Figure 1
<p>Tissue-specific gene enrichment based on target mRNAs for miRNAs altered in the placenta due to BPA exposure. Tissue-specific gene enrichment analysis based on target mRNAs was determined by TissueEnrich [<a href="#B40-biomolecules-14-00995" class="html-bibr">40</a>]. The mouse ENCODE [<a href="#B41-biomolecules-14-00995" class="html-bibr">41</a>] dataset was used for the enrichment analysis with default settings. Enrichments were considered significant if the <span class="html-italic">p</span>-value was ≤0.01 and fold-change ≥2. The target mRNAs were linked to the thymus, cerebellum, olfactory bulb, brain cortex, E 14.5 brain, and heart. E: embryonic age. This figure was published in [<a href="#B34-biomolecules-14-00995" class="html-bibr">34</a>] and reproduced in the current work with permission from Taylor &amp; Francis and Copyright Clearance Center.</p>
Full article ">Figure 2
<p>Transmission electron microscopy and NanoSight analysis of pTGCs and TSCs. (<b>A</b>) Transmission electron microscopy of EVs derived from pTGCs (top row) and TSC (bottom row). Stained with uranyl acetate. (<b>B</b>) Example of a NanoSight report from EVs isolated from culture media containing pTGCs differentiated from TSCs. This analysis provides the average size (mean and mode) of the particles, concentration of particles per ml, and details on Brownian motion based on video analysis of the EVs. A screen capture from the video shows mouse pEVs as viewed with NanoSight. This figure was published in [<a href="#B43-biomolecules-14-00995" class="html-bibr">43</a>] and reproduced in the current work with permission from Oxford University Press and Copyright Clearance Center.</p>
Full article ">Figure 3
<p>Confocal fluorescent microscopy images of isolated and fluorescently labeled EVs from pTGC and TSC and internalization of TB-derived EV by mouse NPC. (<b>A</b>) Isolated and fluorescently labeled EVs from pTGC. (<b>B</b>) Isolated and fluorescently labeled EVs from TSC. (<b>C</b>–<b>E</b>) NPC with EVs derived from pTGC. (<b>F</b>–<b>H</b>) NPC with EVs derived from TSC. This figure was published in [<a href="#B43-biomolecules-14-00995" class="html-bibr">43</a>] and reproduced in the current work with permission from Oxford University Press and Copyright Clearance Center.</p>
Full article ">Figure 4
<p>Potential model of how pEVs might function as messengers between the placenta and developing fetal brain. Cargo contents of these structures might include catecholamines, miRs, mRNA, proteins, and lipids, and such biomolecules can thus assumingly transit from placenta to the brain via the fetal vasculature in the protective structures.</p>
Full article ">
14 pages, 3416 KiB  
Article
Lactoferrin Supplementation during Pregnancy and Lactation Protects Adult Male Rat Offspring from Hypertension Induced by Maternal Adenine Diet
by You-Lin Tain, Chih-Yao Hou, Wei-Ling Chen, Wei-Ting Liao and Chien-Ning Hsu
Nutrients 2024, 16(16), 2607; https://doi.org/10.3390/nu16162607 - 8 Aug 2024
Cited by 1 | Viewed by 861
Abstract
Lactoferrin, a glycoprotein derived from breastmilk, is recognized for its health benefits in infants and children; however, its protective effects when administered during gestation and lactation against offspring hypertension remain unclear. This study aimed to investigate whether maternal lactoferrin supplementation could prevent hypertension [...] Read more.
Lactoferrin, a glycoprotein derived from breastmilk, is recognized for its health benefits in infants and children; however, its protective effects when administered during gestation and lactation against offspring hypertension remain unclear. This study aimed to investigate whether maternal lactoferrin supplementation could prevent hypertension in offspring born to mothers with chronic kidney disease (CKD), with a focus on nitric oxide (NO), renin–angiotensin system (RAS) regulation, and alterations in gut microbiota and short-chain fatty acids (SCFAs). Prior to pregnancy, female rats were subjected to a 0.5% adenine diet for 3 weeks to induce CKD. During pregnancy and lactation, pregnant rats received one of four diets: normal chow, 0.5% adenine diet, 10% lactoferrin diet, or adenine diet supplemented with lactoferrin. Male offspring were euthanized at 12 weeks of age (n = 8 per group). Supplementation with lactoferrin during gestation and lactation prevented hypertension in adult offspring induced by a maternal adenine diet. The maternal adenine diet caused a decrease in the index of NO availability, which was restored by 67% with maternal LF supplementation. Additionally, LF was related to the regulation of the RAS, as evidenced by a reduced renal expression of renin and the angiotensin II type 1 receptor. Combined maternal adenine and LF diets altered beta diversity, shifted the offspring’s gut microbiota, decreased propionate levels, and reduced the renal expression of SCFA receptors. The beneficial effects of lactoferrin are likely mediated through enhanced NO availability, rebalancing the RAS, and alterations in gut microbiota composition and SCFAs. Our findings suggest that maternal lactoferrin supplementation improves hypertension in offspring in a model of adenine-induced CKD, bringing us closer to potentially translating lactoferrin supplementation clinically for children born to mothers with CKD. Full article
