Next Article in Journal
Melatonin-Regulated Chaperone Binding Protein Plays a Key Role in Cadmium Stress Tolerance in Rice, Revealed by the Functional Characterization of a Novel Serotonin N-Acetyltransferase 3 (SNAT3) in Rice
Previous Article in Journal
Identification and Functional Analysis of KH Family Genes Associated with Salt Stress in Rice
Previous Article in Special Issue
Maternal Exercise Prior to and during Gestation Induces Sex-Specific Alterations in the Mouse Placenta
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Developmental Programming of the Fetal Immune System by Maternal Western-Style Diet: Mechanisms and Implications for Disease Pathways in the Offspring

by
Benjamin N. Nelson
1 and
Jacob E. Friedman
1,2,3,*
1
Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
2
Department of Physiology and Biochemistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
3
Department of Pediatrics, Section of Diabetes and Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(11), 5951; https://doi.org/10.3390/ijms25115951
Submission received: 18 April 2024 / Revised: 24 May 2024 / Accepted: 27 May 2024 / Published: 29 May 2024

Abstract

:
Maternal obesity and over/undernutrition can have a long-lasting impact on offspring health during critical periods in the first 1000 days of life. Children born to mothers with obesity have reduced immune responses to stimuli which increase susceptibility to infections. Recently, maternal western-style diets (WSDs), high in fat and simple sugars, have been associated with skewing neonatal immune cell development, and recent evidence suggests that dysregulation of innate immunity in early life has long-term consequences on metabolic diseases and behavioral disorders in later life. Several factors contribute to abnormal innate immune tolerance or trained immunity, including changes in gut microbiota, metabolites, and epigenetic modifications. Critical knowledge gaps remain regarding the mechanisms whereby these factors impact fetal and postnatal immune cell development, especially in precursor stem cells in bone marrow and fetal liver. Components of the maternal microbiota that are transferred from mothers consuming a WSD to their offspring are understudied and identifying cause and effect on neonatal innate and adaptive immune development needs to be refined. Tools including single-cell RNA-sequencing, epigenetic analysis, and spatial location of specific immune cells in liver and bone marrow are critical for understanding immune system programming. Considering the vital role immune function plays in offspring health, it will be important to understand how maternal diets can control developmental programming of innate and adaptive immunity.

1. Introduction

Obesity is a world-wide epidemic that impacts human health at all ages, including pregnant women and their offspring, with socioeconomic consequences across generations in both developed and developing countries. While difficult to approximate, pan-global studies have estimated that about 500 million people are currently obese, and that number is expected to reach over 1 billion by 2030 [1,2]. With the rising global incidence of obesity, especially in children and women of reproductive age, it is important to understand how obesity and poor maternal diet modify neonatal outcomes, particularly in the first 1000 days from conception to 2 years of age. Critical developmental windows in a child’s life can have lifelong impacts on the structure and function of organs and tissues, including developmental programming of the immune system, which plays a significant role in disease susceptibility [3,4,5]. Maternal diet, particularly a western-style diet (WSD) high in fat and simple sugars, has been shown in animal models to drive developmental programming of the immune system, beginning in utero [6,7]. The underlying causes and mechanisms for this remodeling, its impact on future disease pathways, and the critical events that drive developmental programming in the immune system are just beginning to be investigated. The purpose of this review is to explore how exposure to excess maternal fuels during fetal life can influence innate immune cell development and responses, microbial dysbiosis, metabolic effects from metabolites, and epigenetic modifications. How these changes affect the development of mechanisms underlying long-lived chronic inflammatory diseases, such as metabolic dysfunction-associated steatotic liver disease (MASLD, formerly NAFLD), including microglial and liver function in the offspring, are reviewed. Possible interventions in pre-clinical models of disease are also discussed.

2. Immune Cell Development: Hematopoiesis

From embryo to neonate, the developing fetus undergoes many changes to its immune system. To understand the context of these developmental changes to the innate and adaptive immune systems, we provide a description of the tissues and cell types involved in normal development. Central to this process, hematopoiesis undergoes significant changes in the bone and liver during fetal development to yield all blood cells which are derived from a group of self-renewing, multilineage hematopoietic stem and progenitor cells (HSPCs) [8]. However, the notion that there is a single progenitor cell has been replaced by a layered concept, in which the various immune cells arise from predecessor cells in three broadly defined waves (Figure 1) [9,10]. The first cells observed in an embryo are not HSPCs but rather erythrocytes that supply oxygen to the fetal yolk sac [11]. These erythrocytes differ from their adult cousins in that they are nucleated and express embryonic hemoglobin [12]. In addition to these embryonic red blood cells, primitive macrophages develop within the yolk sac that are distinct from circulating macrophages. These cells travel to the fetal liver (establishing a resident population of macrophages and mast cells) before migrating to the brain as microglial cells or the lung as alveolar macrophages [13,14]. This is usually considered the first wave (termed “primitive”), which begins at about 4-weeks post-conception, as these primitive macrophages are the first immune cells to emerge from the yolk sac [15]. The second wave, beginning around 6-weeks post-conception, comes in the form of erythro-myeloid progenitors which are generated from hemogenic endothelial cells [16]. These progenitors have the potential to differentiate into macrophages, monocytes, granulocytes, and mast cells [17,18,19]. Finally, around 10 to 12-weeks post-conception, the third wave, known as definitive hematopoiesis, occurs when adult-type HSPCs emerge from the aorta gonad mesonephros region and seed the fetal liver, followed shortly thereafter by colonization of the fetal bone marrow [20]. It has been shown that this seeding of the bone marrow by HSPCs happens as early as embryonic day 15.5 in mice models [21]. These cells have the potential to generate the full spectrum of immune cells needed for proper functions throughout life [22].
In addition to myeloid cells, common lymphoid progenitor cells go through a series of developments outside of the bone marrow during the maturation of a fetus [23]. Lymphoid cells are a diverse group of immune cells, from the adaptive responses of T and B cells to their innate partners, including natural killer (NK) cells and innate lymphoid cells. Lymphoid progenitors derived from progenitor cells in the yolk sac expand in the fetal bone marrow where they account for ~40% of the total progenitor pool [24,25]. Primary development of these tissue resident immune cells happens early in fetal gestation and, once seeded into tissues, changes to “central” hematopoiesis and has little effect on the peripheral systems. Interestingly, the organs required for normal development (bone marrow and thymus) are not fully formed at this time. Despite this, cells with a similar genetic profile as lymphoid-myeloid progenitors are found within the fetal liver, suggesting that the pathways for the development of T/B cells are present at this stage [26]. Development of T-cell receptor subtypes undergo increasing diversity as development proceeds starting with invariant γδ T cells and proceeding through the more complex αβ T cells [27]. While B cells are traditionally associated with adaptive immunity, where they produce antibodies in response to specific pathogens, they also play an important role in the innate immune system. Human B cell development is a dynamic lifelong process that starts in utero in the bone marrow at around 6-weeks post-conception [28]. B cell-mediated innate immunity is the body’s immediate, nonspecific defense mechanism against pathogens. B cells can interact with gut microbiota, which play a crucial role in metabolic health [29]. Naïve B cells also show lineage divergence depending on when the cells are seeded. Tissue resident B-1 cells can only be differentiated from fetal bone marrow or fetal liver; this contrasts with conventional B-2 cells, which are derived only from adult bone marrow [30].

3. Impact of Maternal Obesity/WSD on Maternal and Placental Immune Function

Normal pregnancy proceeds with many changes in maternal immune function and a highly regulated inflammatory response is required for proper development of the fetus [31]. Maternal obesity can have many negative influences on the regulation of immunity and inflammation over the course of gestation [32]. A majority of studies demonstrate that pregnant women with obesity exhibit increased inflammation and circulating inflammatory cytokines compared with normal-weight pregnant women, and factors including fuel overload, insulin resistance, and excess gestational weight gain contribute to this inflammation [33,34,35,36]. Pre-gravid obesity impacts the systemic maternal immune system, e.g., reducing the numbers of NK T cells and plasmacytoid dendritic cells (DCs) [37,38]. Additionally, changes to the adaptive immune cells have been observed. While the total number of CD4+ T cells remained constant, the number of naïve CD4+ T cells was reported to increase in plasma across gestation in obese pregnancies [37,38]. As for CD8+ T cells, studies on maternal obesity suggest an increase in their numbers from mid to late pregnancy, possibly as a response to the growing demands of the maternal–fetal interface and the need to maintain immune surveillance [37,38].
The placenta plays a crucial role by providing oxygen, nutrients, and immune protection to the developing fetus, as well as serving as an endocrine organ that regulates maternal–fetal communication and maternal physiology. The predominant immune cells in the placenta are the uterine NK (uNK) cells and macrophages (Hofbauer cells) [39,40,41]. Unlike normal NK cells, which are cytotoxic, uNK cells are generally responsible for angiogenesis in the placenta and maternal immune tolerance towards the fetus [42]. Additionally, Hofbauer cells play a role in morphogenesis and homeostasis and display an M2 phenotype (related to tissue repair and renewal for the growing fetus) by affecting the trophoblast cells [43]. Proper regulation of these cells is crucial for normal development of the fetal immune system and maternal obesity/WSD can alter this regulation. Extracellular vesicles can provide some of this regulation by way of membrane-bound proteins and miRNAs. However, the discussion surrounding fetal regulation by extracellular vesicles is an emerging field and has been reviewed elsewhere (see [44,45]).

3.1. Maternal Obesity Changes Placental Immunity

Whether maternal obesity leads to heightened cytokine transport across the placenta, thus directly inducing inflammation in the fetus, is an open question. However, one study revealed that fetal-derived IL-8 helps recruit maternal neutrophils in normal pregnancies [46]. Additionally, it has been shown that pregnancy increases systemic inflammation and pre-gravid obesity increases this further. Excess maternal adiposity has been reported to alter the composition and function of maternal monocytes in peripheral blood, as well as macrophages found in placental and maternal adipose tissues during late gestation, toward a proinflammatory phenotype [33,47]. A recent study showed that Hofbauer cells and microglia share a similar developmental lineage and correlations in gene dysregulation were observed between the populations during maternal obesity [48]. This suggests that the readily abundant cells in the placenta provide insight into the developmental function of neonatal microglia. Higher maternal BMI has also been correlated with increased numbers of B cells during pregnancy [38]. Furthermore, pregnant women with obesity exhibit increased activation of uNK cells promoting TNF-α production [49,50]. The normal cellular population of the placenta is also altered under obese conditions; an increased accumulation of CD68+ macrophages and a reduced number of M1 (proinflammatory) polarized macrophages have been reported [51,52]. Instead of inflammation, the accumulating cells in the placenta are hyporesponsive to TLR stimulation, which is associated with systemic and adipose tissue inflammation in obese pregnancies [53,54]. These cells at the maternal–fetal interface thus become excessively tolerant to stimuli and the tolerance is then imparted onto the fetus by shared developmental lineages.

3.2. Cord Blood Changes in Offspring from Obese Pregnancies

Changes during pregnancy can have a profound impact on the immune cells and maternal-derived inflammatory cytokines, which can alter the developmental trajectory of the neonate (Figure 2). In normal human pregnancy, the fetal immune system has a helper T (Th) 2 phenotype to prevent alloimmune responses against the mother [55]. This is facilitated by a rare B cell subpopulation (regulatory B cells) and primarily by cytokines IL-10, IL-35, and TGF-β [56]. The immune tolerance function of these cells acts by inhibiting Th1 cell activation, Th17 differentiation, and maintenance of Tregs [57]. In cord blood from obese pregnancies, an increase of NK T cells and plasmacytoid DCs with a corresponding drop in CD34+ stems cells, eosinophils, and regulatory T cells (Tregs) have been shown [58]. This drop in hematopoietic stem cells may alter the host’s ability to combat a wide range of insults to the immune system and instead skew it towards predetermined remodeling. Indeed, Sureshchandra et al. found that memory T cells within cord blood from obese pregnancies were increased in number; however, the CD4+ T cells had functional defects and blunted responses to stimuli [59]. This diminished ability of T helper cells impacts their ability to produce IL-4 as it is negatively correlated with increasing maternal BMI [53]. Paradoxically, unusually high levels of inflammatory mediators, such as TNF-α and C-reactive protein, have been observed in cord blood from obese pregnancies [60,61]. All these changes lead to blunted responses in fetal immune cells, decreased placental blood flow/angiogenesis, and increased fatty acid uptake with a corresponding increase in fetal liver mass in obese gravida [62]. Further, ex vivo studies have shown that monocytes in cord blood from obese pregnancies exhibit increased susceptibility and severity to infections, which arose from aberrant monocyte responses caused in part by reduced nuclear translocation of NF-κB [7,51]. These studies showed that reduced availability of proinflammatory gene promoters led to decreased inflammatory mediators, such as IL-6, IL-10, and IL-12. Taken together, maternal obesity has the capacity to change both relative immune cell populations in the pregnant woman and in her fetus to an immune-tolerant phenotype that can impact inflammation pathways postnatally.

