Next Article in Journal
Sex- and Ethnic-Specific Associations of Serum Lipids with Risk of 12 Cancers: Findings from 506,381 Adults in Two Large Cohorts
Previous Article in Journal
Retinoprotective Effects of Abscisic Acid in Ischemic Retinopathy Mouse Model
Previous Article in Special Issue
Prenatal Melatonin Therapy Enhances Postnatal Lung Development in a Mouse Model of Inflammation-Induced Preterm Birth
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Does the Maternal Gut Microbiome Influence the Outcome of Perinatal Asphyxia?

by
Vlad-Petru Morozan
1,2,*,
Mara I. Ionescu
1,3,*,
Carmen M. D. Zahiu
1,
Ana Maria Catrina
4,
Andreea Racoviță
5,
Ana-Teodora Chirilă
1,
Ioana-Alexandra Dogaru
1,
Cristian Ciotei
1,
Gratiela Gradisteanu Pircalabioru
6,7,8 and
Ana-Maria Zăgrean
1
1
Division of Physiology—Neuroscience, Carol Davila University of Medicine and Pharmacy, 02001 Bucharest, Romania
2
Department of Plastic and Reconstructive Microsurgery, Central Military Emergency University Hospital ‘Dr. Carol Davila’, 010825 Bucharest, Romania
3
Department of Pediatrics II, Marie Curie Emergency Children’s Hospital, 75534 Bucharest, Romania
4
Cantacuzino National Military Medical Institute for Research and Development, 050096 Cernica, Romania
5
Department of Dermatology, Elias University Hospital, 011461 Bucharest, Romania
6
Research Institute of the University of Bucharest, 050663 Bucharest, Romania
7
eBio-Hub Centre of Excellence in Bioengineering, National University of Science and Technology Politehnica Bucharest, 061344 Bucharest, Romania
8
Department of Botany and Microbiology, University of Bucharest, 060101 Bucharest, Romania
*
Authors to whom correspondence should be addressed.
Antioxidants 2025, 14(9), 1134; https://doi.org/10.3390/antiox14091134
Submission received: 26 August 2025 / Revised: 13 September 2025 / Accepted: 16 September 2025 / Published: 19 September 2025
(This article belongs to the Special Issue Oxidative Stress in the Newborn)

Abstract

This review explores the maternal gut microbiome’s role in shaping neonatal neurodevelopmental outcomes following perinatal asphyxia (PA), a leading cause of infant mortality and disability with limited therapeutic options beyond hypothermia. We synthesized current evidence on microbiome-mediated neuroprotective mechanisms against hypoxic-ischemic brain injury. The maternal microbiome influences fetal development through bioactive metabolites (short-chain fatty acids, indole derivatives) that cross the placental barrier, bacterial antigen regulation, and infant microbiome colonization. These pathways activate multiple protective mechanisms: anti-inflammatory signaling via NF-κB suppression and regulatory T cell expansion; antioxidant defenses through Nrf2 activation; neural repair via BDNF upregulation and neurogenesis; and oxytocin system modulation. Animal models demonstrate that maternal dysbiosis from high-fat diet or antibiotics exacerbates PA-induced brain damage, increasing inflammatory markers and hippocampal injury. Conversely, probiotic supplementation, dietary fiber, and specific interventions (omega-3, resveratrol) reduce neuroinflammation and oxidative injury. Human studies link maternal dysbiosis-associated conditions (obesity, gestational diabetes) with adverse pregnancy outcomes, though direct clinical evidence for PA severity remains limited. Understanding the maternal microbiome-fetal brain axis opens therapeutic avenues, including prenatal probiotics, dietary modifications, and targeted metabolite supplementation to prevent or mitigate PA-related neurological sequelae, potentially complementing existing neuroprotective strategies.

1. Introduction

Perinatal Asphyxia (PA) is a medical condition in newborns caused by oxygen deprivation and hypercapnia that lasts sufficiently long to cause physical damage, particularly to the nervous system. By accounting for about half of neonatal encephalopathies in term and near-term infants, PA inflicts significant neurodevelopmental disability and ranks as the second-leading contributor to neonatal mortality [1,2,3].
Despite its high prevalence, therapeutic interventions for PA remain limited, with most offering only partial benefit [4]. While hypothermia remains the sole established treatment in clinical practice, its modest effectiveness (one out of seven treated infants avoids death or major disability) and narrow therapeutic window following birth [5,6] call for novel strategies to mitigate the consequences of hypoxic-ischemic events. Emerging evidence suggests that the maternal microbiome may be a novel and beneficial tool to address these therapeutic gaps [7,8,9].
The maternal microbiome influences foetal brain development, particularly through influencing inflammation and oxidative stress, two key drivers of hypoxic-ischemic injury in PA [7,9]. A balanced maternal microbiota creates a potent anti-inflammatory and antioxidative environment that regulates maternal immune activation and modulates microglial activity in the foetus, guiding adequate foetal neurodevelopment [10,11,12,13]. This protective milieu can buffer the developing brain against the detrimental effects of oxygen deprivation, while enhancing its neural repair capacity [10,11].
Conversely, maternal microbiota impairment, known as dysbiosis, has been linked to heightened pro-inflammatory states and disrupted metabolic signalling, exacerbating the severity of hypoxic-ischemic damage [14]. Given these findings, this narrative review aims to examine the mechanistic links and current evidence regarding the role of the maternal microbiota in PA outcomes.