(This article belongs to the Special Issue Breastmilk for Healthy Development)
Show Figures

Figure 1

Figure 1
<p>Effects of maternal adenine diet (CKD) and lactoferrin (LF) on systolic blood pressure in offspring from Week 3 to 12. N = 8/group. * <span class="html-italic">p</span> &lt; 0.05 vs. CN; # <span class="html-italic">p</span> &lt; 0.05 vs. CKD.</p>
Full article ">Figure 2
<p>Effects of maternal adenine diet (CKD) and lactoferrin (LF) on the renin–angiotensin system at Week 12. N = 8/group. * <span class="html-italic">p</span> &lt; 0.05 vs. CN.</p>
Full article ">Figure 3
<p>The evaluation of gut microbial biodiversity in offspring born to dams fed an adenine (CKD) or lactoferrin (LF) diet. (<b>A</b>) Faith’s phylogenic diversity (pd), (<b>B</b>) Shannon index, and (<b>C</b>) principal coordinate analysis (PCoA). Outliers are denoted by dots. Each color corresponds to a different group, with each data point representing one sample.</p>
Full article ">Figure 4
<p>Linear discriminant analysis effect size (LEfSe) with an LDA score &gt; 4 identified significantly differential taxa between groups. The respective group is denoted by the color of the horizontal bar.</p>
Full article ">Figure 5
<p>Bar plots showing the genus-level discrimination between the CKD and CKDLF groups.</p>
Full article ">Figure 6
<p>The comparison of relative abundance of genus <span class="html-italic">Robinsoniella</span> among the four groups. Outliers are denoted by dots. ** <span class="html-italic">p</span> &lt; 0.01. **** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 7
<p>Plasma concentrations of (<b>A</b>) acetate, (<b>B</b>) propionate, and (<b>C</b>) butyrate, and (<b>D</b>) renal mRNA expression of SCFA receptors at Week 12. N = 8/group. * <span class="html-italic">p</span> &lt; 0.05 vs. CN.</p>
Full article ">
13 pages, 294 KiB  
Article
The ORIGINS Project: A Cross-Sectional Analysis of the Nutrition Profile of Pregnant Women in a Longitudinal Birth Cohort
by Poonam K. Pannu, Alexander J. J. Scherini, Desiree T. Silva and Sarah Whalan
Nutrients 2024, 16(15), 2571; https://doi.org/10.3390/nu16152571 - 5 Aug 2024
Viewed by 1129
Abstract
Pregnancy is an opportunistic time for dietary intake to influence future disease susceptibility in offspring later in life. The ORIGINS Project was established to identify the factors that contribute to ‘a healthy start to life’ through a focus supporting childhood health and preventing [...] Read more.
Pregnancy is an opportunistic time for dietary intake to influence future disease susceptibility in offspring later in life. The ORIGINS Project was established to identify the factors that contribute to ‘a healthy start to life’ through a focus supporting childhood health and preventing disease (including non-communicable diseases). We aim to describe the dietary intakes of pregnant women in this cohort and to compare these to the Nutrient Reference Values (NRVs) and Australian Recommended Food Score (ARFS). The usual food and nutrient intakes of women were collected using the Australian Eating Survey (AES), a semi-quantitative food frequency questionnaire (FFQ). A total of 374 women completed the AES FFQ at both 20 weeks and 36 weeks of gestation between December 2016 and January 2023. Macronutrient, micronutrient, and food group intake were explored using descriptive statistics. Overall, it was found that the energy contribution from carbohydrates was low, while that from fat and saturated fat was high; participants were not meeting the recommendations for several key micronutrients (calcium, iron, iodine, and folate); and they had low diet quality scores for all food groups. These findings suggest that despite the ongoing promotion of healthy eating during pregnancy, further exploration into why dietary guidelines during pregnancy are not being adhered to is warranted. Full article
(This article belongs to the Section Nutrition in Women)
14 pages, 2756 KiB  
Article
Chondroitin Sulfate Ameliorates Hypertension in Male Offspring Rat Born to Mothers Fed an Adenine Diet
by You-Lin Tain, Chih-Yao Hou, Guo-Ping Chang-Chien, Shu-Fen Lin and Chien-Ning Hsu
Antioxidants 2024, 13(8), 944; https://doi.org/10.3390/antiox13080944 - 2 Aug 2024
Viewed by 662
Abstract
Pregnant women with chronic kidney disease (CKD) face increased risks of adverse outcomes in their adult offspring. Offspring rats born to dams fed an adenine diet develop hypertension, coinciding with dysregulated hydrogen sulfide (H2S) and nitric oxide (NO) pathways, as well [...] Read more.