4. Impact of Maternal Obesity on the Fetal Liver and Offspring Immunity

MASLD is a chronic, progressive condition affecting about 37% of the global population and is strongly associated with features of the metabolic syndrome, including obesity and type 2 diabetes [63]. In animal models, maternal diet-induced obesity has been shown to modify fetal liver and immune system development and function [64,65]. Studies in rodents and nonhuman primates (NHP), in addition to human neonates, link maternal obesity with increased neonatal liver fat [66,67,68]. Further, exposure to a maternal WSD can increase the risk of the pediatric form of MASLD in adolescent youth [69]. Maternal factors that impact fetal liver development are a complex interplay involving pre-gravid obesity, dietary patterns, and inflammation. Maternal genotypes may influence the development of MASLD in offspring and thrifty genes that influence obesity may be involved. Candidate genes directly involved in obesity that affect energy balance (i.e., hypothalamic satiety receptor MC4R and adipo-genic transcription factor SREBP1 [70,71]) may contribute to genetic risks in the offspring.
MASLD can progress to its active inflammatory form, metabolic dysfunction-associated steatohepatitis (MASH), and both the innate and adaptive immune systems contribute to the progression [72]. Liver resident Kupffer cells respond to nutrient overload and liver damage by releasing chemokines CCL1, CCL2, and CCL5 to recruit monocytes and promote their polarization into M1 proinflammatory macrophages in a CCR8-dependent manner [73,74]. Chemically blocking these mediators has been shown to reduce hepatic fibrosis in human trials [75]. However, inflammation from the innate immune system is not the only driving force for increased complications in the liver, as there is an influx of T and B cells into the liver forming ectopic lymphoid structures [76]. During hepatic steatosis in mice, CD4+ T cells will produce IFN-γ from Th1 cells through the expression of T-bet, further increasing inflammation [77]. Studies using IFN-γ-deficient mice have shown decreased hepatic steatosis and fibrosis when compared with wild-type mice [78]. Similar findings have been corroborated in humans through clinical observations in both adults and children [79,80], suggesting that animal studies on the developmental effects of MASLD are translational to humans.
Maternal WSD can induce a proinflammatory response in fetal HSPCs and macrophages from NHPs [81]. Several adaptive programs have also been described in macrophages, including priming, tolerance, and trained immunity [82,83]. Specific pathways and mechanisms differ between the various adaptive programs in innate immunity, involving epigenetic, transcriptional, and metabolic reprogramming. Innate immune cells can undergo any of these functional adaptive programs, but the exact definition depends on the context under which the cells are studied and the nature of the stimulus used before and after to demonstrate an activated state of programmed immunity or inactivated state of immune tolerance to secondary stimuli [84,85]. Depending on their inflammatory polarization, macrophages in the liver either promote resolution from inflammation and prevent progression to fibrosis or remodel the tissue and provoke fibrosis. Their state of immune tolerance may affect their ability to expand in number, leading to cellular exhaustion in resident tissues in response to secondary challenges, such as infection or a WSD. Given that immune tolerance can lead to downregulation of inflammatory and restorative pathways in the liver, pathogenic immune tolerance may have broad consequences on increasing programming effects through susceptibility, initiation, or progression of fibrosis across a variety of tissues.
Fetal and neonatal T and B cells are not spared from changes to normal effector functions due to external stimuli. CD4+ and CD8+ T cells respond to antigens in HFDs to drive MASH and metabolic syndrome [77,86]. These cells are recruited into the liver by IFN-α and promote insulin resistance and glucose metabolism during HFD feeding [87]. Disruption of the gut microbiota-B cell axis may also contribute to obesity-related inflammation and metabolic dysfunction. B cells have also been implicated in the pathogenesis of MASLD, particularly in its more severe form MASH, as they are shown to be pro-fibrogenic in the form of plasma cells secreting IgA [88]. These are formed by proinflammatory mediators, which stimulate hepatic stellate cells into producing retinoic acid which is detected by the B cells [89]. Additionally, Odaka et al. found that mice born from dams on a high-fat diet (HFD) had increased IgE production [90]. Since IgE is one of the main effectors of allergy, maternal obesity may explain the increasing prevalence of childhood allergies. The adaptive arm of immunity can also be linked back to innate immunity. Independent of T cells, B-1 cells can secrete IgM natural antibodies, which can react with endogenous antigens, such as oxidized phospholipids [91]. These changes both increase the susceptibility to MASLD and drive its progression forward.

5. Microglia Activation by a Maternal WSD

Microglia are the resident immune cells of the brain and have similar functions to macrophages, such as immune defense and brain maintenance [92]. This group of self-renewing cells originate from the fetal yolk sac and any adverse effects on this population at birth can cause long-term consequences on brain function [93]. Many associations have been made between inflammation and neuropsychiatric disorders, including ADHD, autism, schizophrenia, and eating disorders [94]. Mattei et al. found that maternal immune activation with Poly(I:C) led to a downregulation of genes involved in the inflammatory response, microglial development, and phagocytosis in adult offspring [95]. However, genes involved in migration of microglial cells were upregulated, and these transcriptional changes could be reversed with treatment of minocycline, an antibiotic used as a possible treatment for schizophrenia [96]. Studies in an NHP model showed that offspring from dams with WSD-induced obesity have an inflammatory phenotype in the brain that includes increased microglial cell counts [97]. This study showed that, while pre-pregnancy adiposity was positively correlated with increased microglial cell counts, these counts were also negatively correlated with maternal WSD. The exact cause of these relationships is not entirely clear. However, further research by this group found that IL-12 was increased in the dams due to maternal WSD, which caused neurodevelopmental disorders in the offspring [98]. Another group confirmed the previous findings of decreased microglial cell counts due to maternal WSD but also showed that the deceased cell count did not persist one year postnatally [99]. This would suggest that neurodevelopmental trajectory and immune response is heavily dictated by early life events. Additionally, this activation is accompanied with the release of proinflammatory cytokines, e.g., TNF-α, IL-1β, and IL-6, which have been linked to neuronal loss, brain damage, and other neurodegenerative diseases [100]. For example, induction of the inflammasome through NLRP3 causes increased neuroinflammation and cognitive impairment and knocking out its receptor IL-1R1 can restore these deficiencies back to control levels [101]. This increased inflammation damages brain tissue and can alter the proper functioning of other resident cells. IL-6 was also found to be a critical regulator of microglial activation and it effects were abrogated either by knockout mice or neutralizing antibodies or increased by maternal IL-6 administration [102]. Lastly, IL-17A was also shown to increase microglial activation through direct administration to the fetal mouse brain [103].

6. Maternal Obesity Influences the Fetal Microbiome and Immune Development

Maternal diet alters the microbiome of both the mother and the offspring to regulate many processes, including gene expression, immune tolerance, and metabolism [104,105,106,107]. The maternal microbiome has been studied in great detail recently and studies have shown that differences in maternal diet have a profound impact on the diversity and taxa of the maternal and fetal microbiota [108,109,110,111,112,113,114,115]. Individual bacteria and their components, such as endotoxins, have been linked with changes in gut and circulating inflammatory cells, and these changes have been implicated in many chronic western diseases [104,116]. Data suggest that gross changes in abundances of obesity-enriched microbial genes are positively correlated with blood glucose levels in pregnant women with obesity and GDM and can be controlled by maternal diet, but the functionality of these changes remains unknown [117,118]. A portion of the maternal microbiome is vertically transmitted to the neonate, and early-life microbiota of the infant depends in part on the mode of delivery [119]. Infants born vaginally have a microbiome that resembles the mother’s vaginal and gut microbiota, while those born via cesarean section have a microbiome that resembles the mother’s skin microbiota and hospital environment [120]. These differences may have profound impacts on the long-term health of the infant, with research suggesting that offspring born via cesarean section are more prone to allergies, asthma, and obesity [121]. While the exact mechanisms are unclear, data show that newborns’ epigenetic landscape and immune responsiveness to microbes are due to the maternal prenatal immune status in mice, NHP, and possibly humans [6,122,123,124,125].
The process of neonatal colonization, whether it begins in utero, and the mechanisms involved are complex and not completely understood. Commensal microbes make their way to the infant through vertical transmission from the mother and their diversity is affected by perinatal conditions [126,127]. Research has shown that 72% of the neonate gut microbiome comes from the mother, but this number is substantially reduced to 25% in cases of recent maternal antibiotic use [128]. Infants exposed to antibiotics obtained antimicrobial resistant species mostly from the environment rather than through vertical transmission. This shows the potential dangers that can arise with nosocomial infections and antibiotic exposure in neonates. The hygiene hypothesis suggests that reduced diversity of early-life microbial exposure is associated with an elevated risk of allergic diseases later in life; however, studies have also linked susceptibility to allergic disorders to immune regulatory pathways triggered by some commensal microbes [129]. Soderborg et al. showed that, when microbes from 2-week-old infants born to mothers with obesity are transferred to germ-free mice, this imbalance disrupts myeloid cells (macrophage precursors) derived from the bone marrow and provokes obesity and a leaky gut leading to MASLD after a WSD challenge [130]. This metabolic rewiring of maturing macrophages is likely to be accompanied by epigenetic reprogramming that causes reduced bacterial phagocytosis and prevents liver inflammation. This may profoundly disrupt the establishment of the host–microbe symbiosis and suggests that there is a direct impact on offspring’s stem cells, which regulate immune development.
Concepts concerning the origins of developmental immune programming and how early life interactions can shape lifelong alterations and disease susceptibility through immune tolerance or trained immunity of the innate immune system have been reviewed by Hong and Medzhitov [131]. The first 1000 days of life are a critical period for proper development of a healthy gut, dominated by a diverse group of infant-type bifidobacteria, including Bifidobacterium longum, B. bifidum, and B. breve [132,133,134]. These species can metabolize human milk oligosaccharides that are abundant in breast milk and thus have a metabolic advantage in early colonization [135]. Breastfeeding has been correlated with reduced disorders, such as pathogen infection, autoimmune disorders, diabetes, and obesity [136,137]. Likewise, the acquisition of founder strains in the Enterobacteriaceae family in human newborns plays an essential role in development of normal neonatal immunity and susceptibility to metabolic diseases in later life. Enterobacteriaceae, as well as being a facilitative anaerobe, is a producer of lipopolysaccharide (LPS), important for trained innate and adaptive immunity, and is critical for normal immune tolerance [137]. Maternal diet can also shift the community of commensals [114,116]. An HFD can increase the number of anaerobes, such as Bacteroidetes, and obese individuals have a high Bacteroidetes to Firmicutes ratio; however, this is not a universal finding [138]. Table 1 lists key studies exploring the nature between the microbiome and disordered offspring immunity.
The microbiome can also have a wide range of effects on shaping and training the host immune system [144,145,146]. Normal colonization of microbiota has been shown to assist in the proper development of CD8+ T cell immunity through the priming of monocytes and macrophages, which release TNF that signals conventional DCs to produce the CD8+ T cell target IL-12p40 [147]. They can also promote early innate effectors of unconventional T cells, including mucosal-associated invariant T (MAIT) cells, NK cells, and γδ T cells [148]. Furthermore, commensals are necessary for promoting immune tolerance and creating proper immune defenses in the gut. Peripherally induced Tregs (pTregs) can support tolerance in the gut through RORγt while also abrogating Th17 responses [149,150]. Murine studies have shown that early life (2–3 weeks old) is a critical window for pTreg generation and failure to generate Tregs can lead to altered immune homeostasis and increased inflammatory intestinal diseases [151]. These alterations to immune signaling contribute to immunologic and metabolic disorders, which are normally prevented through proper priming of the immune system. This priming can occur as early as the second trimester; memory T cells were found in fetal lymph nodes, which can be activated by Staphylococcus and Lactobacillus [152]. Further, it has been shown that germ-free mice have compromised CD4+ T cell and Treg development, suggesting that microbial interaction is necessary for early immune system development [153]. Finally, serotonin was shown to be highly enriched during the first 3 weeks of life in the luminal contents from the gut of specific pathogen-free neonatal mice and is driven by the gut commensal microbiota to directly alter cell differentiation into Tregs [154].
Microbes can also directly influence the differentiation and effect of bone marrow HSPCs as the fetus develops. Studies have shown that mice lacking proper commensal microbes during development have low HSPC counts and defective myelopoiesis which resulted in impaired resistance to bacterial infections [155,156]. Recolonization of these mice with a healthy microbiota can rescue this phenotype via metabolites, microbial components, and nutritional support [157,158,159]. However, there is a complex interplay at work as not all stimuli provide the same outcome. An example of this was found in mice given different bacterial components and those supplemented with LPS had an increased pathogen killing capacity from myeloid cells compared with those given bacterial DNA instead [160]. The heterogeneity coincides with other studies that have found differences among monocytes in both mice and humans in that inflammatory environmental cues can influence the resulting population of monocytes [161,162]. These differences have lasting effects on both the myeloid cells and HSPCs [163]. Luo et al. found an overall decreased capacity for hematopoiesis with an altered microbiome brought on by HFD feeding in adult mice, and these changes were carried over by fecal transplantation of microbes from the HFD group to normal mice [164]. On the other hand, most studies of early life LPS result in dampening of innate immunity in monocytes in offspring from LPS-treated dams compared with offspring from control pregnancies, suggesting an impaired capacity to respond to infection. In infants born to mothers with chorioamnionitis, for example, monocytes have impaired production of inflammatory cytokines and lower expression of genes involved in antigen presentation and adaptive immunity [165,166]. Taken together, altered microbiomes can have a profound effect both on the proper development and maintenance of immunity during critical periods of fetal development.

7. Metabolites Regulate Fetal Metabolism and Immune Development

As noted earlier, infant-type Bifidobacteria species are well suited for colonizing the infant gut due to their ability to metabolize breast milk [132]. They produce a myriad of metabolites, including various types of short-chain fatty acids (SCFAs) and aromatic amino acids, which can directly regulate and alter host pathways (Table 2) [167,168,169,170]. The most common SCFAs in the gut are acetate, propionate, and butyrate, which are able to influence host processes through different mechanisms [171]. Butyrate is the preferred energy source for gut epithelial cells and can directly modulate genes by acting as histone deacetylases (HDACs) [172]. HDACs have been shown to suppress tumor growth in cancerous cells and have anti-inflammatory effects in healthy cells [173,174,175]. Moreover, butyrate supplementation in mammalian models has been shown to elevate beneficial gut microbiota, improve intestinal barrier function, and promote fat mobilization and utilization, resulting in the attenuation of hepatic steatosis through STAT3 signaling and HDACs [176,177,178]. Acetate and propionate are activators of G protein-coupled receptors and thus promote lipid and glucose metabolism in white adipose tissue [179,180]. Chen et al. found that maternal supplementation with propionate ameliorated intra-uterine growth restriction by downregulating glucose and lipid metabolism caused by hypoxia [181].
Another class of metabolites important for regulating inflammation are derived from the essential amino acid tryptophan (Table 2). Tryptophan regulation and utilization are important during pregnancy and are essential for proper fetal growth [182]. Tryptophan metabolites are derived from gut metabolism and are altered in obesity; higher amounts of the tryptophan product kynurenine and lower amounts of serotonin are associated with higher BMI and visceral fat mass [183,184]. Gut commensals metabolize dietary tryptophan to produce metabolites capable of modulating the host immune system, thereby linking the gut microbiota with nutrition, metabolism, and the innate immune response. The indole derivatives tryptamine and indole-3-acetate were shown to suppress the inflammatory responses of macrophages and reduce lipid production in hepatocytes [185]. Other tryptophan metabolites, e.g., indole-3-ethanol, indole-3-pyruvate, and indole-3-aldehyde, can prevent gut permeability by maintaining tight junctions [186]. Finally, indole-3-carbinol can decrease adipogenesis, thermogenesis, and inflammation by rescuing key transcription factors that become dysregulated by HFD feeding [187]. Central to most of these pathways is the aryl hydrocarbon receptor (AHR); many tryptophan metabolites are AHR ligands [188]. AHR signals the transcription factors NRF2 and NF-κB and has been shown to play a role in the progression of MASLD [189]. Ghiboub et al. reviewed the ongoing research into therapeutic treatments using tryptophan, including direct supplementation, microbiota-derived supplementation, and AHR ligand activation [190].
Table 2. Common metabolites from commensal microbiota, their mechanism of action, and the effect on the host.
Table 2. Common metabolites from commensal microbiota, their mechanism of action, and the effect on the host.
MetaboliteMechanismEffectReferences
AcetateFFAR2 and acetyl-CoA carboxylasePromoted lipid metabolism and reduced appetite[191,192]
PropionateFFAR2 and FFAR3Promoted glucose metabolism[191]
ButyrateFFAR3Anti-inflammatory effects and promoted lipid metabolism[191]
Indole-lactic AcidAHR and HCA3Improved gut health and immune responses[193]
Phenyllactic AcidMineral utilizationIncreased lymphoid cell count without infection[194]
4-Hydroxyphenyllactic AcidDCAR-1Triggered increased innate immune responses[195]
TryptamineAHRSuppressed inflammation[185]
Indole-3-acetateAHRSuppressed inflammation[185]
Indole-3-ethanolMyoIIA, ezrinMaintained gut permeability[186]
Indole-3-pyruvateMyoIIA, ezrinMaintained gut permeability[186]
Indole-3-aldehyeMyoIIA, ezrinMaintained gut permeability[186]
Indole-3-carbinolUCPs, PPAR, sirtuin-1, leptin, aP2Decreased adipogenesis, thermogenesis, and inflammation[187]