2. Perinatal Asphyxia: Pathophysiology and Outcomes

To understand how the maternal microbiota might influence PA outcomes, it is essential to consider the pathophysiological cascade underlying this condition.
Classically, PA undergoes three distinct phases interweaved with a latent period [15]. The primary phase represents the injury itself, where low oxygen and high carbon dioxide levels trigger the chemo-reflexes, which further induce bradycardia and intense peripheral vasoconstriction to maintain perfusion of central organs [16,17]. Hypoxemia increases inflammation and oxidative stress, causes cell oedema and amplifies cell injury, increasing cellular death [18,19]. These neuronal lesions cause waves of anoxic depolarisation, which provide positive feedback in this loop and further aggravate the injuries [20,21].
Following moderate or severe hypoxic injury, a latent and apparently quiescent phase appears [22]. The higher the degree of injury, the shorter the latent period, after which the secondary phase commences with a restart of the inflammatory cascades and a continuation of the cytokine storm [5]. During this phase, trophic factors decrease, the amount of reactive oxygen species (ROS) and the mitochondrial membrane permeability increase, and intracellular calcium concentration rises, all creating a self-sustaining destructive landscape [21,23,24]. This landscape facilitates a pro-excitatory environment, which, combined with the increased glutamate resulting from neuronal loss, leads to massive depolarisation waves and seizure activity that further exacerbate neuronal loss [25,26,27].
Approximately three days after the hypoxic injury, the third phase begins [28]. Depending on the injury severity, it either tilts toward a reparatory phase, with the regeneration of damaged tissues, or, conversely, tilts toward continuing the inflammatory processes and aggravating the existing lesions [29,30,31,32].
PA produces different patterns of neurological injuries, depending on the severity and duration of oxygen deprivation, as well as the infant’s gestational age [5,33]. In preterm neonates, PA leads to a form of white matter injury called periventricular leukomalacia (PVL), which involves oligodendrocyte death and hindered myelination [34,35,36,37]. PVL presents with two distinct forms: cystic and diffuse [34,35]. In full-term neonates, acute sentinel PA damages the grey matter in the thalamus, basal ganglia, and brainstem, while repeated moderate hypoxic events lead to a “watershed” pattern, primarily affecting cortical regions [38,39]. However, depending on the context, neonates can also present multiple patterns simultaneously [5,38,40].
These lesions accompanying PA may lead to outcomes ranging from mild symptoms to neonatal hypoxic-ischemic encephalopathy (HIE), with severe hypoxic events potentially resulting in death [41]. Unfortunately, the neuronal damage often comes with dramatic long-term consequences: cerebral palsy, intellectual disability, psychiatric disorders, epilepsy, neurodevelopmental disorders (e.g., ADHD), learning disabilities, motor disabilities, etc., thus posing a great burden on the individual, as well as on society [40,41,42,43,44,45,46,47,48].
Although extensive work has focused on the pathophysiological cascades triggered by PA and the efficacy of therapeutic hypothermia and pharmacotherapy, important gaps remain in understanding why some newborns are more susceptible to brain damage than others [5,33]. The severity and persistence of PA outcomes underscore the need to investigate additional prenatal factors. In this regard, the maternal microbiome, long overlooked, has surfaced as a key player capable of influencing foetal health through immune regulation and metabolic pathways [7,9,49].

3. The Maternal Gut Microbiome: Composition, Functions, and Impact on Foetal Development

The maternal microbiota represents a complex consortium of microorganisms colonizing various maternal body sites, including the gastrointestinal tract, conjunctiva, airways, vagina, breast tissue, skin, and oral cavity [50]. Among these niches, the gut microbiota is the most extensively studied and undergoes significant shifts during pregnancy, driven by hormonal, metabolic, and immunological changes [51,52]. These adaptations promote enhanced energy storage and immune modulation [14,53].
In the gut, Firmicutes (split into several groups in the Genome Taxonomy Database) and Bacteroidetes (Bacteroidota) phyla comprise approximately 90% of the total bacterial population, with Actinobacteria (Actinomycetota), Proteobacteria (Pseudomonadota), Verrucomicrobia, and archaeal members such as Euryarchaeota accounting for the remaining 10% [51,54]. A balanced, eubiotic microbiota is characterized by diverse species, a beneficial ratio of key taxa, and vital functional capabilities, such as short-chain fatty acids (SCFAs) production, immune system education, maintenance of epithelial barrier integrity, vitamin and nutrient synthesis, and competitive exclusion of pathogens [55,56].
Despite this complexity, deviations from a balanced microbiome or dysbiosis can be categorized into three main forms: type 1, with the loss of beneficial microbes (e.g., Lactobacillus in the vaginal microbiome, Bifidobacterium in the gut); type 2, with an overgrowth of potentially harmful species (e.g., increased Proteobacteria during pregnancy) and type 3, with an overall reduction in microbial diversity, leading to compromised functional capacity and resilience [56].