Pregnant women with chronic kidney disease (CKD) face increased risks of adverse outcomes in their adult offspring. Offspring rats born to dams fed an adenine diet develop hypertension, coinciding with dysregulated hydrogen sulfide (H2S) and nitric oxide (NO) pathways, as well as alterations in gut microbiota. Chondroitin sulfate (CS) is a multifunctional food known for its diverse bioactivities. As a sulfate prebiotic, CS has shown therapeutic potential in various diseases. Here, we investigated the protective effects of maternal CS supplementation against hypertension in offspring induced by an adenine diet. Mother rats were administered regular chow, 0.5% adenine, 3% CS, or a combination throughout gestation and lactation. Maternal CS supplementation effectively protected offspring from hypertension induced by the adenine diet. These beneficial effects of CS were connected with increased renal mRNA and protein levels of 3-mercaptopyruvate sulfurtransferase, an enzyme involved in H2S production. Furthermore, maternal CS treatment significantly enhanced alpha diversity and altered beta diversity of gut microbiota in adult offspring. Specifically, perinatal CS treatment promoted the abundance of beneficial microbes such as Roseburia hominis and Ruminococcus gauvreauii. In conclusion, perinatal CS treatment mitigates offspring hypertension associated with maternal adenine diet, suggesting that early administration of sulfate prebiotics may hold preventive potential. These findings warrant further translational research to explore their clinical implications. Full article
Show Figures

Figure 1

Figure 1
<p>Effects of maternal adenine diet (AD) and chondroitin sulfate (CS) on systolic blood pressure in offspring from Week 3 to 12. N = 8/group; CN = standard diet; AD = standard diet containing 0.5% adenine; CNCS = standard diet containing 3% chondroitin sulfate; ADCS = standard diet containing 0.5% adenine plus 3% CS; AD × CS = interaction of AD × CS.</p>
Full article ">Figure 2
<p>(<b>A</b>) Renal mRNA expression of H<sub>2</sub>S-generating enzymes; (<b>B</b>) representative Western blot protein bands illustrate immunoreactivity to CBS, CSE, and 3MST. The renal cortical protein abundance of (<b>C</b>) CBS (61 kDa), (<b>D</b>) CSE (45 kDa), and (<b>E</b>) 3MST (52 kDa) was quantified. N = 8/group; CN = standard diet; AD = standard diet containing 0.5% adenine; CNCS = standard diet containing 3% chondroitin sulfate; ADCS = standard diet containing 0.5% adenine plus 3% CS; AD × CS = interaction of AD × CS; NS = not significant.</p>
Full article ">Figure 3
<p>Box plots were generated to illustrate (<b>A</b>) Faith’s phylogenetic diversity (PD) index and (<b>B</b>) Shannon index, depicting alpha diversity. * <span class="html-italic">p</span> &lt; 0.05 by the Wilcoxon test. Outliers are denoted by dots. (<b>C</b>) Principal coordinate analysis (PCoA) plots were used to visualize beta diversity, with each data point representing one sample and each color corresponding to a different group. CN = standard diet; AD = standard diet containing 0.5% adenine; CNCS = standard diet containing 3% chondroitin sulfate; ADCS = standard diet containing 0.5% adenine plus 3% CS.</p>
Full article ">Figure 4
<p>Linear discriminant analysis effect size (LEfSe) was employed to identify taxa that were significantly differentially abundant between groups. Taxa with a linear discriminant analysis (LDA) score greater than 4 were primarily highlighted. The color of the horizontal bar represents the respective group. CN = standard diet; AD = standard diet containing 0.5% adenine; CNCS = standard diet containing 3% chondroitin sulfate; ADCS = standard diet containing 0.5% adenine plus 3% CS.</p>
Full article ">Figure 5
<p>Genus-level taxa that significantly differed (false discovery rate (FDR) &lt; 0.05) in relative abundance of (<b>A</b>) <span class="html-italic">Hungatella,</span> (<b>B</b>) <span class="html-italic">Anaerotruncus,</span> and (<b>C</b>) <span class="html-italic">Bifidobacterium</span>. * <span class="html-italic">p</span> &lt; 0.05. *** <span class="html-italic">p</span> &lt; 0.005. **** <span class="html-italic">p</span> &lt; 0.001. Outliers are denoted by dots. CN = standard diet; AD = standard diet containing 0.5% adenine; CNCS = standard diet containing 3% chondroitin sulfate; ADCS = standard diet containing 0.5% adenine plus 3% CS.</p>
Full article ">Figure 5 Cont.
<p>Genus-level taxa that significantly differed (false discovery rate (FDR) &lt; 0.05) in relative abundance of (<b>A</b>) <span class="html-italic">Hungatella,</span> (<b>B</b>) <span class="html-italic">Anaerotruncus,</span> and (<b>C</b>) <span class="html-italic">Bifidobacterium</span>. * <span class="html-italic">p</span> &lt; 0.05. *** <span class="html-italic">p</span> &lt; 0.005. **** <span class="html-italic">p</span> &lt; 0.001. Outliers are denoted by dots. CN = standard diet; AD = standard diet containing 0.5% adenine; CNCS = standard diet containing 3% chondroitin sulfate; ADCS = standard diet containing 0.5% adenine plus 3% CS.</p>
Full article ">Figure 6
<p>Species-level taxa that significantly differed (false discovery rate (FDR) &lt; 0.05) in relative abundance of (<b>A</b>) <span class="html-italic">Roseburia hominis</span> and (<b>B</b>) <span class="html-italic">Ruminococcus gauvreauii</span>. * <span class="html-italic">p</span> &lt; 0.05. **** <span class="html-italic">p</span> &lt; 0.001. Outliers are denoted by dots. AD = standard diet containing 0.5% adenine; ADCS = standard diet containing 0.5% adenine plus 3% CS.</p>
Full article ">
17 pages, 2996 KiB  
Review
Should Pregnant Women Consume Probiotics to Combat Endocrine-Disrupting Chemical-Induced Health Risks to Their Unborn Offspring?
by Cheryl S. Rosenfeld
Biomedicines 2024, 12(8), 1628; https://doi.org/10.3390/biomedicines12081628 - 23 Jul 2024
Viewed by 969
Abstract
Endocrine-disrupting chemicals (EDCs) have become so pervasive in our environment and daily lives that it is impossible to avoid contact with such compounds, including pregnant women seeking to minimize exposures to themselves and their unborn children. Developmental exposure of humans and rodent models [...] Read more.