8. Role of Epigenetic Programming in Immune Cells from Offspring Exposed to WSD

Epigenetic changes due to maternal diet/obesity can occur throughout neonatal tissues and cells with lasting impact on neonatal development and beyond. Recent studies have shown that cord blood DNA methylation in neonates born to mothers with obesity correlated with maternal triglycerides and childhood adiposity [196]. In NHP studies, both HSPCs and bone marrow-derived macrophages (BMDM) from maternal WSD-exposed 3-year-old offspring were epigenetically modified to a more proinflammatory signature [6], despite weaning at 7 months to a normal chow diet. More open chromatin (higher transcription) in regions promoting glycolysis and more closed chromatin in regions relating to oxidative phosphorylation were observed in fetal mononuclear cells, suggestive of immune cell chromatin remodeling by exposure to maternal WSD. These progeny cells were then able to transform into macrophages with an increase in genes that mark early development of proinflammatory macrophages, such as FOS/JUN, NF-κB, C/EBPβ, and STAT6 [6]. These findings indicate that maternal WSD led to a classic shift, increasing glycolytic metabolism that favors the proinflammatory M1 cell type [6]. This effect may also be further modified by the tissue microenvironment which affect their proper function [197,198]. Suter et al. showed that, in NHPs, maternal WSD can alter the expression of NPAS2 (regulator of circadian genes in peripheral organs) in the offspring liver through histone modification at the promoter regions of genes favoring liver lipid deposition [199,200].
Maternal overnutrition also causes epigenetic changes within the hypothalamus of the fetal and postnatal brain [201,202,203]. Mouse studies showed that maternal WSD alters the arcuate nucleus region that controls NPY and AgRP and the appetite suppression neurons of POMC and CART [204]. Maternal overnutrition can increase the expression of NPY/AgRP while reducing POMC in the adult offspring’s brain [205,206]. Overall, this led to less sensitivity to the appetite suppression hormones leptin and insulin [205,207,208,209]. Specifically, hypermethylation occurs at the enhancer and promoter regions for POMC [210,211]. Interestingly, hypermethylation of the enhancer regions (but not the promoter) persisted into adulthood showing the prolonged programming effects of overnutrition [210]. Modulation of genes can also occur though HDACs. Suter et al. showed in the NHP model that a maternal WSD increased acetylation of histone H3 through the downregulation of the lysine deacetylase SIRT1 in the liver [212]. A study in mice showed that, in response to an HFD, Hdac5 and Hdac8 levels were increased in the hypothalamus, suggesting that they also play a role in gene expression under different metabolic conditions [213]. These alterations to the environment can alter the local hypothalamic region and, coupled with the proinflammatory response of activated microglia and astrocytes, suggests that metabolic reprogramming during gestation will drive microglia innate immune memory to shape their immune reactivity, leading to abnormalities of the brain in areas of both appetite stimulation and suppression [214,215].

9. Conclusions and Future Directions

As outlined in this review, due to maternal factors, the developing immune system in offspring goes through many changes, both functionally and superficially in development, beginning with the fetus, to the neonatal period and beyond (Figure 3). However, many challenges remain as the interpretation and translation of these findings are hampered by our relatively limited mechanistic understanding of immune programming. As single-cell omic technologies are maturing and more high-throughput data analyses are conducted, more information is being gathered on different cell populations, especially immune cell types, rare cell types, disease state cells, and the whole spectrum of cells in between, which lead to complex interactions within tissues [216]. The signals underlying development of HSPCs, specifically in the fetal liver and bone marrow under the influence of maternal WSD, and the timing and distribution of cells are of critical importance. Tools with single-cell resolution for analyses of the transcriptome, epigenetic changes, splice variants, surface protein expression, and spatial location will enable us to uncover their interactions. Analyzing these types of datasets in tandem will allow us to discover novel interactions. Additionally, despite evidence that T and B cells can be derived from the yolk sac in mice, no definitive proof of this has been reported in humans, as fate-mapping in adults from their developmental origins is not yet possible [23]. Which HSPC populations function primarily during fetal development and which populations are lifelong will be key to understanding how early factors influence disease development.
Maternal proinflammatory dietary factors, including high fat/high sugar, can alter the microbiome of both the mother and their offspring and the metabolites these microbes produce. However, the critical roles of a complex microbiome in influencing HSPCs and fetal programming/immunity are needed. The exact composition of the human maternal microbiota that is transferred to their offspring is vastly different between individuals. Therefore, protocols and mouse models mimicking the human condition for discovery of these relationships during critical windows of development need to be refined. Alterations in the maternal microbiome can lead to functional changes in offspring immune cell populations; this research will generate a diverse array of datasets, including transcriptomics, proteomics, metabolomics and, integrated together, meta-omics, to create a functional understanding of the processes of fetal immune development. This information can better inform us of the treatments needed and pre/probiotic usage during pregnancy for healthier outcomes.
Lastly, questions remain as to how to correct the immune system, especially if we medically supplement with an external source. Studies have sought to use maternal diets as an intervention for altered immunological programming and neural defects in offspring [217]. Will it be possible in the future to introduce targeted genetically modified immune cells as therapy for metabolically rewiring the immune system [218,219]? Given the increasing numbers of children and adolescents suffering from metabolic and behavioral disorders, and the role that inflammation may play in triggering the origins of disease, understanding the relationship between poor maternal diets and developmental programming of the immune system during early life may play a crucial role in maintaining the delicate balance between health and disease in the 21st century.

Author Contributions

Conceptualization, B.N.N. and J.E.F.; writing—original draft preparation, B.N.N.; writing—review and editing, B.N.N. and J.E.F.; visualization, B.N.N.; supervision, J.E.F.; project administration, J.E.F.; funding acquisition, J.E.F. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Institute of Diabetes and Digestive and Kidney Diseases grants R01DK121951 (J.E.F.) and R01DK128416 (J.E.F.).