3.1. Factors Influencing Microbiome Composition

Multiple factors influence the maternal microbiome during pregnancy, leading to unique microbial signatures that can impact both maternal and foetal health (Table 1). These influences create a dynamic, ever-shifting microbial landscape in the mother, which, in turn, shapes foetal development, systemic inflammation, the mother’s health, and the maturation of the foetal gut–brain axis [10,11,12,13,14,57,58,59,60,61,62,63].

3.2. Maternal Health Implications

Maternal gut microbiome shapes maternal health throughout pregnancy (Figure 1). Dysregulated maternal gut communities are associated with elevated proinflammatory cytokines, altered oestrogen metabolism, and impaired metabolic health [84,85]. Gestational dysbiosis is associated with conditions such as preeclampsia and hypertension [13,86,87]. Probiotic administration decreases blood glucose levels, lowers gestational diabetes markers, reduces insulin levels, and improves glucose tolerance; however, some studies argue for minimal or no probiotic effect [86,88].
Beyond maternal metabolic health, gut microbes can modulate the maternal immune system by influencing populations of granulocytes and regulatory T cells (Treg) [13]. Diets rich in fibre, known to improve the gut microbiota, are associated with lower proportions of proinflammatory immune cells. At the same time, dysbiosis increases M1 macrophages and proinflammatory cytokines (e.g., TNF-α, IL-1β, IL-6, IL-17), and reduces anti-inflammatory cytokines such as IL-10 [13]. Moreover, dysbiosis can lead to maternal immune activation, with important consequences for both maternal and foetal health [89]. Some studies conclude that dysbiosis increases the risk of preterm birth; however, the evidence remains conflicting [90,91,92,93].
Maternal microbiota influences placental development, and dysbiosis is accompanied by lower placental weight and volume, as well as impaired maternal-foetal circulation interfaces [94]. These conditions are attenuated by SCFAs administration [94] and certain bacterial species (e.g., Bifidobacterium breve) [95]. Although a wave of scientific discoveries pinpointed that the placenta might present a microbiome of its own, scientific consensus tilts towards no microbiome [73,94,95,96,97,98].

3.3. Direct Effects on Foetal Development

Aside from maternal health, pregnancy well-being, and placental development, the maternal gut microbiota influences the development of foetal immune and nervous systems, and the infant’s future metabolic health (Figure 2) [86,88].

3.3.1. Immune System Maturation

From in utero to early childhood, the immune system undergoes a set of fine-tuned, sequential developmental processes [99] that evolve in parallel with the maternal gut microbiota, engaging in a bidirectional interplay [100] that shapes the immune cell populations. Dysbiosis can alter normal immune system development and can shift the balance either towards more immune reactivity with increased chances to develop autoimmune diseases or atopy, or towards a less reactive immune system, incapable of defending itself against threats [13,100,101,102,103,104,105,106,107]. For example, a maternal high-fat diet, known to induce dysbiosis, increases the expression of receptors that compromise the intestinal mucus barrier (e.g., LRRC19) [108], leading to increased bacterial translocation and heightened inflammatory state. Also, it upregulates proinflammatory RORγt-positive cells (precursors of IL-17-producing cells) [58,109] and elevates the levels of TNF-α and IL-6 [110]. In contrast, probiotics and prebiotic administration push the immune balance toward a more tolerogenic state, marked by an increase in regulatory T cells (Tregs) and in IL-10 and TGF-β levels [111,112].
The mechanisms underlying immune system maturation involve four primary pathways: (1) microbial metabolites—SCFAs [100,107,113,114,115,116], aryl hydrocarbon receptor (AhR) ligands [10], bile salts [117], peptidoglycan fragments [118], trimethylated microbial metabolites [10], (2) IgG-bound microbial antigens [119], (3) microbiota derived extracellular vesicles [120] and (4) bacterial translocation [58].
Immune system maturation, although an autonomous process, is closely intertwined with neurodevelopment [121,122], each influencing the other’s course [123].

3.3.2. Neurodevelopmental Processes

Maternal microbial communities and their metabolites impact the fetal neural circuits’ development, such as thalamocortical axons [57]. Germ-free animal models revealed altered gene expression related to myelination and axon development, highlighting the pivotal role of microbiota in neurodevelopment [10]. In addition, differentiation of GABAergic neurons appears to depend on AhR signaling [124], which is activated by microbial metabolites in the foetus.
Recently, the role of the maternal gut-fetal brain axis in fetal neurodevelopment has been increasingly considered [14]. Dysbiotic conditions lead to maternal immune activation, which has been linked to autism-like phenotypes in offspring, via IL-17-producing Th17 cells [63,89,125,126,127]. In addition, specific bacterial taxa (e.g., Fusobacteria) in the maternal gut have also been associated with better fine motor outcomes in children [128] and enhanced neurogenesis [129].
Maternal diet, which is known to impact microbiome, can either exacerbate [130] or mitigate [131] long-term neurodevelopmental deficits. Antibiotic use during gestation, known to induce dysbiosis, alters hippocampal development and emotional regulation in offspring [9,70], suggesting that the maternal microbiome profoundly impacts fetal brain health and development.