Endocrine-disrupting chemicals (EDCs) have become so pervasive in our environment and daily lives that it is impossible to avoid contact with such compounds, including pregnant women seeking to minimize exposures to themselves and their unborn children. Developmental exposure of humans and rodent models to bisphenol A (BPA) and other EDCs is linked to increased anxiogenic behaviors, learning and memory deficits, and decreased socio-sexual behaviors. Prenatal exposure to BPA and other EDCs leads to longstanding and harmful effects on gut microbiota with reductions in beneficial bacteria, i.e., gut dysbiosis, and such microbial changes are linked to host changes in fecal metabolites, including those involved in carbohydrate metabolism and synthesis, and neurobehavioral alterations in adulthood, in particular, social and cognitive deficits. Gut dysbiosis is increasingly being recognized as a key driver of a myriad of diseases, ranging from metabolic, cardiovascular, reproductive, and neurobehavioral disorders via the gut-microbiome–brain axis. Thus, EDCs might induce indirect effects on physical and mental health by acting as microbiome-disrupting chemicals. Findings raise the important question as to whether pregnant women should consume a probiotic supplement to mitigate pernicious effects of EDCs, especially BPA, on themselves and their unborn offspring. Current studies investigating the effects of maternal probiotic supplementation on pregnant women’s health and that of their unborn offspring will be reviewed. Data will inform on the potential application of probiotic supplementation to reverse harmful effects of EDCs, especially BPA, in pregnant women unwittingly exposed to these compounds and striving to give their offspring the best start in life. Full article
(This article belongs to the Special Issue Environmental Exposures and Human Diseases—Molecular Insights)
Show Figures

Figure 1

Figure 1
<p>Hormonal and behavioral changes observed in germ-free (GF) mice that established the existence of a gut-microbiome–brain axis. GF mice must be raised in aseptic conditions to prevent exposure to microorganisms that might otherwise colonize the gut and other organ systems. The absence of gut bacteria results in profound changes in stress-hormone signaling pathways, morphological and gene-expression changes in the brain, altered responses to pathogenic organisms due to changes in microglial cells and increased blood–brain-barrier (BBB) permeability, and altered behavioral responses. This figure is from [<a href="#B94-biomedicines-12-01628" class="html-bibr">94</a>] and reproduced from permissions from Oxford University Press and Copyright Clearance Center, Inc.</p>
Full article ">Figure 2
<p>Select mechanisms by which the gut microbiome may influence the brain and other organs, reproduced from [<a href="#B19-biomedicines-12-01628" class="html-bibr">19</a>]. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution, or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice.</p>
Full article ">Figure 3
<p>A mixOmics analysis approach to integrate, in this case, developmental exposure to genistein (GEN) vs. AIN (phytoestrogen-free diet) on gut microbiota changes with fecal metabolite and neurobehavioral changes in vocalizations and social behaviors. This integrative approach is one of the few that permits correlations between multiple omics approaches and phenotypic changes. Positive correlations are shown by the red lines linking specific categories together, whereas inverse or negative correlations between two categories are represented in blue. The correlation strength was set to 0.9, which is quite stringent [<a href="#B17-biomedicines-12-01628" class="html-bibr">17</a>]. This figure was reproduced with permissions from Copyright Clearance Center, Inc.</p>
Full article ">Figure 4
<p>The triad relationship between developmental exposure to EDCs, such as BPA, gut-microbiome changes, and host genetic/epigenetic/phenotypic status. If individuals cannot eliminate exposure to EDCs and it is unrealistic to reverse direct effects of EDCs on our genetic, epigenetic, and disease status, then EDCs acting as microbiome-disrupting chemicals might provide an avenue to intervene and abate the harmful effects of EDCs.</p>
Full article ">Figure 5
<p>Intervention mechanisms to modify gut microbiota and/or their products. In so doing, such approaches might be used to prevent/treat human diseases, including those originating due to early exposure to EDCs, such as BPA. This figure is from [<a href="#B100-biomedicines-12-01628" class="html-bibr">100</a>]. This publication is an open-access article distributed under the terms of the Creative Commons (CC BY) license, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.</p>
Full article ">
17 pages, 1070 KiB  
Review
A Review of Fetal Development in Pregnancies with Maternal Type 2 Diabetes Mellitus (T2DM)-Associated Hypothalamic-Pituitary-Adrenal (HPA) Axis Dysregulation: Possible Links to Pregestational Prediabetes
by Mathuli Ngema, Nombuso D. Xulu, Phikelelani S. Ngubane and Andile Khathi
Biomedicines 2024, 12(6), 1372; https://doi.org/10.3390/biomedicines12061372 - 20 Jun 2024
Viewed by 953
Abstract
Research has identified fetal risk factors for adult diseases, forming the basis for the Developmental Origins of Health and Disease (DOHaD) hypothesis. DOHaD suggests that maternal insults during pregnancy cause structural and functional changes in fetal organs, increasing the risk of chronic diseases [...] Read more.