Acknowledgments

All figures were created with BioRender.com accessed on 18 April 2024.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Finucane, M.M.; Stevens, G.A.; Cowan, M.J.; Danaei, G.; Lin, J.K.; Paciorek, C.J.; Singh, G.M.; Gutierrez, H.R.; Lu, Y.; Bahalim, A.N.; et al. National, regional, and global trends in body-mass index since 1980: Systematic analysis of health examination surveys and epidemiological studies with 960 country-years and 9.1 million participants. Lancet 2011, 377, 557–567. [Google Scholar] [CrossRef]
  2. Kelly, T.; Yang, W.; Chen, C.S.; Reynolds, K.; He, J. Global burden of obesity in 2005 and projections to 2030. Int. J. Obes. 2008, 32, 1431–1437. [Google Scholar] [CrossRef] [PubMed]
  3. Brines, J.; Rigourd, V.; Billeaud, C. The First 1000 Days of Infant. Healthcare 2022, 10, 106. [Google Scholar] [CrossRef]
  4. Likhar, A.; Patil, M.S. Importance of Maternal Nutrition in the First 1000 Days of Life and Its Effects on Child Development: A Narrative Review. Cureus 2022, 14, e30083. [Google Scholar] [CrossRef] [PubMed]
  5. Simione, M.; Moreno-Galarraga, L.; Perkins, M.; Price, S.N.; Luo, M.; Kotelchuck, M.; Blake-Lamb, T.L.; Taveras, E.M. Effects of the First 1000 Days Program, a systems-change intervention, on obesity risk factors during pregnancy. BMC Pregnancy Childbirth 2021, 21, 729. [Google Scholar] [CrossRef] [PubMed]
  6. Nash, M.J.; Dobrinskikh, E.; Soderborg, T.K.; Janssen, R.C.; Takahashi, D.L.; Dean, T.A.; Varlamov, O.; Hennebold, J.D.; Gannon, M.; Aagaard, K.M.; et al. Maternal diet alters long-term innate immune cell memory in fetal and juvenile hematopoietic stem and progenitor cells in nonhuman primate offspring. Cell Rep. 2023, 42, 112393. [Google Scholar] [CrossRef]
  7. Sureshchandra, S.; Doratt, B.M.; Mendza, N.; Varlamov, O.; Rincon, M.; Marshall, N.E.; Messaoudi, I. Maternal obesity blunts antimicrobial responses in fetal monocytes. eLife 2023, 12, e81320. [Google Scholar] [CrossRef] [PubMed]
  8. Kobayashi, M.; Yoshimoto, M. Multiple waves of fetal-derived immune cells constitute adult immune system. Immunol. Rev. 2023, 315, 11–30. [Google Scholar] [CrossRef] [PubMed]
  9. Mass, E.; Gentek, R. Fetal-Derived Immune Cells at the Roots of Lifelong Pathophysiology. Front. Cell Dev. Biol. 2021, 9, 648313. [Google Scholar] [CrossRef]
  10. Donald, K.; Finlay, B.B. Early-life interactions between the microbiota and immune system: Impact on immune system development and atopic disease. Nat. Rev. Immunol. 2023, 23, 735–748. [Google Scholar] [CrossRef]
  11. Palis, J.; Yoder, M.C. Yolk-sac hematopoiesis: The first blood cells of mouse and man. Exp. Hematol. 2001, 29, 927–936. [Google Scholar] [CrossRef] [PubMed]
  12. Brittain, T. Molecular aspects of embryonic hemoglobin function. Mol. Asp. Med. 2002, 23, 293–342. [Google Scholar] [CrossRef] [PubMed]
  13. Chia, S.L.; Kapoor, S.; Carvalho, C.; Bajenoff, M.; Gentek, R. Mast cell ontogeny: From fetal development to life-long health and disease. Immunol. Rev. 2023, 315, 31–53. [Google Scholar] [CrossRef] [PubMed]
  14. Ginhoux, F.; Greter, M.; Leboeuf, M.; Nandi, S.; See, P.; Gokhan, S.; Mehler, M.F.; Conway, S.J.; Ng, L.G.; Stanley, E.R.; et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 2010, 330, 841–845. [Google Scholar] [CrossRef] [PubMed]
  15. Tober, J.; Koniski, A.; McGrath, K.E.; Vemishetti, R.; Emerson, R.; de Mesy-Bentley, K.K.; Waugh, R.; Palis, J. The megakaryocyte lineage originates from hemangioblast precursors and is an integral component both of primitive and of definitive hematopoiesis. Blood 2007, 109, 1433–1441. [Google Scholar] [CrossRef] [PubMed]
  16. McGrath, K.E.; Frame, J.M.; Fegan, K.H.; Bowen, J.R.; Conway, S.J.; Catherman, S.C.; Kingsley, P.D.; Koniski, A.D.; Palis, J. Distinct Sources of Hematopoietic Progenitors Emerge before HSCs and Provide Functional Blood Cells in the Mammalian Embryo. Cell Rep. 2015, 11, 1892–1904. [Google Scholar] [CrossRef] [PubMed]
  17. Gomez Perdiguero, E.; Klapproth, K.; Schulz, C.; Busch, K.; Azzoni, E.; Crozet, L.; Garner, H.; Trouillet, C.; de Bruijn, M.F.; Geissmann, F.; et al. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature 2015, 518, 547–551. [Google Scholar] [CrossRef] [PubMed]
  18. Hoeffel, G.; Chen, J.; Lavin, Y.; Low, D.; Almeida, F.F.; See, P.; Beaudin, A.E.; Lum, J.; Low, I.; Forsberg, E.C.; et al. C-Myb+ erythro-myeloid progenitor-derived fetal monocytes give rise to adult tissue-resident macrophages. Immunity 2015, 42, 665–678. [Google Scholar] [CrossRef] [PubMed]
  19. Li, Z.; Liu, S.; Xu, J.; Zhang, X.; Han, D.; Liu, J.; Xia, M.; Yi, L.; Shen, Q.; Xu, S.; et al. Adult Connective Tissue-Resident Mast Cells Originate from Late Erythro-Myeloid Progenitors. Immunity 2018, 49, 640–653.e5. [Google Scholar] [CrossRef]
  20. Calvanese, V.; Mikkola, H.K.A. The genesis of human hematopoietic stem cells. Blood 2023, 142, 519–532. [Google Scholar] [CrossRef]
  21. Hall, T.; Sriram, P.; McKinney-Freeman, S. Murine Fetal Bone Marrow HSPCs Undergo a Dramatic Shift in Frequency at Birth. Blood 2019, 134, 2471. [Google Scholar] [CrossRef]
  22. Zhang, Y.; Liu, F. The evolving views of hematopoiesis: From embryo to adulthood and from in vivo to in vitro. J. Genet. Genom. 2023, 51, 3–15. [Google Scholar] [CrossRef] [PubMed]
  23. Sun, S.; Wijanarko, K.; Liani, O.; Strumila, K.; Ng, E.S.; Elefanty, A.G.; Stanley, E.G. Lymphoid cell development from fetal hematopoietic progenitors and human pluripotent stem cells. Immunol. Rev. 2023, 315, 154–170. [Google Scholar] [CrossRef] [PubMed]
  24. O’Byrne, S.; Elliott, N.; Rice, S.; Buck, G.; Fordham, N.; Garnett, C.; Godfrey, L.; Crump, N.T.; Wright, G.; Inglott, S.; et al. Discovery of a CD10-negative B-progenitor in human fetal life identifies unique ontogeny-related developmental programs. Blood 2019, 134, 1059–1071. [Google Scholar] [CrossRef] [PubMed]
  25. Yoshimoto, M.; Porayette, P.; Glosson, N.L.; Conway, S.J.; Carlesso, N.; Cardoso, A.A.; Kaplan, M.H.; Yoder, M.C. Autonomous murine T-cell progenitor production in the extra-embryonic yolk sac before HSC emergence. Blood 2012, 119, 5706–5714. [Google Scholar] [CrossRef] [PubMed]
  26. Luis, T.C.; Luc, S.; Mizukami, T.; Boukarabila, H.; Thongjuea, S.; Woll, P.S.; Azzoni, E.; Giustacchini, A.; Lutteropp, M.; Bouriez-Jones, T.; et al. Initial seeding of the embryonic thymus by immune-restricted lympho-myeloid progenitors. Nat. Immunol. 2016, 17, 1424–1435. [Google Scholar] [CrossRef] [PubMed]
  27. Krangel, M.S. Mechanics of T cell receptor gene rearrangement. Curr. Opin. Immunol. 2009, 21, 133–139. [Google Scholar] [CrossRef] [PubMed]
  28. Jackson, T.R.; Ling, R.E.; Roy, A. The Origin of B-cells: Human Fetal B Cell Development and Implications for the Pathogenesis of Childhood Acute Lymphoblastic Leukemia. Front. Immunol. 2021, 12, 637975. [Google Scholar] [CrossRef] [PubMed]
  29. Pabst, O.; Nowosad, C.R. B cells and the intestinal microbiome in time, space and place. Semin. Immunol. 2023, 69, 101806. [Google Scholar] [CrossRef]
  30. Dorshkind, K.; Crooks, G. Layered immune system development in mice and humans. Immunol. Rev. 2023, 315, 5–10. [Google Scholar] [CrossRef]
  31. Racicot, K.; Kwon, J.Y.; Aldo, P.; Silasi, M.; Mor, G. Understanding the complexity of the immune system during pregnancy. Am. J. Reprod. Immunol. 2014, 72, 107–116. [Google Scholar] [CrossRef] [PubMed]
  32. Wanaditya, G.K.; Putra, I.W.A.; Aryana, M.B.D.; Mulyana, R.S. Obesity in Pregnant Women and Its Impact on Maternal and Neonatal Morbidity. Eur. J. Med. Health Sci. 2023, 5, 17–21. [Google Scholar] [CrossRef]
  33. Bravo-Flores, E.; Mancilla-Herrera, I.; Espino, Y.S.S.; Ortiz-Ramirez, M.; Flores-Rueda, V.; Ibarguengoitia-Ochoa, F.; Ibanez, C.A.; Zambrano, E.; Solis-Paredes, M.; Perichart-Perera, O.; et al. Macrophage Populations in Visceral Adipose Tissue from Pregnant Women: Potential Role of Obesity in Maternal Inflammation. Int. J. Mol. Sci. 2018, 19, 1074. [Google Scholar] [CrossRef] [PubMed]
  34. Pendeloski, K.P.T.; Ono, E.; Torloni, M.R.; Mattar, R.; Daher, S. Maternal obesity and inflammatory mediators: A controversial association. Am. J. Reprod. Immunol. 2017, 77, e12674. [Google Scholar] [CrossRef] [PubMed]
  35. Tinius, R.A.; Blankenship, M.M.; Furgal, K.E.; Cade, W.T.; Pearson, K.J.; Rowland, N.S.; Pearson, R.C.; Hoover, D.L.; Maples, J.M. Metabolic flexibility is impaired in women who are pregnant and overweight/obese and related to insulin resistance and inflammation. Metabolism 2020, 104, 154142. [Google Scholar] [CrossRef] [PubMed]
  36. Pantham, P.; Aye, I.L.; Powell, T.L. Inflammation in maternal obesity and gestational diabetes mellitus. Placenta 2015, 36, 709–715. [Google Scholar] [CrossRef] [PubMed]
  37. Sureshchandra, S.; Marshall, N.E.; Wilson, R.M.; Barr, T.; Rais, M.; Purnell, J.Q.; Thornburg, K.L.; Messaoudi, I. Inflammatory Determinants of Pregravid Obesity in Placenta and Peripheral Blood. Front. Physiol. 2018, 9, 1089. [Google Scholar] [CrossRef] [PubMed]
  38. Sen, S.; Iyer, C.; Klebenov, D.; Histed, A.; Aviles, J.A.; Meydani, S.N. Obesity impairs cell-mediated immunity during the second trimester of pregnancy. Am. J. Obstet. Gynecol. 2013, 208, 139.e1–139.e8. [Google Scholar] [CrossRef] [PubMed]
  39. Moffett-King, A. Natural killer cells and pregnancy. Nat. Rev. Immunol. 2002, 2, 656–663. [Google Scholar] [CrossRef]
  40. Nagamatsu, T.; Schust, D.J. The contribution of macrophages to normal and pathological pregnancies. Am. J. Reprod. Immunol. 2010, 63, 460–471. [Google Scholar] [CrossRef]
  41. Suryawanshi, H.; Morozov, P.; Straus, A.; Sahasrabudhe, N.; Max, K.E.A.; Garzia, A.; Kustagi, M.; Tuschl, T.; Williams, Z. A single-cell survey of the human first-trimester placenta and decidua. Sci. Adv. 2018, 4, eaau4788. [Google Scholar] [CrossRef] [PubMed]
  42. Burke, S.D.; Barrette, V.F.; Gravel, J.; Carter, A.L.; Hatta, K.; Zhang, J.; Chen, Z.; Leno-Duran, E.; Bianco, J.; Leonard, S.; et al. Uterine NK cells, spiral artery modification and the regulation of blood pressure during mouse pregnancy. Am. J. Reprod. Immunol. 2010, 63, 472–481. [Google Scholar] [CrossRef] [PubMed]
  43. Yao, Y.; Xu, X.H.; Jin, L. Macrophage Polarization in Physiological and Pathological Pregnancy. Front. Immunol. 2019, 10, 792. [Google Scholar] [CrossRef] [PubMed]
  44. Paul, N.; Sultana, Z.; Fisher, J.J.; Maiti, K.; Smith, R. Extracellular vesicles- crucial players in human pregnancy. Placenta 2023, 140, 30–38. [Google Scholar] [CrossRef] [PubMed]
  45. Zhang, T.N.; Wang, W.; Huang, X.M.; Gao, S.Y. Non-Coding RNAs and Extracellular Vehicles: Their Role in the Pathogenesis of Gestational Diabetes Mellitus. Front. Endocrinol. 2021, 12, 664287. [Google Scholar] [CrossRef]
  46. Mir, I.N.; Chalak, L.F.; Liao, J.; Johnson-Welch, S.; Brown, L.S.; Longoria, C.; Savani, R.C.; Rosenfeld, C.R. Fetal-placental crosstalk occurs through fetal cytokine synthesis and placental clearance. Placenta 2018, 69, 1–8. [Google Scholar] [CrossRef] [PubMed]
  47. Sacks, G.P.; Studena, K.; Sargent, K.; Redman, C.W. Normal pregnancy and preeclampsia both produce inflammatory changes in peripheral blood leukocytes akin to those of sepsis. Am. J. Obs. Gynecol. 1998, 179, 80–86. [Google Scholar] [CrossRef] [PubMed]
  48. Batorsky, R.; Ceasrine, A.M.; Shook, L.L.; Kislal, S.; Bordt, E.A.; Devlin, B.A.; Perlis, R.H.; Slonim, D.K.; Bilbo, S.D.; Edlow, A.G. Hofbauer cells and fetal brain microglia share transcriptional profiles and responses to maternal diet-induced obesity. bioRxiv 2023. [Google Scholar] [CrossRef]
  49. Castellana, B.; Perdu, S.; Kim, Y.; Chan, K.; Atif, J.; Marziali, M.; Beristain, A.G. Maternal obesity alters uterine NK activity through a functional KIR2DL1/S1 imbalance. Immunol. Cell Biol. 2018, 96, 805–819. [Google Scholar] [CrossRef]
  50. Fukui, A.; Funamizu, A.; Yokota, M.; Yamada, K.; Nakamua, R.; Fukuhara, R.; Kimura, H.; Mizunuma, H. Uterine and circulating natural killer cells and their roles in women with recurrent pregnancy loss, implantation failure and preeclampsia. J. Reprod. Immunol. 2011, 90, 105–110. [Google Scholar] [CrossRef]
  51. Cifuentes-Zuniga, F.; Arroyo-Jousse, V.; Soto-Carrasco, G.; Casanello, P.; Uauy, R.; Krause, B.J.; Castro-Rodriguez, J.A. IL-10 expression in macrophages from neonates born from obese mothers is suppressed by IL-4 and LPS/INFgamma. J. Cell. Physiol. 2017, 232, 3693–3701. [Google Scholar] [CrossRef]
  52. Challier, J.C.; Basu, S.; Bintein, T.; Minium, J.; Hotmire, K.; Catalano, P.M.; Hauguel-de Mouzon, S. Obesity in pregnancy stimulates macrophage accumulation and inflammation in the placenta. Placenta 2008, 29, 274–281. [Google Scholar] [CrossRef]
  53. Wilson, R.M.; Marshall, N.E.; Jeske, D.R.; Purnell, J.Q.; Thornburg, K.; Messaoudi, I. Maternal obesity alters immune cell frequencies and responses in umbilical cord blood samples. Pediatr. Allergy Immunol. 2015, 26, 344–351. [Google Scholar] [CrossRef]