4. Neuroprotective Mechanisms Mediated by the Maternal Microbiome

The response of the immature brain to metabolic deprivation conditions, such as hypoxia/ischemia, involves mechanisms still to be revealed, which could explain why some newborns walk away from the same hypoxic insult while others live with lifelong disability [132,133,134,135]. The maternal microbiome could be an important factor influencing this response: dysbiosis magnifies cytokine-driven injury [136], while eubiosis, by promoting anti-inflammatory mechanisms, redox buffering, and reparatory signals (Figure 3), blunts the secondary wave of neuronal loss that could determine whether recovery or lifelong disability prevails [137].

4.1. Anti-Inflammatory Mechanisms

Within hours following a hypoxic-ischemic insult, a secondary cytokine storm unleashes, with increased concentrations of IL-1β, TNF-α, and IL-6. These cytokines amplify the initial lesion size, and their levels hold predictive power for outcome forecast [138,139,140,141].
Maternal gut metabolites, mainly SCFAs and indole derivatives, can dampen that cytokine surge at several, hierarchically layered checkpoints: (i) epigenetic silencing of NF-κB inside microglia [142], (ii) tightening of gut-placenta-blood/cerebrospinal fluid barriers that stop endotoxin leakage [143,144], and (iii) prenatal expansion of Tregs that pre-set a tolerogenic baseline [145].
SCFAs inhibit the histone deacetylase (HDAC), which reduces the expression of inflammatory cytokines [146] and prevents NF-κB translocation, both directly, as well as through HDAC inhibition [147], thus steering microglia towards an Arg-1/IL-10 rich M2 anti-inflammatory phenotype [142,148]. In a middle cerebral artery occlusion stroke model in adult mice, butyrate, a key SCFA, almost halved infarcted volume when administered alone [149]. Moreover, in a model of neuronal damage due to cardiac arrest, butyrate attenuated the microglial inflammation through the NF-κB pathway [150] and reduced brain inflammation through activation of the JAK-STAT pathway [151]. Studies both in the adult and neonate found that the microglia respond similarly to SCFAs, by reducing NF-κB translocation, thus providing an anti-inflammatory and protective response to hypoxia [152].
SCFAs bind G-protein-coupled receptors (GPCRs) in brain endothelial and intestinal cells, providing a second layer of protection. GPCR activation increases the expression of claudin 5 and occludin in both cell types, thus tightening the barriers and limiting endotoxin accumulation in the brain [143,153]. Furthermore, SCFAs induce actin cytoskeleton rearrangements in a blood-brain barrier (BBB) in vitro model, contributing to improved BBB integrity [144]. A structurally sound BBB promotes a lower microglial activation, thus limiting the initial lesions and inflammation [154,155].
In parallel, SCFAs boost Treg cells’ expansion [113,156] in the thymic and placental tissues, which lowers the fetal IL-6/IL-1β basal levels and increases the anti-inflammatory cytokine IL-10 production. SCFAs may exert these effects by directly inhibiting HDACs in naïve T cells, activating free fatty acid receptors (FFARs) on both dendritic cells and naïve T cells, and priming thymic epithelial cells to select Treg cells [113,156,157].
Gut-derived indole derivatives cross the placenta and reach fetal brain tissues [158]. In animal models of ischemia, they affect microglia through AhR activation, which suppresses p65 phosphorylation and decreases IL-1β/TNF-α expression through the NF-κB pathway [159,160,161]. As shown in a stroke model, reduction of NF-κB translocation provides an anti-inflammatory and protective response to hypoxia in both adult and neonate mouse brains [152]. Experimental evidence shows that AhR-ligands interact with microglia in the fetal brain and reduce brain inflammation [162].
Epigenetic silencing of NF-κB, GPCR-driven BBB tightening, systemic Treg expansion, and AhR activation form together a multilayered anti-inflammatory buffer that shields the newborn brain against PA damage.