Research has identified fetal risk factors for adult diseases, forming the basis for the Developmental Origins of Health and Disease (DOHaD) hypothesis. DOHaD suggests that maternal insults during pregnancy cause structural and functional changes in fetal organs, increasing the risk of chronic diseases like type 2 diabetes mellitus (T2DM) in adulthood. It is proposed that altered maternal physiology, such as increased glucocorticoid (GC) levels associated with a dysregulated hypothalamic-pituitary-adrenal (HPA) axis in maternal stress and T2DM during pregnancy, exposes the fetus to excess GC. Prenatal glucocorticoid exposure reduces fetal growth and programs the fetal HPA axis, permanently altering its activity into adulthood. This programmed HPA axis is linked to increased risks of hypertension, cardiovascular diseases, and mental disorders in adulthood. With the global rise in T2DM, particularly among young adults of reproductive age, it is crucial to prevent its onset. T2DM is often preceded by a prediabetic state, a condition that does not show any symptoms, causing many to unknowingly progress to T2DM. Studying prediabetes is essential, as it is a reversible stage that may help prevent T2DM-related pregnancy complications. The existing literature focuses on HPA axis dysregulation in T2DM pregnancies and its link to fetal programming. However, the effects of prediabetes on HPA axis function, specifically glucocorticoid in pregnancy and fetal outcomes, are not well understood. This review consolidates research on T2DM during pregnancy, its impact on fetal programming via the HPA axis, and possible links with pregestational prediabetes. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>A schematic presentation of maternal HPA axis and GC signalling between mother, placenta, and fetus. Glucocorticoids tightly control HPA axis activity through glucocorticoid receptors (GR) and mineralocorticoid receptors (MR) in the pituitary and hypothalamus to inhibit CRH release, ACTH, and its own secretion [<a href="#B83-biomedicines-12-01372" class="html-bibr">83</a>,<a href="#B88-biomedicines-12-01372" class="html-bibr">88</a>]. In pregnancy, the placenta secretes large quantities of CRH into the maternal bloodstream as the pregnancy progresses, which promotes the production of GC [<a href="#B16-biomedicines-12-01372" class="html-bibr">16</a>,<a href="#B77-biomedicines-12-01372" class="html-bibr">77</a>]. Increased placental CRH secretion and GC also increase GR, promoting negative feedback and, therefore, maintaining the HPA axis activity in pregnancy. Nevertheless, the fetus is shielded from excess maternal GC exposure by the increased activity of 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2). The minimal transfer of GC from the placenta to the fetal compartment plays a vital role in the development of fetal organs, particularly the brain’s HPA axis and the maturation of the lungs.</p>
Full article ">Figure 2
<p>The schematic diagram presents the summary of maternal T2DM pregnancy complications, leading to fetal consequences in utero that persist until adulthood.</p>
Full article ">
19 pages, 2681 KiB  
Article
Daily Eicosapentaenoic Acid Infusion in IUGR Fetal Lambs Reduced Systemic Inflammation, Increased Muscle ADRβ2 Content, and Improved Myoblast Function and Muscle Growth
by Haley N. Beer, Taylor A. Lacey, Rachel L. Gibbs, Micah S. Most, Zena M. Hicks, Pablo C. Grijalva, Eileen S. Marks-Nelson, Ty B. Schmidt, Jessica L. Petersen and Dustin T. Yates
Metabolites 2024, 14(6), 340; https://doi.org/10.3390/metabo14060340 - 18 Jun 2024
Viewed by 869
Abstract
Intrauterine growth-restricted (IUGR) fetuses exhibit systemic inflammation that contributes to programmed deficits in myoblast function and muscle growth. Thus, we sought to determine if targeting fetal inflammation improves muscle growth outcomes. Heat stress-induced IUGR fetal lambs were infused with eicosapentaenoic acid (IUGR+EPA; n [...] Read more.
Intrauterine growth-restricted (IUGR) fetuses exhibit systemic inflammation that contributes to programmed deficits in myoblast function and muscle growth. Thus, we sought to determine if targeting fetal inflammation improves muscle growth outcomes. Heat stress-induced IUGR fetal lambs were infused with eicosapentaenoic acid (IUGR+EPA; n = 9) or saline (IUGR; n = 8) for 5 days during late gestation and compared to saline-infused controls (n = 11). Circulating eicosapentaenoic acid was 42% less (p < 0.05) for IUGR fetuses but was recovered in IUGR+EPA fetuses. The infusion did not improve placental function or fetal O2 but resolved the 67% greater (p < 0.05) circulating TNFα observed in IUGR fetuses. This improved myoblast function and muscle growth, as the 23% reduction (p < 0.05) in the ex vivo differentiation of IUGR myoblasts was resolved in IUGR+EPA myoblasts. Semitendinosus, longissimus dorsi, and flexor digitorum superficialis muscles were 24–39% lighter (p < 0.05) for IUGR but not for IUGR+EPA fetuses. Elevated (p < 0.05) IL6R and reduced (p < 0.05) β2 adrenoceptor content in IUGR muscle indicated enhanced inflammatory sensitivity and diminished β2 adrenergic sensitivity. Although IL6R remained elevated, β2 adrenoceptor deficits were resolved in IUGR+EPA muscle, demonstrating a unique underlying mechanism for muscle dysregulation. These findings show that fetal inflammation contributes to IUGR muscle growth deficits and thus may be an effective target for intervention. Full article
(This article belongs to the Special Issue Unlocking the Mysteries of Muscle Metabolism in the Animal Sciences)
Show Figures

Figure 1

Figure 1