  54. Basu, S.; Haghiac, M.; Surace, P.; Challier, J.C.; Guerre-Millo, M.; Singh, K.; Waters, T.; Minium, J.; Presley, L.; Catalano, P.M.; et al. Pregravid obesity associates with increased maternal endotoxemia and metabolic inflammation. Obesity 2011, 19, 476–482. [Google Scholar] [CrossRef] [PubMed]
  55. Esteve-Sole, A.; Luo, Y.; Vlagea, A.; Deya-Martinez, A.; Yague, J.; Plaza-Martin, A.M.; Juan, M.; Alsina, L. B Regulatory Cells: Players in Pregnancy and Early Life. Int. J. Mol. Sci. 2018, 19, 2099. [Google Scholar] [CrossRef] [PubMed]
  56. Rosser, E.C.; Mauri, C. Regulatory B cells: Origin, phenotype, and function. Immunity 2015, 42, 607–612. [Google Scholar] [CrossRef]
  57. Flores-Borja, F.; Bosma, A.; Ng, D.; Reddy, V.; Ehrenstein, M.R.; Isenberg, D.A.; Mauri, C. CD19+CD24hiCD38hi B cells maintain regulatory T cells while limiting TH1 and TH17 differentiation. Sci. Transl. Med. 2013, 5, 173ra123. [Google Scholar] [CrossRef] [PubMed]
  58. Gonzalez-Espinosa, L.O.; Montiel-Cervantes, L.A.; Guerra-Marquez, A.; Penaflor-Juarez, K.; Reyes-Maldonado, E.; Vela-Ojeda, J. Maternal obesity associated with increase in natural killer T cells and CD8+ regulatory T cells in cord blood units. Transfusion 2016, 56, 1075–1081. [Google Scholar] [CrossRef] [PubMed]
  59. Sureshchandra, S.; Mendoza, N.; Jankeel, A.; Wilson, R.M.; Marshall, N.E.; Messaoudi, I. Phenotypic and Epigenetic Adaptations of Cord Blood CD4+ T Cells to Maternal Obesity. Front. Immunol. 2021, 12, 617592. [Google Scholar] [CrossRef]
  60. Dosch, N.C.; Guslits, E.F.; Weber, M.B.; Murray, S.E.; Ha, B.; Coe, C.L.; Auger, A.P.; Kling, P.J. Maternal Obesity Affects Inflammatory and Iron Indices in Umbilical Cord Blood. J. Pediatr. 2016, 172, 20–28. [Google Scholar] [CrossRef]
  61. McCloskey, K.; Ponsonby, A.L.; Collier, F.; Allen, K.; Tang, M.L.K.; Carlin, J.B.; Saffery, R.; Skilton, M.R.; Cheung, M.; Ranganathan, S.; et al. The association between higher maternal pre-pregnancy body mass index and increased birth weight, adiposity and inflammation in the newborn. Pediatr. Obes. 2018, 13, 46–53. [Google Scholar] [CrossRef] [PubMed]
  62. Sureshchandra, S.; Marshall, N.E.; Messaoudi, I. Impact of pregravid obesity on maternal and fetal immunity: Fertile grounds for reprogramming. J. Leukoc. Biol. 2019, 106, 1035–1050. [Google Scholar] [CrossRef] [PubMed]
  63. Chan, W.K.; Chuah, K.H.; Rajaram, R.B.; Lim, L.L.; Ratnasingam, J.; Vethakkan, S.R. Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): A State-of-the-Art Review. J. Obes. Metab. Syndr. 2023, 32, 197–213. [Google Scholar] [CrossRef]
  64. Alfaradhi, M.Z.; Fernandez-Twinn, D.S.; Martin-Gronert, M.S.; Musial, B.; Fowden, A.; Ozanne, S.E. Oxidative stress and altered lipid homeostasis in the programming of offspring fatty liver by maternal obesity. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2014, 307, R26–R34. [Google Scholar] [CrossRef] [PubMed]
  65. Gallardo, J.M.; Gomez-Lopez, J.; Medina-Bravo, P.; Juarez-Sanchez, F.; Contreras-Ramos, A.; Galicia-Esquivel, M.; Sanchez-Urbina, R.; Klunder-Klunder, M. Maternal obesity increases oxidative stress in the newborn. Obesity 2015, 23, 1650–1654. [Google Scholar] [CrossRef] [PubMed]
  66. Brumbaugh, D.E.; Friedman, J.E. Developmental origins of nonalcoholic fatty liver disease. Pediatr. Res. 2014, 75, 140–147. [Google Scholar] [CrossRef] [PubMed]
  67. McCurdy, C.E.; Bishop, J.M.; Williams, S.M.; Grayson, B.E.; Smith, M.S.; Friedman, J.E.; Grove, K.L. Maternal high-fat diet triggers lipotoxicity in the fetal livers of nonhuman primates. J. Clin. Investig. 2009, 119, 323–335. [Google Scholar] [CrossRef] [PubMed]
  68. Mouralidarane, A.; Soeda, J.; Visconti-Pugmire, C.; Samuelsson, A.M.; Pombo, J.; Maragkoudaki, X.; Butt, A.; Saraswati, R.; Novelli, M.; Fusai, G.; et al. Maternal obesity programs offspring nonalcoholic fatty liver disease by innate immune dysfunction in mice. Hepatology 2013, 58, 128–138. [Google Scholar] [CrossRef] [PubMed]
  69. Cohen, C.C.; Francis, E.C.; Perng, W.; Sauder, K.A.; Scherzinger, A.; Sundaram, S.S.; Shankar, K.; Dabelea, D. Exposure to maternal fuels during pregnancy and offspring hepatic fat in early childhood: The healthy start study. Pediatr. Obes. 2022, 17, e12902. [Google Scholar] [CrossRef]
  70. O’Rourke, R.W. Metabolic thrift and the genetic basis of human obesity. Ann. Surg. 2014, 259, 642–648. [Google Scholar] [CrossRef]
  71. Wells, J.C. Thrift: A guide to thrifty genes, thrifty phenotypes and thrifty norms. Int. J. Obes. 2009, 33, 1331–1338. [Google Scholar] [CrossRef] [PubMed]
  72. Sutti, S.; Albano, E. Adaptive immunity: An emerging player in the progression of NAFLD. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 81–92. [Google Scholar] [CrossRef] [PubMed]
  73. Tacke, F. Targeting hepatic macrophages to treat liver diseases. J. Hepatol. 2017, 66, 1300–1312. [Google Scholar] [CrossRef] [PubMed]
  74. Heymann, F.; Hammerich, L.; Storch, D.; Bartneck, M.; Huss, S.; Russeler, V.; Gassler, N.; Lira, S.A.; Luedde, T.; Trautwein, C.; et al. Hepatic macrophage migration and differentiation critical for liver fibrosis is mediated by the chemokine receptor C-C motif chemokine receptor 8 in mice. Hepatology 2012, 55, 898–909. [Google Scholar] [CrossRef] [PubMed]
  75. Friedman, S.L.; Ratziu, V.; Harrison, S.A.; Abdelmalek, M.F.; Aithal, G.P.; Caballeria, J.; Francque, S.; Farrell, G.; Kowdley, K.V.; Craxi, A.; et al. A randomized, placebo-controlled trial of cenicriviroc for treatment of nonalcoholic steatohepatitis with fibrosis. Hepatology 2018, 67, 1754–1767. [Google Scholar] [CrossRef] [PubMed]
  76. Pitzalis, C.; Jones, G.W.; Bombardieri, M.; Jones, S.A. Ectopic lymphoid-like structures in infection, cancer and autoimmunity. Nat. Rev. Immunol. 2014, 14, 447–462. [Google Scholar] [CrossRef] [PubMed]
  77. Sutti, S.; Jindal, A.; Locatelli, I.; Vacchiano, M.; Gigliotti, L.; Bozzola, C.; Albano, E. Adaptive immune responses triggered by oxidative stress contribute to hepatic inflammation in NASH. Hepatology 2014, 59, 886–897. [Google Scholar] [CrossRef] [PubMed]
  78. Luo, X.Y.; Takahara, T.; Kawai, K.; Fujino, M.; Sugiyama, T.; Tsuneyama, K.; Tsukada, K.; Nakae, S.; Zhong, L.; Li, X.K. IFN-γ deficiency attenuates hepatic inflammation and fibrosis in a steatohepatitis model induced by a methionine- and choline-deficient high-fat diet. Am. J. Physiol. Gastrointest. Liver Physiol. 2013, 305, G891–G899. [Google Scholar] [CrossRef]
  79. Ferreyra Solari, N.E.; Inzaugarat, M.E.; Baz, P.; De Matteo, E.; Lezama, C.; Galoppo, M.; Galoppo, C.; Chernavsky, A.C. The role of innate cells is coupled to a Th1-polarized immune response in pediatric nonalcoholic steatohepatitis. J. Clin. Immunol. 2012, 32, 611–621. [Google Scholar] [CrossRef]
  80. Inzaugarat, M.E.; Ferreyra Solari, N.E.; Billordo, L.A.; Abecasis, R.; Gadano, A.C.; Chernavsky, A.C. Altered phenotype and functionality of circulating immune cells characterize adult patients with nonalcoholic steatohepatitis. J. Clin. Immunol. 2011, 31, 1120–1130. [Google Scholar] [CrossRef]
  81. Sureshchandra, S.; Chan, C.N.; Robino, J.J.; Parmelee, L.K.; Nash, M.J.; Wesolowski, S.R.; Pietras, E.M.; Friedman, J.E.; Takahashi, D.; Shen, W.; et al. Maternal Western-style diet remodels the transcriptional landscape of fetal hematopoietic stem and progenitor cells in rhesus macaques. Stem Cell Rep. 2022, 17, 2595–2609. [Google Scholar] [CrossRef]
  82. Iwasaki, A.; Medzhitov, R. Control of adaptive immunity by the innate immune system. Nat. Immunol. 2015, 16, 343–353. [Google Scholar] [CrossRef] [PubMed]
  83. Netea, M.G.; Joosten, L.A.; Latz, E.; Mills, K.H.; Natoli, G.; Stunnenberg, H.G.; O’Neill, L.A.; Xavier, R.J. Trained immunity: A program of innate immune memory in health and disease. Science 2016, 352, aaf1098. [Google Scholar] [CrossRef] [PubMed]
  84. Kucuksezer, U.C.; Ozdemir, C.; Akdis, M.; Akdis, C.A. Influence of Innate Immunity on Immune Tolerance. Acta Med. Acad. 2020, 49, 164–180. [Google Scholar] [CrossRef] [PubMed]
  85. Bauer, M.; Weis, S.; Netea, M.G.; Wetzker, R. Remembering Pathogen Dose: Long-Term Adaptation in Innate Immunity. Trends Immunol. 2018, 39, 438–445. [Google Scholar] [CrossRef] [PubMed]
  86. Wolf, M.J.; Adili, A.; Piotrowitz, K.; Abdullah, Z.; Boege, Y.; Stemmer, K.; Ringelhan, M.; Simonavicius, N.; Egger, M.; Wohlleber, D.; et al. Metabolic activation of intrahepatic CD8+ T cells and NKT cells causes nonalcoholic steatohepatitis and liver cancer via cross-talk with hepatocytes. Cancer Cell 2014, 26, 549–564. [Google Scholar] [CrossRef] [PubMed]
  87. Ghazarian, M.; Revelo, X.S.; Nohr, M.K.; Luck, H.; Zeng, K.; Lei, H.; Tsai, S.; Schroer, S.A.; Park, Y.J.; Chng, M.H.Y.; et al. Type I Interferon Responses Drive Intrahepatic T cells to Promote Metabolic Syndrome. Sci. Immunol. 2017, 2, eaai7616. [Google Scholar] [CrossRef] [PubMed]
  88. Grohmann, M.; Wiede, F.; Dodd, G.T.; Gurzov, E.N.; Ooi, G.J.; Butt, T.; Rasmiena, A.A.; Kaur, S.; Gulati, T.; Goh, P.K.; et al. Obesity Drives STAT-1-Dependent NASH and STAT-3-Dependent HCC. Cell 2018, 175, 1289–1306.e20. [Google Scholar] [CrossRef] [PubMed]
  89. Thapa, M.; Chinnadurai, R.; Velazquez, V.M.; Tedesco, D.; Elrod, E.; Han, J.H.; Sharma, P.; Ibegbu, C.; Gewirtz, A.; Anania, F.; et al. Liver fibrosis occurs through dysregulation of MyD88-dependent innate B-cell activity. Hepatology 2015, 61, 2067–2079. [Google Scholar] [CrossRef]
  90. Odaka, Y.; Nakano, M.; Tanaka, T.; Kaburagi, T.; Yoshino, H.; Sato-Mito, N.; Sato, K. The influence of a high-fat dietary environment in the fetal period on postnatal metabolic and immune function. Obesity 2010, 18, 1688–1694. [Google Scholar] [CrossRef]
  91. Tsiantoulas, D.; Sage, A.P.; Mallat, Z.; Binder, C.J. Targeting B cells in atherosclerosis: Closing the gap from bench to bedside. Arter. Thromb. Vasc. Biol. 2015, 35, 296–302. [Google Scholar] [CrossRef] [PubMed]
  92. Magalhaes, M.S.; Potter, H.G.; Ahlback, A.; Gentek, R. Developmental programming of macrophages by early life adversity. Int. Rev. Cell Mol. Biol. 2022, 368, 213–259. [Google Scholar] [CrossRef] [PubMed]
  93. Saijo, K.; Glass, C.K. Microglial cell origin and phenotypes in health and disease. Nat. Rev. Immunol. 2011, 11, 775–787. [Google Scholar] [CrossRef] [PubMed]
  94. Rivera, H.M.; Christiansen, K.J.; Sullivan, E.L. The role of maternal obesity in the risk of neuropsychiatric disorders. Front. Neurosci. 2015, 9, 194. [Google Scholar] [CrossRef] [PubMed]
  95. Mattei, D.; Ivanov, A.; Ferrai, C.; Jordan, P.; Guneykaya, D.; Buonfiglioli, A.; Schaafsma, W.; Przanowski, P.; Deuther-Conrad, W.; Brust, P.; et al. Maternal immune activation results in complex microglial transcriptome signature in the adult offspring that is reversed by minocycline treatment. Transl. Psychiatry 2017, 7, e1120. [Google Scholar] [CrossRef] [PubMed]
  96. Solmi, M.; Veronese, N.; Thapa, N.; Facchini, S.; Stubbs, B.; Fornaro, M.; Carvalho, A.F.; Correll, C.U. Systematic review and meta-analysis of the efficacy and safety of minocycline in schizophrenia. CNS Spectr. 2017, 22, 415–426. [Google Scholar] [CrossRef] [PubMed]
  97. Dunn, G.A.; Mitchell, A.J.; Selby, M.; Fair, D.A.; Gustafsson, H.C.; Sullivan, E.L. Maternal diet and obesity shape offspring central and peripheral inflammatory outcomes in juvenile non-human primates. Brain Behav. Immun. 2022, 102, 224–236. [Google Scholar] [CrossRef] [PubMed]
  98. Mitchell, A.J.; Khambadkone, S.G.; Dunn, G.; Bagley, J.; Tamashiro, K.L.K.; Fair, D.; Gustafsson, H.; Sullivan, E.L. Maternal Western-style diet reduces social engagement and increases idiosyncratic behavior in Japanese macaque offspring. Brain Behav. Immun. 2022, 105, 109–121. [Google Scholar] [CrossRef] [PubMed]
  99. Papadakis, S.; Thompson, J.R.; Feczko, E.; Miranda-Dominguez, O.; Dunn, G.A.; Selby, M.; Mitchell, A.J.; Sullivan, E.L.; Fair, D.A. Perinatal Western-style diet exposure associated with decreased microglial counts throughout the arcuate nucleus of the hypothalamus in Japanese macaques. J. Neurophysiol. 2024, 131, 241–260. [Google Scholar] [CrossRef]
  100. Subhramanyam, C.S.; Wang, C.; Hu, Q.; Dheen, S.T. Microglia-mediated neuroinflammation in neurodegenerative diseases. Semin. Cell Dev. Biol. 2019, 94, 112–120. [Google Scholar] [CrossRef]
  101. Guo, D.H.; Yamamoto, M.; Hernandez, C.M.; Khodadadi, H.; Baban, B.; Stranahan, A.M. Visceral adipose NLRP3 impairs cognition in obesity via IL-1R1 on CX3CR1+ cells. J. Clin. Investig. 2020, 130, 1961–1976. [Google Scholar] [CrossRef] [PubMed]
  102. Smith, S.E.; Li, J.; Garbett, K.; Mirnics, K.; Patterson, P.H. Maternal immune activation alters fetal brain development through interleukin-6. J. Neurosci. 2007, 27, 10695–10702. [Google Scholar] [CrossRef] [PubMed]