4.2. Antioxidant Pathways: Maternal Gut Barrier, Placenta, Blood-Brain-Barrier, Glial Cells, and Neurons

The hypoxic injury begins with a primary burst of ROS and a larger oxidative aftershock, which result in mitochondrial failure and excitotoxicity, which together form a self-amplifying destructive loop [18]. Beyond classical apoptosis and necrosis, ferroptosis, an iron-dependent, lipid-peroxidation-driven form of regulated cell death, has emerged as an additional contributor in neonatal HIE [163,164]. Moreover, experimental data link Nrf2 activation to lower ferroptosis and improved outcomes in PA [163,165,166].
The maternal gut microbiome shapes antioxidant defences in both the mother and foetus, playing a key role in antioxidant capacity transfer to the foetus [167,168]. In mice, the maternal microbiome drives distinct metabolomics profiles in the placenta, foetal gut, and foetal brain, confirming transplacental transfer of microbial metabolites [158].
This antioxidant “defence line” spans several interconnected compartments along the maternal-foetal axis: maternal gut, placenta, epithelial barriers (BBB and placento-fetal), neurons, and glial cells.
Within the maternal gut, eubiosis sustains intestinal barrier function and neutralises ROS, preventing leakage of pro-oxidant and inflammatory molecules into the circulation [169]. A well-colonized maternal gut contributes to antioxidant protection in two complementary ways: first, gut bacteria neutralise reactive oxygen species with antioxidant enzymes—NADH peroxidase and SOD—as well as with metabolites with ROS-scavenging activity [170,171]; second, microbial metabolites increase antioxidant defence through Nrf2 signalling and upregulate tight-junction proteins [169,170,171]. SCFAs bind FFAR, inhibit HDAC, and activate AMPK, which increases Nrf2 nuclear translocation and induces antioxidant defences [170]. AhR ligands increase Nrf2 nuclear translocation and induce tight junction proteins, as well as increased enzymatic antioxidant defences [169].
In the placenta, maternal antioxidant reserves and microbiota-derived metabolites help neutralize reactive oxygen species before they reach the foetus, supporting glutathione balance and redox homeostasis [172,173], thus protecting the placenta and the foetus [174]. AhR ligands induce Nrf2 translocation, which enhances enzymatic antioxidant systems and provides antioxidant buffers themselves through interaction with IDO (indoleamine 2,3-dioxygenase) [172]. Direct placental Nrf2 activation by SCFAs in vivo remains to be demonstrated; although current evidence supports SCFA transfer and sensing, with anti-inflammatory effects through NF-κB that lower oxidative stress at the placenta [94,174].
In the foetal BBB, an increased permeability allows for more ROS increased oxidative damage in PA [175]. The microbiome regulates maturation of the BBB, controlling the expression of tight junction proteins: occludins and claudins [176]. Because SCFAs reach foetal circulation, FFAR activation and Nrf2 signalling could enhance antioxidant capacity and maintain BBB integrity [154,177].
In neurons, SCFAs protect against oxidative ischemic damage in animal models [178,179]. SCFAs inhibit neuronal HDAC [180], increase Nrf2 nuclear translocation, enhance antioxidant enzyme activity, and lower ferroptosis [181]. Additionally, they act through the BDNF-Akt3 pathway to reduce oxidative stress and apoptosis [178]. AhR ligands could exhibit direct antioxidant effects. Thus, tryptophan metabolites related to gut microbiota, which are endogenous AhR ligands, attenuated neuronal damage in an ischemic hippocampus model [182].
Glial cells, including microglia and astrocytes, further contribute by fine-tuning inflammatory responses and limiting oxidative injury, roles that have been demonstrated in ischemia and related injury models [159,165]. Mechanistically, this could involve NF-κB inhibition, which triggers M2 phenotype with lower ROS production, both through SCFA and AhR ligands [150,159], although direct evidence in PA is currently lacking.
Together, these layers form a coordinated antioxidant network that may critically influence vulnerability or resilience to oxidative injury in PA.

4.3. Neurogenesis, Neuroplasticity, and Neural Repair

The maternal microbiota modulates neurotrophic factor expression through pathways that lead to increased antioxidant enzymes. Short-chain fatty acids, particularly butyrate, enhance brain-derived neurotrophic factor (BDNF) expression by direct histone deacetylase inhibition and increased H3 crotonylation, which together amplify antioxidant gene transcription [183,184,185]. This SCFA-mediated BDNF upregulation activates the PI3K/AKT signaling cascade, which phosphorylates and inactivates pro-apoptotic proteins BDNF and increases antioxidant expression [186], as shown in a model of ischemia reperfusion injury [178]. In contrast, maternal high-fat diet-induced dysbiosis correlates with reduced offspring BDNF levels, along with functional impairments [187,188,189,190].
By promoting a strong anti-inflammatory phenotype in microglia, adequate synaptic pruning and development occur [191], leading to a greater neuronal reserve and improved wiring.
In dysbiotic animal models (induced through high-fat diet), butyrate improves mitochondrial function in the cerebral cortex and the synaptic fraction [192], enhances neuroplasticity [193], and shows an impressive neuroprotective effect in ischemia [142,194], all increasing the neural functional reserve. One possible mechanism underlying this protective effect could be the ability of sodium butyrate to stimulate neurogenesis, as demonstrated in rat models of perinatal asphyxia [142,195].
In addition, the maternal gut microbiome affects oligodendrocyte activation and myelination processes in the offspring’s central nervous system [196]. Through HDAC inhibition in precursor oligodendrocytes, SCFAs guide their maturation and proper myelination, creating additional resistance against PA [196]. Moreover, SCFAs reduce brain inflammation, lowering the risk of precursor oligodendrocyte disruption.
Maternal eubiosis likely primes the foetus towards a reparative phenotype during phase 3, providing direct neuroprotection through enhanced neurogenesis and neural repair, thus improving neuronal reserve and mitigating the PA effects.