<p>Lipid accumulation and fibrotic area in placentomes from IUGR fetal lambs administered daily with eicosapentaenoic acid. Representative images for Trichrome (top row, scale bar = 400 μm) and Oil Red O (bottom row, scale bar = 50 μm) staining are shown in frame (<b>A</b>). Staining was performed in control (<span class="html-italic">n</span> = 11), IUGR (<span class="html-italic">n</span> = 8), and IUGR+EPA fetuses (<span class="html-italic">n</span> = 9). Data are presented for relative collagen area (<b>B</b>), average lipid droplet size (<b>C</b>), and lipid droplet density (<b>D</b>). Effects of the experimental group were evaluated and are noted where significant (<span class="html-italic">p</span> &lt; 0.05). <sup>a,b</sup> Means with different superscripts differ (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>Placental insufficiency indicators in IUGR fetal lambs administered daily with eicosapentaenoic acid. Daily whole blood samples were collected from control (<span class="html-italic">n</span> = 11), IUGR (<span class="html-italic">n</span> = 8), and IUGR+EPA fetuses (<span class="html-italic">n</span> = 9) simultaneously with maternal blood samples. On the top row, data are presented for maternal glucose (<b>A</b>), fetal glucose (<b>B</b>), and maternofetal glucose gradients (<b>C</b>). On the bottom row, data are presented for maternal pO<sub>2</sub> (<b>D</b>), fetal pO<sub>2</sub> (<b>E</b>), and maternofetal pO<sub>2</sub> gradient (<b>F</b>), Effects of the experimental group (GRP), day of gestation, and group x day interaction (G*D) were evaluated and are noted where significant (<span class="html-italic">p</span> &lt; 0.05). <sup>a–c</sup> Means with different superscripts differ (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>Circulating leukocytes in IUGR fetal lambs administered daily with eicosapentaenoic acid. Complete blood counts were performed on daily whole blood samples collected from control (<span class="html-italic">n</span> = 11), IUGR (<span class="html-italic">n</span> = 8), and IUGR+EPA fetuses (<span class="html-italic">n</span> = 9). Data are presented for circulating concentrations of total white blood cells (<b>A</b>), lymphocytes (<b>B</b>), monocytes (<b>C</b>), and granulocytes (<b>D</b>), as well as granulocyte-to-lymphocyte (<b>E</b>) and lymphocyte-to-monocyte ratios (<b>F</b>). Effects of the experimental group (GRP), day of gestation, and group x day interaction (G*D) were evaluated and are noted where significant (<span class="html-italic">p</span> &lt; 0.05). <sup>a,b</sup> Means with different superscripts differ (<span class="html-italic">p</span> &lt; 0.05). <sup>x,y</sup> Means with different superscripts tend to differ (<span class="html-italic">p</span> &lt; 0.10).</p>
Full article ">Figure 4
<p>Systemic inflammation in IUGR fetal lambs administered daily with eicosapentaenoic acid. Plasma was isolated from daily blood samples collected from control (<span class="html-italic">n</span> = 11), IUGR (<span class="html-italic">n</span> = 8), and IUGR+EPA fetuses (<span class="html-italic">n</span> = 9). Data are presented for fetal plasma eicosapentaenoic acid (<b>A</b>) and TNFα (<b>B</b>) concentrations. Effects of the experimental group (GRP), day of gestation, and group × day interaction were evaluated and are noted where significant (<span class="html-italic">p</span> &lt; 0.05). <sup>a,b</sup> Means with different superscripts differ (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>Skeletal muscle hormone receptor content for IUGR fetal lambs administered daily with eicosapentaenoic acid. Total protein was isolated from <span class="html-italic">semitendinosus</span> muscle samples collected from control (<span class="html-italic">n</span> = 11), IUGR (<span class="html-italic">n</span> = 8), and IUGR+EPA fetuses (<span class="html-italic">n</span> = 9). Data are presented for protein immunoblot analysis of muscle β2 adrenoceptor (<b>A</b>) and IL-6 receptor (<b>B</b>) content. Effects of the experimental group (GRP) were evaluated and are noted where significant (<span class="html-italic">p</span> &lt; 0.05). <sup>a,b</sup> Means with different superscripts differ (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 6
<p>Myoblast profiles in skeletal muscle from IUGR fetal lambs administered daily with eicosapentaenoic acid. Immunohistochemistry was performed on fixed <span class="html-italic">semitendinosus</span> muscle cross-sectional samples collected from control (<span class="html-italic">n</span> = 11), IUGR (<span class="html-italic">n</span> = 8), and IUGR+EPA fetuses (<span class="html-italic">n</span> = 9). Data are presented for total myoblasts (<b>A</b>), proliferating myoblasts (<b>B</b>), differentiated myoblasts (<b>C</b>), and average cross-sectional muscle fiber area (<b>D</b>). Effects of the experimental group (GRP) were evaluated and are noted where significant (<span class="html-italic">p</span> &lt; 0.05). <sup>a–c</sup> Means with different superscripts differ (<span class="html-italic">p</span> &lt; 0.05). Representative staining images are included in the <a href="#app1-metabolites-14-00340" class="html-app">Supplemental Materials</a>.</p>
Full article ">Figure 7
<p>Ex vivo myoblast function for IUGR fetal lambs administered daily with eicosapentaenoic acid. Primary myoblasts were isolated from the hindlimb muscles of control (<span class="html-italic">n</span> = 11), IUGR (<span class="html-italic">n</span> = 8), and IUGR+EPA fetuses (<span class="html-italic">n</span> = 9) and studied in culture. Data are presented for proliferation rates (<b>A</b>) during a 2 h EdU pulse and for differentiation rates (<b>B</b>) following a 4-day induction of differentiation. Effects of the experimental group (GRP), incubation media, and group × media interaction were evaluated and are noted where significant (<span class="html-italic">p</span> &lt; 0.05). <sup>a,b</sup> Means with different superscripts differ (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">
20 pages, 1147 KiB  
Review
Amino Acids during Pregnancy and Offspring Cardiovascular–Kidney–Metabolic Health
by You-Lin Tain and Chien-Ning Hsu
Nutrients 2024, 16(9), 1263; https://doi.org/10.3390/nu16091263 - 24 Apr 2024
Viewed by 3905
Abstract
Amino acids are essential for normal pregnancy and fetal development. Disruptions in maternal amino acid metabolism have been associated with various adult diseases later in life, a phenomenon referred to as the developmental origins of health and disease (DOHaD). In this review, we [...] Read more.