  103. Sasaki, T.; Tome, S.; Takei, Y. Intraventricular IL-17A administration activates microglia and alters their localization in the mouse embryo cerebral cortex. Mol. Brain 2020, 13, 93. [Google Scholar] [CrossRef] [PubMed]
  104. Aron-Wisnewsky, J.; Vigliotti, C.; Witjes, J.; Le, P.; Holleboom, A.G.; Verheij, J.; Nieuwdorp, M.; Clement, K. Gut microbiota and human NAFLD: Disentangling microbial signatures from metabolic disorders. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 279–297. [Google Scholar] [CrossRef] [PubMed]
  105. Conway, J.; Duggal, N.A. Ageing of the gut microbiome: Potential influences on immune senescence and inflammageing. Ageing Res. Rev. 2021, 68, 101323. [Google Scholar] [CrossRef] [PubMed]
  106. Nichols, R.G.; Davenport, E.R. The relationship between the gut microbiome and host gene expression: A review. Hum. Genet. 2021, 140, 747–760. [Google Scholar] [CrossRef] [PubMed]
  107. Cox, T.O.; Lundgren, P.; Nath, K.; Thaiss, C.A. Metabolic control by the microbiome. Genome Med. 2022, 14, 80. [Google Scholar] [CrossRef]
  108. Human Microbiome Project, C. Structure, function and diversity of the healthy human microbiome. Nature 2012, 486, 207–214. [Google Scholar] [CrossRef]
  109. Turnbaugh, P.J.; Ley, R.E.; Hamady, M.; Fraser-Liggett, C.M.; Knight, R.; Gordon, J.I. The human microbiome project. Nature 2007, 449, 804–810. [Google Scholar] [CrossRef]
  110. Ley, R.E.; Backhed, F.; Turnbaugh, P.; Lozupone, C.A.; Knight, R.D.; Gordon, J.I. Obesity alters gut microbial ecology. Proc. Natl. Acad. Sci. USA 2005, 102, 11070–11075. [Google Scholar] [CrossRef]
  111. Turnbaugh, P.J.; Ley, R.E.; Mahowald, M.A.; Magrini, V.; Mardis, E.R.; Gordon, J.I. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006, 444, 1027–1031. [Google Scholar] [CrossRef] [PubMed]
  112. David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef] [PubMed]
  113. Wilson, A.S.; Koller, K.R.; Ramaboli, M.C.; Nesengani, L.T.; Ocvirk, S.; Chen, C.; Flanagan, C.A.; Sapp, F.R.; Merritt, Z.T.; Bhatti, F.; et al. Diet and the Human Gut Microbiome: An International Review. Dig. Dis. Sci. 2020, 65, 723–740. [Google Scholar] [CrossRef] [PubMed]
  114. Singh, R.K.; Chang, H.W.; Yan, D.; Lee, K.M.; Ucmak, D.; Wong, K.; Abrouk, M.; Farahnik, B.; Nakamura, M.; Zhu, T.H.; et al. Influence of diet on the gut microbiome and implications for human health. J. Transl. Med. 2017, 15, 73. [Google Scholar] [CrossRef] [PubMed]
  115. Koren, O.; Konnikova, L.; Brodin, P.; Mysorekar, I.U.; Collado, M.C. The maternal gut microbiome in pregnancy: Implications for the developing immune system. Nat. Rev. Gastroenterol. Hepatol. 2024, 21, 35–45. [Google Scholar] [CrossRef] [PubMed]
  116. Jennison, E.; Byrne, C.D. The role of the gut microbiome and diet in the pathogenesis of non-alcoholic fatty liver disease. Clin. Mol. Hepatol. 2021, 27, 22–43. [Google Scholar] [CrossRef]
  117. Kuang, Y.S.; Lu, J.H.; Li, S.H.; Li, J.H.; Yuan, M.Y.; He, J.R.; Chen, N.N.; Xiao, W.Q.; Shen, S.Y.; Qiu, L.; et al. Connections between the human gut microbiome and gestational diabetes mellitus. Gigascience 2017, 6, gix058. [Google Scholar] [CrossRef]
  118. Sugino, K.Y.; Hernandez, T.L.; Barbour, L.A.; Kofonow, J.M.; Frank, D.N.; Friedman, J.E. A maternal higher-complex carbohydrate diet increases bifidobacteria and alters early life acquisition of the infant microbiome in women with gestational diabetes mellitus. Front. Endocrinol. 2022, 13, 921464. [Google Scholar] [CrossRef]
  119. Miko, E.; Csaszar, A.; Bodis, J.; Kovacs, K. The Maternal-Fetal Gut Microbiota Axis: Physiological Changes, Dietary Influence, and Modulation Possibilities. Life 2022, 12, 424. [Google Scholar] [CrossRef]
  120. Arboleya, S.; Suarez, M.; Fernandez, N.; Mantecon, L.; Solis, G.; Gueimonde, M.; de Los Reyes-Gavilan, C.G. C-section and the Neonatal Gut Microbiome Acquisition: Consequences for Future Health. Ann. Nutr. Metab. 2018, 73 (Suppl. S3), 17–23. [Google Scholar] [CrossRef]
  121. Rios-Covian, D.; Langella, P.; Martin, R. From Short- to Long-Term Effects of C-Section Delivery on Microbiome Establishment and Host Health. Microorganisms 2021, 9, 2122. [Google Scholar] [CrossRef] [PubMed]
  122. DeVries, A.; McCauley, K.; Fadrosh, D.; Fujimura, K.E.; Stern, D.A.; Lynch, S.V.; Vercelli, D. Maternal prenatal immunity, neonatal trained immunity, and early airway microbiota shape childhood asthma development. Allergy 2022, 77, 3617–3628. [Google Scholar] [CrossRef] [PubMed]
  123. Thorburn, A.N.; McKenzie, C.I.; Shen, S.; Stanley, D.; Macia, L.; Mason, L.J.; Roberts, L.K.; Wong, C.H.; Shim, R.; Robert, R.; et al. Evidence that asthma is a developmental origin disease influenced by maternal diet and bacterial metabolites. Nat. Commun. 2015, 6, 7320. [Google Scholar] [CrossRef] [PubMed]
  124. Kimura, I.; Miyamoto, J.; Ohue-Kitano, R.; Watanabe, K.; Yamada, T.; Onuki, M.; Aoki, R.; Isobe, Y.; Kashihara, D.; Inoue, D.; et al. Maternal gut microbiota in pregnancy influences offspring metabolic phenotype in mice. Science 2020, 367, eaaw8429. [Google Scholar] [CrossRef] [PubMed]
  125. van Dijk, S.J.; Zhou, J.; Peters, T.J.; Buckley, M.; Sutcliffe, B.; Oytam, Y.; Gibson, R.A.; McPhee, A.; Yelland, L.N.; Makrides, M.; et al. Effect of prenatal DHA supplementation on the infant epigenome: Results from a randomized controlled trial. Clin. Epigenet. 2016, 8, 114. [Google Scholar] [CrossRef] [PubMed]
  126. Yassour, M.; Vatanen, T.; Siljander, H.; Hamalainen, A.M.; Harkonen, T.; Ryhanen, S.J.; Franzosa, E.A.; Vlamakis, H.; Huttenhower, C.; Gevers, D.; et al. Natural history of the infant gut microbiome and impact of antibiotic treatment on bacterial strain diversity and stability. Sci. Transl. Med. 2016, 8, 343ra381. [Google Scholar] [CrossRef] [PubMed]
  127. Wang, S.; Ryan, C.A.; Boyaval, P.; Dempsey, E.M.; Ross, R.P.; Stanton, C. Maternal Vertical Transmission Affecting Early-Life Microbiota Development. Trends Microbiol. 2020, 28, 28–45. [Google Scholar] [CrossRef] [PubMed]
  128. Li, W.; Tapiainen, T.; Brinkac, L.; Lorenzi, H.A.; Moncera, K.; Tejesvi, M.V.; Salo, J.; Nelson, K.E. Vertical Transmission of Gut Microbiome and Antimicrobial Resistance Genes in Infants Exposed to Antibiotics at Birth. J. Infect. Dis. 2021, 224, 1236–1246. [Google Scholar] [CrossRef] [PubMed]
  129. Guarner, F.; Bourdet-Sicard, R.; Brandtzaeg, P.; Gill, H.S.; McGuirk, P.; van Eden, W.; Versalovic, J.; Weinstock, J.V.; Rook, G.A. Mechanisms of disease: The hygiene hypothesis revisited. Nat. Clin. Pract. Gastroenterol. Hepatol. 2006, 3, 275–284. [Google Scholar] [CrossRef]
  130. Soderborg, T.K.; Clark, S.E.; Mulligan, C.E.; Janssen, R.C.; Babcock, L.; Ir, D.; Young, B.; Krebs, N.; Lemas, D.J.; Johnson, L.K.; et al. The gut microbiota in infants of obese mothers increases inflammation and susceptibility to NAFLD. Nat. Commun. 2018, 9, 4462. [Google Scholar] [CrossRef]
  131. Hong, J.Y.; Medzhitov, R. On developmental programming of the immune system. Trends Immunol. 2023, 44, 877–889. [Google Scholar] [CrossRef] [PubMed]
  132. Lin, C.; Lin, Y.; Zhang, H.; Wang, G.; Zhao, J.; Zhang, H.; Chen, W. Intestinal ‘Infant-Type’ Bifidobacteria Mediate Immune System Development in the First 1000 Days of Life. Nutrients 2022, 14, 1498. [Google Scholar] [CrossRef] [PubMed]
  133. Turroni, F.; Peano, C.; Pass, D.A.; Foroni, E.; Severgnini, M.; Claesson, M.J.; Kerr, C.; Hourihane, J.; Murray, D.; Fuligni, F.; et al. Diversity of bifidobacteria within the infant gut microbiota. PLoS ONE 2012, 7, e36957. [Google Scholar] [CrossRef] [PubMed]
  134. Turroni, F.; van Sinderen, D.; Ventura, M. Genomics and ecological overview of the genus Bifidobacterium. Int. J. Food Microbiol. 2011, 149, 37–44. [Google Scholar] [CrossRef] [PubMed]
  135. Asakuma, S.; Hatakeyama, E.; Urashima, T.; Yoshida, E.; Katayama, T.; Yamamoto, K.; Kumagai, H.; Ashida, H.; Hirose, J.; Kitaoka, M. Physiology of consumption of human milk oligosaccharides by infant gut-associated bifidobacteria. J. Biol. Chem. 2011, 286, 34583–34592. [Google Scholar] [CrossRef] [PubMed]
  136. Russell, J.T.; Roesch, L.F.W.; Ordberg, M.; Ilonen, J.; Atkinson, M.A.; Schatz, D.A.; Triplett, E.W.; Ludvigsson, J. Genetic risk for autoimmunity is associated with distinct changes in the human gut microbiome. Nat. Commun. 2019, 10, 3621. [Google Scholar] [CrossRef] [PubMed]
  137. Vatanen, T.; Kostic, A.D.; d’Hennezel, E.; Siljander, H.; Franzosa, E.A.; Yassour, M.; Kolde, R.; Vlamakis, H.; Arthur, T.D.; Hamalainen, A.M.; et al. Variation in Microbiome LPS Immunogenicity Contributes to Autoimmunity in Humans. Cell 2016, 165, 842–853. [Google Scholar] [CrossRef] [PubMed]
  138. Ley, R.E.; Turnbaugh, P.J.; Klein, S.; Gordon, J.I. Microbial ecology: Human gut microbes associated with obesity. Nature 2006, 444, 1022–1023. [Google Scholar] [CrossRef]
  139. Weaver, L.K.; Minichino, D.; Biswas, C.; Chu, N.; Lee, J.J.; Bittinger, K.; Albeituni, S.; Nichols, K.E.; Behrens, E.M. Microbiota-dependent signals are required to sustain TLR-mediated immune responses. JCI Insight 2019, 4, e124370. [Google Scholar] [CrossRef]
  140. Soderborg, T.K.; Carpenter, C.M.; Janssen, R.C.; Weir, T.L.; Robertson, C.E.; Ir, D.; Young, B.E.; Krebs, N.F.; Hernandez, T.L.; Barbour, L.A.; et al. Gestational Diabetes Is Uniquely Associated With Altered Early Seeding of the Infant Gut Microbiota. Front. Endocrinol. 2020, 11, 603021. [Google Scholar] [CrossRef]
  141. Chen, T.; Qin, Y.; Chen, M.; Zhang, Y.; Wang, X.; Dong, T.; Chen, G.; Sun, X.; Lu, T.; White, R.A., III; et al. Gestational diabetes mellitus is associated with the neonatal gut microbiota and metabolome. BMC Med. 2021, 19, 120. [Google Scholar] [CrossRef] [PubMed]
  142. Pinto, Y.; Frishman, S.; Turjeman, S.; Eshel, A.; Nuriel-Ohayon, M.; Shrossel, O.; Ziv, O.; Walters, W.; Parsonnet, J.; Ley, C.; et al. Gestational diabetes is driven by microbiota-induced inflammation months before diagnosis. Gut 2023, 72, 918–928. [Google Scholar] [CrossRef] [PubMed]
  143. Jones, J.M.; Reinke, S.N.; Mousavi-Derazmahalleh, M.; Garssen, J.; Jenmalm, M.C.; Srinivasjois, R.; Silva, D.; Keelan, J.; Prescott, S.L.; Palmer, D.J.; et al. Maternal prebiotic supplementation during pregnancy and lactation modifies the microbiome and short chain fatty acid profile of both mother and infant. Clin. Nutr. 2024, 43, 969–980. [Google Scholar] [CrossRef] [PubMed]
  144. Reynolds, H.M.; Bettini, M.L. Early-life microbiota-immune homeostasis. Front. Immunol. 2023, 14, 1266876. [Google Scholar] [CrossRef] [PubMed]
  145. Barman, P.K.; Goodridge, H.S. Microbial Sensing by Hematopoietic Stem and Progenitor Cells. Stem Cells 2022, 40, 14–21. [Google Scholar] [CrossRef] [PubMed]
  146. Paucar Iza, Y.A.; Brown, C.C. Early life imprinting of intestinal immune tolerance and tissue homeostasis. Immunol. Rev. 2024, 323, 303–315. [Google Scholar] [CrossRef] [PubMed]
  147. Kohler, A.; Delbauve, S.; Smout, J.; Torres, D.; Flamand, V. Very early-life exposure to microbiota-induced TNF drives the maturation of neonatal pre-cDC1. Gut 2021, 70, 511–521. [Google Scholar] [CrossRef]
  148. Constantinides, M.G.; Belkaid, Y. Early-life imprinting of unconventional T cells and tissue homeostasis. Science 2021, 374, eabf0095. [Google Scholar] [CrossRef] [PubMed]
  149. Sefik, E.; Geva-Zatorsky, N.; Oh, S.; Konnikova, L.; Zemmour, D.; McGuire, A.M.; Burzyn, D.; Ortiz-Lopez, A.; Lobera, M.; Yang, J.; et al. MUCOSAL IMMUNOLOGY. Individual intestinal symbionts induce a distinct population of RORγ+ regulatory T cells. Science 2015, 349, 993–997. [Google Scholar] [CrossRef]
  150. Eshleman, E.M.; Shao, T.Y.; Woo, V.; Rice, T.; Engleman, L.; Didriksen, B.J.; Whitt, J.; Haslam, D.B.; Way, S.S.; Alenghat, T. Intestinal epithelial HDAC3 and MHC class II coordinate microbiota-specific immunity. J. Clin. Investig. 2023, 133, 1–13. [Google Scholar] [CrossRef]
  151. Al Nabhani, Z.; Dulauroy, S.; Marques, R.; Cousu, C.; Al Bounny, S.; Dejardin, F.; Sparwasser, T.; Berard, M.; Cerf-Bensussan, N.; Eberl, G. A Weaning Reaction to Microbiota Is Required for Resistance to Immunopathologies in the Adult. Immunity 2019, 50, 1276–1288.e5. [Google Scholar] [CrossRef] [PubMed]
  152. Mishra, A.; Lai, G.C.; Yao, L.J.; Aung, T.T.; Shental, N.; Rotter-Maskowitz, A.; Shepherdson, E.; Singh, G.S.N.; Pai, R.; Shanti, A.; et al. Microbial exposure during early human development primes fetal immune cells. Cell 2021, 184, 3394–3409.e20. [Google Scholar] [CrossRef] [PubMed]
  153. Hu, M.; Eviston, D.; Hsu, P.; Marino, E.; Chidgey, A.; Santner-Nanan, B.; Wong, K.; Richards, J.L.; Yap, Y.A.; Collier, F.; et al. Decreased maternal serum acetate and impaired fetal thymic and regulatory T cell development in preeclampsia. Nat. Commun. 2019, 10, 3031. [Google Scholar] [CrossRef] [PubMed]