4.4. The Maternal Microbiome, Oxytocin Signaling, and Perinatal Asphyxia—Direct Neuroprotection?

The maternal microbiome’s neuroprotective effects extend beyond direct metabolite production to include modulation of critical neuropeptide systems, particularly oxytocin (OXT) [197]. We believe this neuroendocrine pathway represents an underexplored mechanism through which the maternal gut bacteria influence fetal resilience to hypoxic injury.
Maternal gut dysbiosis, particularly from high-fat diets, depletes beneficial species like Lactobacillus reuteri, correlating with diminished hypothalamic OXT expression in offspring and impaired stress regulation [197]. Crucially, L. reuteri supplementation during pregnancy enhances maternal OXT signaling via the vagus nerve stimulation [198,199], with strain-specific effects not replicated by other Lactobacillus species [200].
The microbiome-OXT connection is particularly relevant for PA outcomes, as OXT facilitates the critical transition from excitatory to inhibitory GABA signaling in the fetal brain, a key protective mechanism against hypoxia-induced excitotoxicity [201]. In experimental PA models, OXT administration reduces neuronal loss, seizure severity, and hippocampal damage, with efficacy dependent on injury severity [202,203,204,205].
The neuroprotective capacity is enhanced by OXT’s co-release with arginine vasopressin during perinatal stress, which suppresses excessive neuronal excitability during oxygen deprivation [206,207,208]. Further, germ-free and antibiotic-exposed [209] animal models display altered OXT signaling and social deficits reminiscent of neurodevelopmental disorders, whereas offspring treatment with L. reuteri reversed maternal high-fat diet-induced social and synaptic deficits [197], suggesting that early microbial colonization is essential for normal oxytocinergic development.
Therefore, maternal dysbiosis may compromise this endogenous neuroprotective system, increase HIE severity, and disrupt OXT signalling. Microbiome-based interventions that target OXT could enhance fetal neuroprotection against PA.

5. The Maternal Gut Microbiome Dysbiosis and Perinatal Asphyxia Outcomes

While maternal eubiosis provides multiple layers of neuroprotection against hypoxic-ischemic injury, dysbiosis creates a contrasting scenario of increased vulnerability. Disrupted maternal microbial communities compromise the very pathways that normally shield the developing brain from oxidative damage, inflammatory cascades, and metabolic dysfunction.
This section examines the experimental evidence demonstrating how the maternal microbiome imbalance amplifies the pathophysiological processes underlying PA, transforming what might be a manageable hypoxic insult into severe, lasting neurological injury.

5.1. Mechanisms of Dysbiosis-Induced Neuronal Vulnerability

PA creates a storm of oxidative vulnerability that, amplified by maternal (and subsequent fetal) dysbiosis, amplifies PA’s neurological injury. This section examines how dysbiosis-derived endotoxins amplify the oxidative damage cascade specific to hypoxic-ischemic events.
As dysbiosis results in a higher lipopolysaccharide (LPS) concentration in the bloodstream [155], LPS can amplify the oxidative and proinflammatory stress [210,211], with increased ROS production, TNF-α level, and antioxidant enzymes, as shown in a model of PA, combined with LPS exposure. LPS action seems to also depend on timing [211], while even hypothermia could bring minimal benefit against a LPS sensitised neural tissue [212].
Mitochondrial dysfunction from PA is another factor of ROS and triggers multiple forms of cellular death, including ferroptosis [18]. Dysbiosis reduces bacterial metabolites that promote a tolerogenic and anti-inflammatory status, and increase oxidative stress and pro-oxidative and inflammatory cytokines such as IL-6, IL-1β, and TNF-α [181,213], thus amplifying the effect of PA.
By allowing more LPS into the bloodstream, dysbiosis affects BBB integrity [176,214,215,216], leading to enhanced neuroinflammation and, ultimately, more severe outcomes.
Lastly, by increasing pro-inflammatory cytokines and decreasing beneficial bacterial metabolites, dysbiosis transforms microglia into a pro-inflammatory phenotype with increased ROS production and neuronal damage [217,218].