Amino acids are essential for normal pregnancy and fetal development. Disruptions in maternal amino acid metabolism have been associated with various adult diseases later in life, a phenomenon referred to as the developmental origins of health and disease (DOHaD). In this review, we examine the recent evidence highlighting the significant impact of amino acids on fetal programming, their influence on the modulation of gut microbiota, and their repercussions on offspring outcomes, particularly in the context of cardiovascular–kidney–metabolic (CKM) syndrome. Furthermore, we delve into experimental studies that have unveiled the protective effects of therapies targeting amino acids. These interventions have demonstrated the potential to reprogram traits associated with CKM in offspring. The discussion encompasses the challenges of translating the findings from animal studies to clinical applications, emphasizing the complexity of this process. Additionally, we propose potential solutions to overcome these challenges. Ultimately, as we move forward, future research endeavors should aim to pinpoint the most effective amino-acid-targeted therapies, determining the optimal dosage and mode of administration. This exploration is essential for maximizing the reprogramming effects, ultimately contributing to the enhancement of cardiovascular–kidney–metabolic health in offspring. Full article
(This article belongs to the Section Nutrition and Public Health)
Show Figures

Figure 1

Figure 1
<p>Flow diagram of the literature search and selection.</p>
Full article ">Figure 2
<p>A cartoon showing placental amino acid transporters that determine the amino acid pool between the mother, placenta, and fetus. Flow diagram of the literature search and selection.</p>
Full article ">Figure 3
<p>An overview of the role of maternal amino acid metabolism in the developmental programming of offspring cardiovascular–kidney–metabolic (CKM) syndrome.</p>
Full article ">
22 pages, 1363 KiB  
Review
The Impact of the Aryl Hydrocarbon Receptor on Antenatal Chemical Exposure-Induced Cardiovascular–Kidney–Metabolic Programming
by You-Lin Tain and Chien-Ning Hsu
Int. J. Mol. Sci. 2024, 25(9), 4599; https://doi.org/10.3390/ijms25094599 - 23 Apr 2024
Cited by 1 | Viewed by 1470
Abstract
Early life exposure lays the groundwork for the risk of developing cardiovascular–kidney–metabolic (CKM) syndrome in adulthood. Various environmental chemicals to which pregnant mothers are commonly exposed can disrupt fetal programming, leading to a wide range of CKM phenotypes. The aryl hydrocarbon receptor (AHR) [...] Read more.
Early life exposure lays the groundwork for the risk of developing cardiovascular–kidney–metabolic (CKM) syndrome in adulthood. Various environmental chemicals to which pregnant mothers are commonly exposed can disrupt fetal programming, leading to a wide range of CKM phenotypes. The aryl hydrocarbon receptor (AHR) has a key role as a ligand-activated transcription factor in sensing these environmental chemicals. Activating AHR through exposure to environmental chemicals has been documented for its adverse impacts on cardiovascular diseases, hypertension, diabetes, obesity, kidney disease, and non-alcoholic fatty liver disease, as evidenced by both epidemiological and animal studies. In this review, we compile current human evidence and findings from animal models that support the connection between antenatal chemical exposures and CKM programming, focusing particularly on AHR signaling. Additionally, we explore potential AHR modulators aimed at preventing CKM syndrome. As the pioneering review to present evidence advocating for the avoidance of toxic chemical exposure during pregnancy and deepening our understanding of AHR signaling, this has the potential to mitigate the global burden of CKM syndrome in the future. Full article
Show Figures

Figure 1

Figure 1
<p>Schema outlining the structure of the aryl hydrocarbon receptor.</p>
Full article ">Figure 2
<p>Classical and non-classical AHR signaling pathways.</p>
Full article ">Figure 3
<p>The AHR connects antenatal chemical exposure to CKM programming and reprogramming.</p>
Full article ">
15 pages, 2096 KiB  
Article
Effect of Probiotic Lacticaseibacillus rhamnosus LB1.5 on Anxiety-like Behavior, Neuroprotection and Neuroinflammation Markers of Male Mice Fed a High-Fat Diet
by Natália Perin Schmidt, Patrícia Molz, Brenda Santos Fraga, Nicole Hiller Bondarczuk, Priscila Dutra Silveira, Milena Henrique Ferri, Thais Busatto Crestani, Gabriela Merker Breyer, Giuliano Rizzoto Guimarães, Amanda de Souza da Motta, Renata Padilha Guedes and Márcia Giovenardi
Nutrients 2024, 16(6), 879; https://doi.org/10.3390/nu16060879 - 18 Mar 2024
Cited by 1 | Viewed by 1360
Abstract
Probiotic supplementation has been identified as a potential target to reduce inflammatory mediators associated with obesity. Therefore, this study assessed the effect of probiotic Lacticaseibacillus rhamnosus LB1.5 on anxiety-like behavior, gene expression in the prefrontal cortex, and neuroinflammation in the cerebral cortex and [...] Read more.