  154. Sanidad, K.Z.; Rager, S.L.; Carrow, H.C.; Ananthanarayanan, A.; Callaghan, R.; Hart, L.R.; Li, T.; Ravisankar, P.; Brown, J.A.; Amir, M.; et al. Gut bacteria-derived serotonin promotes immune tolerance in early life. Sci. Immunol. 2024, 9, eadj4775. [Google Scholar] [CrossRef] [PubMed]
  155. Khosravi, A.; Yanez, A.; Price, J.G.; Chow, A.; Merad, M.; Goodridge, H.S.; Mazmanian, S.K. Gut microbiota promote hematopoiesis to control bacterial infection. Cell Host Microbe 2014, 15, 374–381. [Google Scholar] [CrossRef] [PubMed]
  156. Josefsdottir, K.S.; Baldridge, M.T.; Kadmon, C.S.; King, K.Y. Antibiotics impair murine hematopoiesis by depleting the intestinal microbiota. Blood 2017, 129, 729–739. [Google Scholar] [CrossRef] [PubMed]
  157. Staffas, A.; Burgos da Silva, M.; Slingerland, A.E.; Lazrak, A.; Bare, C.J.; Holman, C.D.; Docampo, M.D.; Shono, Y.; Durham, B.; Pickard, A.J.; et al. Nutritional Support from the Intestinal Microbiota Improves Hematopoietic Reconstitution after Bone Marrow Transplantation in Mice. Cell Host Microbe 2018, 23, 447–457.e4. [Google Scholar] [CrossRef]
  158. Trompette, A.; Gollwitzer, E.S.; Yadava, K.; Sichelstiel, A.K.; Sprenger, N.; Ngom-Bru, C.; Blanchard, C.; Junt, T.; Nicod, L.P.; Harris, N.L.; et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat. Med. 2014, 20, 159–166. [Google Scholar] [CrossRef]
  159. Clarke, T.B.; Davis, K.M.; Lysenko, E.S.; Zhou, A.Y.; Yu, Y.; Weiser, J.N. Recognition of peptidoglycan from the microbiota by Nod1 enhances systemic innate immunity. Nat. Med. 2010, 16, 228–231. [Google Scholar] [CrossRef]
  160. Yanez, A.; Coetzee, S.G.; Olsson, A.; Muench, D.E.; Berman, B.P.; Hazelett, D.J.; Salomonis, N.; Grimes, H.L.; Goodridge, H.S. Granulocyte-Monocyte Progenitors and Monocyte-Dendritic Cell Progenitors Independently Produce Functionally Distinct Monocytes. Immunity 2017, 47, 890–902.e4. [Google Scholar] [CrossRef]
  161. Menezes, S.; Melandri, D.; Anselmi, G.; Perchet, T.; Loschko, J.; Dubrot, J.; Patel, R.; Gautier, E.L.; Hugues, S.; Longhi, M.P.; et al. The Heterogeneity of Ly6Chi Monocytes Controls Their Differentiation into iNOS+ Macrophages or Monocyte-Derived Dendritic Cells. Immunity 2016, 45, 1205–1218. [Google Scholar] [CrossRef] [PubMed]
  162. Villani, A.C.; Satija, R.; Reynolds, G.; Sarkizova, S.; Shekhar, K.; Fletcher, J.; Griesbeck, M.; Butler, A.; Zheng, S.; Lazo, S.; et al. Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science 2017, 356, eaah4573. [Google Scholar] [CrossRef] [PubMed]
  163. Jonscher, K.R.; Abrams, J.; Friedman, J.E. Maternal Diet Alters Trained Immunity in the Pathogenesis of Pediatric NAFLD. J. Cell. Immunol. 2020, 2, 315–325. [Google Scholar] [CrossRef] [PubMed]
  164. Luo, Y.; Chen, G.L.; Hannemann, N.; Ipseiz, N.; Kronke, G.; Bauerle, T.; Munos, L.; Wirtz, S.; Schett, G.; Bozec, A. Microbiota from Obese Mice Regulate Hematopoietic Stem Cell Differentiation by Altering the Bone Niche. Cell Metab. 2015, 22, 886–894. [Google Scholar] [CrossRef]
  165. Bermick, J.; Schaller, M. Chorioamnionitis exposure dampens the preterm monocyte response to subsequent challenges. Immunol. Cell Biol. 2018, 96, 789–791. [Google Scholar] [CrossRef] [PubMed]
  166. de Jong, E.; Hancock, D.G.; Wells, C.; Richmond, P.; Simmer, K.; Burgner, D.; Strunk, T.; Currie, A.J. Exposure to chorioamnionitis alters the monocyte transcriptional response to the neonatal pathogen Staphylococcus epidermidis. Immunol. Cell Biol. 2018, 96, 792–804. [Google Scholar] [CrossRef]
  167. Rios-Covian, D.; Ruas-Madiedo, P.; Margolles, A.; Gueimonde, M.; de Los Reyes-Gavilan, C.G.; Salazar, N. Intestinal Short Chain Fatty Acids and their Link with Diet and Human Health. Front. Microbiol. 2016, 7, 185. [Google Scholar] [CrossRef] [PubMed]
  168. Koh, A.; De Vadder, F.; Kovatcheva-Datchary, P.; Backhed, F. From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites. Cell 2016, 165, 1332–1345. [Google Scholar] [CrossRef] [PubMed]
  169. Hsu, C.N.; Tain, Y.L. Developmental Programming and Reprogramming of Hypertension and Kidney Disease: Impact of Tryptophan Metabolism. Int. J. Mol. Sci. 2020, 21, 8705. [Google Scholar] [CrossRef]
  170. Patil, N.Y.; Friedman, J.E.; Joshi, A.D. Role of Hepatic Aryl Hydrocarbon Receptor in Non-Alcoholic Fatty Liver Disease. Receptors 2023, 2, 1–15. [Google Scholar] [CrossRef]
  171. Cummings, J.H.; Pomare, E.W.; Branch, W.J.; Naylor, C.P.; Macfarlane, G.T. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut 1987, 28, 1221–1227. [Google Scholar] [CrossRef] [PubMed]
  172. Johnstone, R.W. Histone-deacetylase inhibitors: Novel drugs for the treatment of cancer. Nat. Rev. Drug Discov. 2002, 1, 287–299. [Google Scholar] [CrossRef] [PubMed]
  173. Chang, P.V.; Hao, L.; Offermanns, S.; Medzhitov, R. The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc. Natl. Acad. Sci. USA 2014, 111, 2247–2252. [Google Scholar] [CrossRef]
  174. Flint, H.J.; Scott, K.P.; Louis, P.; Duncan, S.H. The role of the gut microbiota in nutrition and health. Nat. Rev. Gastroenterol. Hepatol. 2012, 9, 577–589. [Google Scholar] [CrossRef] [PubMed]
  175. Singh, N.; Thangaraju, M.; Prasad, P.D.; Martin, P.M.; Lambert, N.A.; Boettger, T.; Offermanns, S.; Ganapathy, V. Blockade of dendritic cell development by bacterial fermentation products butyrate and propionate through a transporter (Slc5a8)-dependent inhibition of histone deacetylases. J. Biol. Chem. 2010, 285, 27601–27608. [Google Scholar] [CrossRef]
  176. Beisner, J.; Filipe Rosa, L.; Kaden-Volynets, V.; Stolzer, I.; Gunther, C.; Bischoff, S.C. Prebiotic Inulin and Sodium Butyrate Attenuate Obesity-Induced Intestinal Barrier Dysfunction by Induction of Antimicrobial Peptides. Front. Immunol. 2021, 12, 678360. [Google Scholar] [CrossRef]
  177. Fang, W.; Xue, H.; Chen, X.; Chen, K.; Ling, W. Supplementation with Sodium Butyrate Modulates the Composition of the Gut Microbiota and Ameliorates High-Fat Diet-Induced Obesity in Mice. J. Nutr. 2019, 149, 747–754. [Google Scholar] [CrossRef] [PubMed]
  178. Zhou, D.; Pan, Q.; Xin, F.Z.; Zhang, R.N.; He, C.X.; Chen, G.Y.; Liu, C.; Chen, Y.W.; Fan, J.G. Sodium butyrate attenuates high-fat diet-induced steatohepatitis in mice by improving gut microbiota and gastrointestinal barrier. World J. Gastroenterol. 2017, 23, 60–75. [Google Scholar] [CrossRef] [PubMed]
  179. Kimura, I.; Ozawa, K.; Inoue, D.; Imamura, T.; Kimura, K.; Maeda, T.; Terasawa, K.; Kashihara, D.; Hirano, K.; Tani, T.; et al. The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43. Nat. Commun. 2013, 4, 1829. [Google Scholar] [CrossRef]
  180. Le Poul, E.; Loison, C.; Struyf, S.; Springael, J.Y.; Lannoy, V.; Decobecq, M.E.; Brezillon, S.; Dupriez, V.; Vassart, G.; Van Damme, J.; et al. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J. Biol. Chem. 2003, 278, 25481–25489. [Google Scholar] [CrossRef]
  181. Chen, D.; Wang, Y.Y.; Li, S.P.; Zhao, H.M.; Jiang, F.J.; Wu, Y.X.; Tong, Y.; Pang, Q.F. Maternal propionate supplementation ameliorates glucose and lipid metabolic disturbance in hypoxia-induced fetal growth restriction. Food Funct. 2022, 13, 10724–10736. [Google Scholar] [CrossRef] [PubMed]
  182. Badawy, A.A. Tryptophan metabolism, disposition and utilization in pregnancy. Biosci. Rep. 2015, 35, e00261. [Google Scholar] [CrossRef] [PubMed]
  183. Cussotto, S.; Delgado, I.; Anesi, A.; Dexpert, S.; Aubert, A.; Beau, C.; Forestier, D.; Ledaguenel, P.; Magne, E.; Mattivi, F.; et al. Tryptophan Metabolic Pathways Are Altered in Obesity and Are Associated With Systemic Inflammation. Front. Immunol. 2020, 11, 557. [Google Scholar] [CrossRef] [PubMed]
  184. Lischka, J.; Schanzer, A.; Baumgartner, M.; de Gier, C.; Greber-Platzer, S.; Zeyda, M. Tryptophan Metabolism Is Associated with BMI and Adipose Tissue Mass and Linked to Metabolic Disease in Pediatric Obesity. Nutrients 2022, 14, 286. [Google Scholar] [CrossRef] [PubMed]
  185. Krishnan, S.; Ding, Y.; Saedi, N.; Choi, M.; Sridharan, G.V.; Sherr, D.H.; Yarmush, M.L.; Alaniz, R.C.; Jayaraman, A.; Lee, K. Gut Microbiota-Derived Tryptophan Metabolites Modulate Inflammatory Response in Hepatocytes and Macrophages. Cell Rep. 2018, 23, 1099–1111. [Google Scholar] [CrossRef] [PubMed]
  186. Scott, S.A.; Fu, J.; Chang, P.V. Microbial tryptophan metabolites regulate gut barrier function via the aryl hydrocarbon receptor. Proc. Natl. Acad. Sci. USA 2020, 117, 19376–19387. [Google Scholar] [CrossRef] [PubMed]
  187. Choi, Y.; Kim, Y.; Park, S.; Lee, K.W.; Park, T. Indole-3-carbinol prevents diet-induced obesity through modulation of multiple genes related to adipogenesis, thermogenesis or inflammation in the visceral adipose tissue of mice. J. Nutr. Biochem. 2012, 23, 1732–1739. [Google Scholar] [CrossRef]
  188. Shinde, R.; McGaha, T.L. The Aryl Hydrocarbon Receptor: Connecting Immunity to the Microenvironment. Trends Immunol. 2018, 39, 1005–1020. [Google Scholar] [CrossRef] [PubMed]
  189. Bock, K.W. Aryl hydrocarbon receptor (AHR), integrating energy metabolism and microbial or obesity-mediated inflammation. Biochem. Pharmacol. 2021, 184, 114346. [Google Scholar] [CrossRef]
  190. Ghiboub, M.; Verburgt, C.M.; Sovran, B.; Benninga, M.A.; de Jonge, W.J.; Van Limbergen, J.E. Nutritional Therapy to Modulate Tryptophan Metabolism and Aryl Hydrocarbon-Receptor Signaling Activation in Human Diseases. Nutrients 2020, 12, 2846. [Google Scholar] [CrossRef]
  191. den Besten, G.; van Eunen, K.; Groen, A.K.; Venema, K.; Reijngoud, D.J.; Bakker, B.M. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J. Lipid Res. 2013, 54, 2325–2340. [Google Scholar] [CrossRef] [PubMed]
  192. Frost, G.; Sleeth, M.L.; Sahuri-Arisoylu, M.; Lizarbe, B.; Cerdan, S.; Brody, L.; Anastasovska, J.; Ghourab, S.; Hankir, M.; Zhang, S.; et al. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat. Commun. 2014, 5, 3611. [Google Scholar] [CrossRef]
  193. Laursen, M.F.; Sakanaka, M.; von Burg, N.; Morbe, U.; Andersen, D.; Moll, J.M.; Pekmez, C.T.; Rivollier, A.; Michaelsen, K.F.; Molgaard, C.; et al. Bifidobacterium species associated with breastfeeding produce aromatic lactic acids in the infant gut. Nat. Microbiol. 2021, 6, 1367–1382. [Google Scholar] [CrossRef] [PubMed]
  194. Wang, J.P.; Yoo, J.S.; Lee, J.H.; Jang, H.D.; Kim, H.J.; Shin, S.O.; Seong, S.I.; Kim, I.H. Effects of phenyllactic acid on growth performance, nutrient digestibility, microbial shedding, and blood profile in pigs. J. Anim. Sci. 2009, 87, 3235–3243. [Google Scholar] [CrossRef]
  195. Zugasti, O.; Bose, N.; Squiban, B.; Belougne, J.; Kurz, C.L.; Schroeder, F.C.; Pujol, N.; Ewbank, J.J. Activation of a G protein-coupled receptor by its endogenous ligand triggers the innate immune response of Caenorhabditis elegans. Nat. Immunol. 2014, 15, 833–838. [Google Scholar] [CrossRef]
  196. Waldrop, S.W.; Niemiec, S.; Wood, C.; Gyllenhammer, L.E.; Jansson, T.; Friedman, J.E.; Tryggestad, J.B.; Borengasser, S.J.; Davidson, E.J.; Yang, I.V.; et al. Cord blood DNA methylation of immune and lipid metabolism genes is associated with maternal triglycerides and child adiposity. Obesity 2024, 32, 187–199. [Google Scholar] [CrossRef] [PubMed]
  197. van der Heijden, C.; Noz, M.P.; Joosten, L.A.B.; Netea, M.G.; Riksen, N.P.; Keating, S.T. Epigenetics and Trained Immunity. Antioxid. Redox Signal 2018, 29, 1023–1040. [Google Scholar] [CrossRef] [PubMed]
  198. Fanucchi, S.; Dominguez-Andres, J.; Joosten, L.A.B.; Netea, M.G.; Mhlanga, M.M. The Intersection of Epigenetics and Metabolism in Trained Immunity. Immunity 2021, 54, 32–43. [Google Scholar] [CrossRef] [PubMed]
  199. Suter, M.; Bocock, P.; Showalter, L.; Hu, M.; Shope, C.; McKnight, R.; Grove, K.; Lane, R.; Aagaard-Tillery, K. Epigenomics: Maternal high-fat diet exposure in utero disrupts peripheral circadian gene expression in nonhuman primates. FASEB J. 2011, 25, 714–726. [Google Scholar] [CrossRef]
  200. Suter, M.A.; Takahashi, D.; Grove, K.L.; Aagaard, K.M. Postweaning exposure to a high-fat diet is associated with alterations to the hepatic histone code in Japanese macaques. Pediatr. Res. 2013, 74, 252–258. [Google Scholar] [CrossRef]
  201. Harmancioglu, B.; Kabaran, S. Maternal high fat diets: Impacts on offspring obesity and epigenetic hypothalamic programming. Front. Genet. 2023, 14, 1158089. [Google Scholar] [CrossRef]
  202. Borengasser, S.J.; Zhong, Y.; Kang, P.; Lindsey, F.; Ronis, M.J.; Badger, T.M.; Gomez-Acevedo, H.; Shankar, K. Maternal obesity enhances white adipose tissue differentiation and alters genome-scale DNA methylation in male rat offspring. Endocrinology 2013, 154, 4113–4125. [Google Scholar] [CrossRef] [PubMed]
  203. Reynolds, C.M.; Segovia, S.A.; Vickers, M.H. Experimental Models of Maternal Obesity and Neuroendocrine Programming of Metabolic Disorders in Offspring. Front. Endocrinol. 2017, 8, 245. [Google Scholar] [CrossRef] [PubMed]
  204. Ross, M.G.; Desai, M. Developmental programming of appetite/satiety. Ann. Nutr. Metab. 2014, 64 (Suppl. S1), 36–44. [Google Scholar] [CrossRef] [PubMed]