5.2. Experimental Findings

Detrimental microbiome alterations consistently exacerbate PA-induced brain damage (Table 2). Empirical findings reveal that a high-fat gestational diet linked to gut dysbiosis heightens neuroinflammation and cellular injury in the hippocampus of rat pups subjected to PA, as shown by increased TNF-α, IL-1β, and S100B [219]. Similarly, antibiotic-induced maternal dysbiosis amplifies PA vulnerability and exacerbates brain injuries, with altered neurodevelopmental reflexes and increased hippocampal S100B [9], while neonatal antibiotic exposure reduces SCFA-producing bacteria and promotes astrocytic gliosis and microglial activation during perinatal ischemia [7].
Conversely, interventions that display neuroprotective effects have been shown to positively affect microbiome homeostasis (Table 2). Nutritional supplements, including trans-resveratrol [220], citicoline [221], and omega-3 PUFAs [222], not only provide direct neuroprotection through mechanisms such as SIRT1 activation and membrane stabilization but also promote beneficial bacteria that may protect against PA [223,224,225].
Table 2. Experimental Evidence Linking Microbiome Modulation to Perinatal Asphyxia (PA) Outcomes.
Table 2. Experimental Evidence Linking Microbiome Modulation to Perinatal Asphyxia (PA) Outcomes.
InterventionModelPrimary ActionMicrobiome EffectPerinatal Asphyxia (PA) OutcomesKey MarkersReferences
High-fat dietSprague-Dawley (SD) rats, PA P7Metabolic dysregulation↓ Bacteroidetes, ↑ Firmicutes/Bacteroidetes ratio↑ Hippocampal injury↑ TNF-α, IL-1β, S100B[219]
Gestational antibioticsSD rats, PA birthMicrobiome depletion #↓ Diversity, ↓ Lactobacillus/Bifidobacterium↑ Brain injury, altered reflexes↑ S100B[9]
Neonatal antibioticsMice, PAMicrobiome disruption #↓ short-chain fatty acid (SCFA) producers↑ Neuronal damage, gliosis↑ GFAP, Iba1[7]
Omega-3 PUFAMice, HI P9Anti-inflammatory↑ Butyrate producers 1*↓ Injury volume (5 weeks)↓ NF-κB, apoptosis[222]
ResveratrolSD rats, PA P7SIRT1 activationLactobacillus and Bifidobacterium 2*↓ Hippocampal damage↓ IL-1β, TNF-α[220]
CiticolineSD rats, PA P7Membrane stabilizationMaintains homeostasis 3*Preserved hippocampus↓ Inflammation[221]
LactoferrinRats, PA↑ Nrf2, ↓ ferroptosisBifidobacterium and Lactobacillus 4*↓ Neuronal death↓ Ferroptosis[226,227]
SCFAsSD rats, PA P7Metabolic dysregulationDirect metabolite↓ 30% infarct, ↑ neurogenesis↓ IL-1β, COX-2[194]
Probiotics SD rats, PA birthMicrobiome depletion #Lactobacillus and Bifidobacterium, ↑ SCFAsBlood-brain barrier (BBB) protection↓ Microglial activation[8]
↑ increased/higher levels; ↓ decreased/lower levels; # Primary mechanism is microbiome-related; * Microbiome effects documented separately from PA studies: 1* [225]; 2* [223]; 3* [224]; 4* [227]; L. acidophilus + B. infantis; IL-1β inflammatory model.
Lactoferrin supplementation reduces PA-induced neuronal death through Nrf2-mediated ferroptosis inhibition [226], while also promoting beneficial bacteria and inhibiting harmful ones [227].
Direct microbiome-targeted interventions provide compelling evidence for causality. SCFA administration after neonatal hypoxia reduces brain infarct volume by 30%, decreases IL-1β and COX-2 expression, and enhances neurogenesis through histone crotonylation and HDAC inhibition [142,185,194,195].
Maternal probiotic supplementation with Lactobacillus acidophilus and Bifidobacterium infantis protects blood-brain barrier integrity and reduces microglial activation even in inflammatory challenge models [8], suggesting that microbiome optimization creates resilience against multiple injury pathways.
These experimental findings demonstrate that the maternal microbiome represents a modifiable factor influencing offspring vulnerability to hypoxic-ischemic injury, with interventions showing efficacy through both direct neuroprotective mechanisms and indirect microbiome-mediated pathways.

5.3. Vertical Transmission and Neonatal Gut-Brain Axis Disruption

Neonates born to mothers with gut microbiome dysbiosis may vertically inherit an altered microbial community [50], potentially predisposing them to an impaired gut-brain axis function and heightened susceptibility to PA-induced neurological injury [136]. While infant gut microbiome colonization timing remains debated, most evidence indicates postnatal establishment [97], with the maternal gut microbiome serving as the primary inoculum source [50]. Multiple factors shape this vertical transmission, including maternal health status, delivery mode, breastfeeding practices, and maternal care habits [49,228,229].
This inherited dysbiosis creates a foundation of vulnerability that compounds PA pathophysiology. Because of the infant gut’s rich vascularization and the underdeveloped fetal circulatory system, gastrointestinal tissues show increased vulnerability to PA [230,231]. Animal models of global hypoxemia or mesenteric artery occlusion report upregulation of IL-1 and IL-17, gut barrier disruption, and increased bacterial translocation, reflected by higher blood LPS levels [49,232,233].
Furthermore, hypoxemic conditions consistently alter intestinal microbiome composition, with murine models showing increased Firmicutes and decreased Bacteroides and Proteobacteria populations [234,235]. This demonstrates that hypoxemia directly modifies gut microbiome composition, establishing a self-reinforcing cycle where inherited dysbiosis increases PA susceptibility while PA-induced hypoxemia further destabilizes the gut microbiome [49,236].