Probiotic supplementation has been identified as a potential target to reduce inflammatory mediators associated with obesity. Therefore, this study assessed the effect of probiotic Lacticaseibacillus rhamnosus LB1.5 on anxiety-like behavior, gene expression in the prefrontal cortex, and neuroinflammation in the cerebral cortex and hippocampus of male mice fed a high-fat diet. Mice aged 21 days were divided into four groups: control (CONT), control plus probiotic (CONT + PROB), high-fat diet (HFD), and high-fat diet plus probiotic (HFD + PROB), and fed for 13 weeks. The probiotic Lact. rhamnosus 1.5 (3.1 × 108 CFU/mL, derived from raw buffalo milk) was administered by gavage three times a week. Probiotic supplementation provided an anxiolytic effect in CONT and HFD. The IL-6 showed lower levels after probiotic supplementation in the HFD. Regarding immunoreactivity for GFAP in the cerebral cortex, we demonstrated that animals HFD-fed had a reduction in cells number compared to CONT. In the hippocampus, we found an interaction between diet and supplementation, as well as an effect of probiotic supplementation. A higher number of Th positive cells was observed in the cerebral cortex in mice fed HFD. Lact. rhamnosus LB1.5 supplementation decreased serum IL-6 levels in HFD-fed mice and promoted a reduction in anxiety-like behavior. Full article
(This article belongs to the Section Prebiotics and Probiotics)
Show Figures

Figure 1

Figure 1
<p>Analysis of serum levels of cytokines of the adult males fed with standard (CONT, <span class="html-italic">n</span> = 6–8), standard plus probiotic (CONT + PROB, <span class="html-italic">n</span> = 6–7), high-fat (HFD, <span class="html-italic">n</span> = 5–7) and high-fat plus probiotic (HFD + PROB, <span class="html-italic">n</span> = 4–7) diets for 13 weeks by two-way ANOVA followed by Bonferroni’s multiple comparison test. Results, presented as mean ± SEM, indicated significant differences (* <span class="html-italic">p</span> &lt; 0.05) compared to the high-fat diet group (HFD). Abbreviations: interleukin 6 (IL-6), interleukin 1α (IL-1α), interleukin 1β (IL-1β) and interleukin 10 (IL-10).</p>
Full article ">Figure 2
<p>Number of IBA1 positive cells in the cerebral cortex and hippocampus (CA1 area and dentate gyrus) of the adult males fed with standard (CONT), standard plus probiotic (CONT + PROB), high-fat (HFD) and high-fat plus probiotic (HFD + PROB) diets for 13 weeks by two-way ANOVA followed by Bonferroni’s multiple comparison test. Results were presented as mean ± SEM, with  5 animals per/group.</p>
Full article ">Figure 3
<p>Number of GFAP positive cells in the cerebral cortex and hippocampus (CA1 area and dentate gyrus) of the adult males fed with standard (CONT), standard plus probiotic (CONT + PROB), high-fat (HFD) and high-fat plus probiotic (HFD + PROB) diets for 13 weeks by two-way ANOVA followed by Bonferroni’s multiple comparison test. (<b>A</b>) Representative images of GFAP positive cells in the cerebral cortex immunofluorescent under confocal microscopy (bar = 200 μm). (<b>B</b>) Representative images of GFAP positive cells in the cerebral hippocampus immunofluorescent under confocal microscopy (bar = 100 μm). Results, presented as mean ± SEM, indicated significant differences (** <span class="html-italic">p</span> &lt; 0.01) compared to the high-fat diet group (HFD). Scale bar = 100 µm and 200 µm, cerebral cortex and hippocampus, respectively. <span class="html-italic">n</span> = 5/group.</p>
Full article ">Figure 4
<p>Number of Th positive cells in the cerebral cortex of the adult males fed with standard (CONT), standard plus probiotic (CONT + PROB), high-fat (HFD) and high-fat plus probiotic (HFD + PROB) diets for 13 weeks by Bonferroni’s multiple comparison test. Results were presented as mean ± SEM. Scale bar = 100 µm. <span class="html-italic">n</span> = 5/group.</p>
Full article ">Figure 5
<p>Relative gene expression of Sirt1, Nrf2 and BDNF in the prefrontal cortex of the adult males fed with standard (CONT, <span class="html-italic">n</span> = 6–8), standard plus probiotic (CONT + PROB, <span class="html-italic">n</span> = 6–7), high-fat (HFD, <span class="html-italic">n</span> = 5–7) and high-fat plus probiotic (HFD + PROB, <span class="html-italic">n</span> = 4–7) diets for 13 weeks by two-way ANOVA followed by Bonferroni’s multiple comparison test. Results were presented as mean ± SEM.</p>
Full article ">
Back to TopTop