  205. Chen, H.; Simar, D.; Morris, M.J. Hypothalamic neuroendocrine circuitry is programmed by maternal obesity: Interaction with postnatal nutritional environment. PLoS ONE 2009, 4, e6259. [Google Scholar] [CrossRef]
  206. Vickers, M.H.; Breier, B.H.; Cutfield, W.S.; Hofman, P.L.; Gluckman, P.D. Fetal origins of hyperphagia, obesity, and hypertension and postnatal amplification by hypercaloric nutrition. Am. J. Physiol. Endocrinol. Metab. 2000, 279, E83–E87. [Google Scholar] [CrossRef]
  207. Desai, M.; Han, G.; Ross, M.G. Programmed hyperphagia in offspring of obese dams: Altered expression of hypothalamic nutrient sensors, neurogenic factors and epigenetic modulators. Appetite 2016, 99, 193–199. [Google Scholar] [CrossRef]
  208. Sardinha, F.L.; Telles, M.M.; Albuquerque, K.T.; Oyama, L.M.; Guimaraes, P.A.; Santos, O.F.; Ribeiro, E.B. Gender difference in the effect of intrauterine malnutrition on the central anorexigenic action of insulin in adult rats. Nutrition 2006, 22, 1152–1161. [Google Scholar] [CrossRef]
  209. Galjaard, S.; Devlieger, R.; Van Assche, F.A. Fetal growth and developmental programming. J. Perinat. Med. 2013, 41, 101–105. [Google Scholar] [CrossRef]
  210. Gali Ramamoorthy, T.; Allen, T.J.; Davies, A.; Harno, E.; Sefton, C.; Murgatroyd, C.; White, A. Maternal overnutrition programs epigenetic changes in the regulatory regions of hypothalamic Pomc in the offspring of rats. Int. J. Obes. 2018, 42, 1431–1444. [Google Scholar] [CrossRef]
  211. Plagemann, A.; Harder, T.; Brunn, M.; Harder, A.; Roepke, K.; Wittrock-Staar, M.; Ziska, T.; Schellong, K.; Rodekamp, E.; Melchior, K.; et al. Hypothalamic proopiomelanocortin promoter methylation becomes altered by early overfeeding: An epigenetic model of obesity and the metabolic syndrome. J. Physiol. 2009, 587, 4963–4976. [Google Scholar] [CrossRef] [PubMed]
  212. Suter, M.A.; Chen, A.; Burdine, M.S.; Choudhury, M.; Harris, R.A.; Lane, R.H.; Friedman, J.E.; Grove, K.L.; Tackett, A.J.; Aagaard, K.M. A maternal high-fat diet modulates fetal SIRT1 histone and protein deacetylase activity in nonhuman primates. FASEB J. 2012, 26, 5106–5114. [Google Scholar] [CrossRef] [PubMed]
  213. Funato, H.; Oda, S.; Yokofujita, J.; Igarashi, H.; Kuroda, M. Fasting and high-fat diet alter histone deacetylase expression in the medial hypothalamus. PLoS ONE 2011, 6, e18950. [Google Scholar] [CrossRef] [PubMed]
  214. Dalvi, P.S.; Chalmers, J.A.; Luo, V.; Han, D.Y.; Wellhauser, L.; Liu, Y.; Tran, D.Q.; Castel, J.; Luquet, S.; Wheeler, M.B.; et al. High fat induces acute and chronic inflammation in the hypothalamus: Effect of high-fat diet, palmitate and TNF-α on appetite-regulating NPY neurons. Int. J. Obes. 2017, 41, 149–158. [Google Scholar] [CrossRef] [PubMed]
  215. Grayson, B.E.; Levasseur, P.R.; Williams, S.M.; Smith, M.S.; Marks, D.L.; Grove, K.L. Changes in melanocortin expression and inflammatory pathways in fetal offspring of nonhuman primates fed a high-fat diet. Endocrinology 2010, 151, 1622–1632. [Google Scholar] [CrossRef] [PubMed]
  216. Cheng, C.; Chen, W.; Jin, H.; Chen, X. A Review of Single-Cell RNA-Seq Annotation, Integration, and Cell-Cell Communication. Cells 2023, 12, 1970. [Google Scholar] [CrossRef] [PubMed]
  217. Chen, B.; de Launoit, E.; Renier, N.; Schneeberger, M. Maternal nutritional programming shapes the cerebral landscape. Trends Endocrinol. Metab. 2023, 35, 367–370. [Google Scholar] [CrossRef] [PubMed]
  218. Wu, R.; Prachyathipsakul, T.; Zhuang, J.; Liu, H.; Han, Y.; Liu, B.; Gong, S.; Qiu, J.; Wong, S.; Ribbe, A.; et al. Conferring liver selectivity to a thyromimetic using a novel nanoparticle increases therapeutic efficacy in a diet-induced obesity animal model. PNAS Nexus 2023, 2, pgad252. [Google Scholar] [CrossRef]
  219. Ayyangar, U.; Karkhanis, A.; Tay, H.; Afandi, A.F.B.; Bhattacharjee, O.; Ks, L.; Lee, S.H.; Chan, J.; Raghavan, S. Metabolic rewiring of macrophages by epidermal-derived lactate promotes sterile inflammation in the murine skin. EMBO J. 2024, 43, 1113–1134. [Google Scholar] [CrossRef]
Figure 1. Simplified overview of fetal immune cell development. Immune cells develop in three overlapping waves: firstly, primitive macrophages emerge from the yolk sac and, secondly, erythro-myeloid progenitors (EMP) form in the fetal liver. During the second and third trimesters, fully functioning hematopoietic stem and progenitor cells (HSPC) seed the fetal liver and migrate from the liver to the fetal bone marrow. The relative contribution of each wave is plotted against fetal gestational age in the number of weeks post-conception (wpc) until birth. T and B cells require further development through primary and secondary lymphoid organs starting from the common lymphoid progenitor (CLP) in the bone marrow (simplified here). Circulating leukocytes are replenished from this central reservoir in the bone marrow.
Figure 1. Simplified overview of fetal immune cell development. Immune cells develop in three overlapping waves: firstly, primitive macrophages emerge from the yolk sac and, secondly, erythro-myeloid progenitors (EMP) form in the fetal liver. During the second and third trimesters, fully functioning hematopoietic stem and progenitor cells (HSPC) seed the fetal liver and migrate from the liver to the fetal bone marrow. The relative contribution of each wave is plotted against fetal gestational age in the number of weeks post-conception (wpc) until birth. T and B cells require further development through primary and secondary lymphoid organs starting from the common lymphoid progenitor (CLP) in the bone marrow (simplified here). Circulating leukocytes are replenished from this central reservoir in the bone marrow.
Ijms 25 05951 g001
Figure 2. Changes to immune system effectors in the mother, fetus, and neonate due to maternal obesity. Maternal obesity is accompanied by increased inflammation and abnormal uterine natural killer (NK) cell activation. These changes alter the blood supply to the fetus by reducing fetal pluripotent CD34+ cells and regulatory capabilities and altering macrophage polarization. The neonate born to a mother with obesity may have imbalanced T helper subtypes, increased circulating IgE, and altered hematopoiesis leading to an increased chance of childhood allergies and autoimmune diseases. Bregs, B regulatory cells; NK, natural killer; NKT, NK T cells; pDCs, plasmacytoid dendritic cells; Th, T helper; Tregs, T regulatory cells.
Figure 2. Changes to immune system effectors in the mother, fetus, and neonate due to maternal obesity. Maternal obesity is accompanied by increased inflammation and abnormal uterine natural killer (NK) cell activation. These changes alter the blood supply to the fetus by reducing fetal pluripotent CD34+ cells and regulatory capabilities and altering macrophage polarization. The neonate born to a mother with obesity may have imbalanced T helper subtypes, increased circulating IgE, and altered hematopoiesis leading to an increased chance of childhood allergies and autoimmune diseases. Bregs, B regulatory cells; NK, natural killer; NKT, NK T cells; pDCs, plasmacytoid dendritic cells; Th, T helper; Tregs, T regulatory cells.
Ijms 25 05951 g002
Figure 3. Maternal factors underlie developmental programming and immune cell development. Several maternal factors brought on by a maternal WSD include increased immune activation, altered microbiome, and changes to bacterial metabolite composition. These exposures underlie dysregulated hematopoiesis, excessive immune tolerance, epigenetic modifications, and disordered infant microbiome. The outcomes affect postnatal developmental programming in organs and tissues with sustained and long-term metabolic and neurodevelopmental effects on future health.
Figure 3. Maternal factors underlie developmental programming and immune cell development. Several maternal factors brought on by a maternal WSD include increased immune activation, altered microbiome, and changes to bacterial metabolite composition. These exposures underlie dysregulated hematopoiesis, excessive immune tolerance, epigenetic modifications, and disordered infant microbiome. The outcomes affect postnatal developmental programming in organs and tissues with sustained and long-term metabolic and neurodevelopmental effects on future health.
Ijms 25 05951 g003
Table 1. Maternal obesity impacts commensal microbiota and immunity in the offspring.
Table 1. Maternal obesity impacts commensal microbiota and immunity in the offspring.
Subjects (Year)Physiological OutcomesProgramming EffectsReference
222 human infants and NOD mice (2016)
  • Bacteroides spp. are highly abundant in the at-risk human group
  • E. coli LPS decreases frequency of diabetes compared to B. dorei LPS in mice
  • Early microbiota trains innate immune signaling and tolerance
  • Different LPS subtypes can alter autoimmunity through immune system modulation
[137]
Germ-free mice colonized with microbes of infants from obese or NW mothers (2018)
  • Higher amounts of SCFAs and increased gut permeability
  • Macrophages were hyporesponsive to LPS and showed reduced phagocytic capability
  • Microbes were able to induce metabolic and inflammatory changes in the liver and bone marrow
  • Mice were susceptible to increased weight gain and fatty liver after a WD challenge
[130]
17,055 neonates from the general population (2019)
  • Increased risk for T1D was correlated with haplotypes DR3 and/or DR4
  • Intestinibacter and Romboutsia are associated with protective haplotypes
  • Risk for T1D is correlated with core microbiome and diversity
  • Probiotic treatment may be suitable due to conserved antigen among taxa present in high-risk individuals
[136]
Antibiotic-treated and germ-free mice (2019)
  • Myeloid progenitors reduced in antibiotic-treated mice
  • Commensals primed JAK signaling to promote TLR-enhanced myelopoiesis
  • Host microbiota primes TLR-enhanced myelopoiesis
  • Regulation of myelopoiesis required for sustained and proper immune responses
[139]
46 full-term neonates from obese mother with and without GDM (2020)
  • Microbes involved in immune suppression increased in GDM group
  • SCFA levels and microbial relative abundances were correlated
  • Innate immunity was reprogrammed by altered microbiota
  • Changed to gut microbiota can influence host metabolism
[140]
418 mothers and their neonates (2021)
  • Reduction in alpha diversity in neonates from women with GDM including increased Firmicutes and decreased Proteobacteria
  • Metabolites indicate increased taurine/hypo-taurine, pyrimidine, beta-alanine, and bile acid metabolism
  • An increase in opportunistic pathogens may cause childhood metabolic disorders
  • Pathways associated with increased metabolic function and fuels which may facilitate the development of childhood obesity
[141]
394 pregnant women in the first trimester and germ-free mice (2023)
  • Increased levels of IL-4, IL-6, IL-8, GM-CSF, and TNF-α and decreased amounts of SCFAs in women who later developed GDM
  • Decreased Prevotella in women who later developed GDM
  • GDM can be predicted as early as the first trimester
  • Markers for GDM diagnosis can include inflammation and microbiota
  • Microbial composition drove inflammation and insulin resistance
[142]
65 mother-infant pairs, half receiving a prebiotic (2024)
  • Prebiotics were associated with decreased Negativicutes in mothers and infants
  • Significant increases of the SCFA acetic acid were found in the prebiotic mother group
  • Negativicutes were correlated with c-section births and are less competitive in a prebiotic-rich nutritional environment
  • Acetate is likely due to increased abundance of Bifidobacterium
[143]
GDM, gestational diabetes mellitus; LPS, lipopolysaccharide; SCFA, short-chain fatty acid; T1D, type 1 diabetes.
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

Nelson, B.N.; Friedman, J.E. Developmental Programming of the Fetal Immune System by Maternal Western-Style Diet: Mechanisms and Implications for Disease Pathways in the Offspring. Int. J. Mol. Sci. 2024, 25, 5951. https://doi.org/10.3390/ijms25115951

AMA Style

Nelson BN, Friedman JE. Developmental Programming of the Fetal Immune System by Maternal Western-Style Diet: Mechanisms and Implications for Disease Pathways in the Offspring. International Journal of Molecular Sciences. 2024; 25(11):5951. https://doi.org/10.3390/ijms25115951

Chicago/Turabian Style

Nelson, Benjamin N., and Jacob E. Friedman. 2024. "Developmental Programming of the Fetal Immune System by Maternal Western-Style Diet: Mechanisms and Implications for Disease Pathways in the Offspring" International Journal of Molecular Sciences 25, no. 11: 5951. https://doi.org/10.3390/ijms25115951

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