6. Therapeutic Interventions Targeting the Maternal Microbiome

Given the profound impact of the maternal gut microbiome on fetal neurodevelopment and resilience to hypoxic injury, therapeutic strategies targeting maternal microbial communities represent a promising avenue for preventing or mitigating perinatal asphyxia outcomes. These interventions range from simple dietary modifications to sophisticated microbiome-engineering approaches, each offering distinct mechanisms for enhancing neuroprotection through modulation of oxidative stress and inflammatory pathways.

6.1. Dietary Modifications

Maternal diet shapes its microbiome, and it also shapes the infant gut microbiome [237,238]. Despite appearing relatively straightforward, dietary interventions demonstrate significant therapeutic potential.
Dietary fibre serves as the primary substrate for SCFA production, with demonstrated anti-inflammatory effects [239,240]. High-fibre diets during pregnancy increase SCFA-producing bacteria, enhancing SCFAs levels [67,241]. Among the most studied, the Mediterranean diet, rich in vegetables, fibres, and antioxidants, seems to provide a more anti-inflammatory microbiome [242,243], with effects reverberating into early childhood, hence protection against PA [237]. A randomized control trial analysing the impact of the Mediterranean diet showed better neurodevelopment outcomes at the 2-year mark (Bayley-III) [244].
Alongside fibres, another important aspect of dietary modifications could be related to the amount of polyphenols found in the Mediterranean diet. While having an impact on the gut microbiome and on the inflammatory reverberations of the gut microbiome [224,245], polyphenols serve as antioxidants and have been found to reduce the impact of brain ischemic damage in animal models [220,246,247,248,249]. Polyphenols enhance the development of SCFAs-producing bacteria, especially Akkermansia and Bifidobacterium, and inhibit potentially harmful bacteria growth [250,251,252]. Dietary polyphenols are metabolized mainly in the colon into bioactive components, such as equol, urolithin, and protocatechuic acid, by several bacterial species (Bifidobacterium sp., Lactobacillus sp., Bacteroides sp., etc.) [251,253]. Through the interaction with the maternal microbiome, resveratrol, transformed into 4-hydroxyphenylacetic acid, acts through the SIRT1 pathway and prevents obesity [254].
Omega-3 fatty acid supplementation deserves special emphasis, with maternal DHA-enriched diets preventing neuronal apoptosis and reducing brain injury volume in HIE models through anti-inflammatory effects and microglial NF-κB signalling suppression [222,255], an effect that was present at 5 weeks postnatal. Alongside direct effects in animal studies, in humans, omega-3 has been associated with fewer pre-term births and better neonatal health [256]. Omega-3 fatty acids also influence maternal microbiome and the inflammatory status of the placenta and the offspring, respectively [225]. These supplements maintain the intestinal wall integrity and modulate host immune response through modulating the gut composition, promoting butyrate-producing bacteria and Bacteroidetes sp. [257].
Fermented foods such as kombucha, yogurt, and kimchi show promise for establishing a balanced maternal microbiome [258]. Furthermore, these foods have been reported to offer benefits for pregnancy overall [259]. These effects likely stem from their complex composition; while serving as a vehicle for probiotics, these foods also supply essential macronutrients (carbohydrates, proteins, fats), alongside other bioactive compounds such as prebiotics and microbial metabolites [259,260]. A review on fermented foods shows the benefits on immune system maturation, brain development and maternal health, and infant microbiota [261].
In addition, maternal administration of lactoferrin improved outcomes in PA [226] through Nrf2 signalling, increasing antioxidant buffer and reducing ferroptosis. In addition, lactoferrin modulates gut diversity and promotes beneficial bacterial growth (e.g., Bifidobacterium and Lactobacillus) [227].
Conversely, a Western-style high-fat/low-fibre dietary pattern promotes dysbiosis, elevates maternal and placental cytokine levels, and predisposes offspring to neuroinflammation [187].

6.2. Emerging Microbiome-Based Therapeutic Strategies

Alongside dietary and lifestyle interventions, targeted manipulation of the maternal gut microbiome through prebiotics, probiotics, postbiotics, and synbiotics represents an expanding area of research with significant implications for maternal–infant health and neurodevelopment. These interventions are distinct yet complementary, each influencing microbial composition and function via different mechanisms.

6.2.1. Prebiotics: Enhancing Endogenous Microbial Metabolism

Prebiotics are defined as selectively fermentable, non-digestible food components, most notably galacto-oligosaccharides (GOS) and fructo-oligosaccharides (FOS), that stimulate the growth and activity of beneficial gut microorganisms. During pregnancy, prebiotic supplementation alters microbial fermentation patterns, leading to increased production of SCFAs such as acetate, propionate, and butyrate. These metabolites cross the placental barrier and exert systemic effects, enhancing antioxidant defense and modulating immune tolerance in the offspring [112,262]. Notably, prebiotics induce a tolerogenic immune shift by promoting regulatory B and T cells expansion and elevating acetate and butyrate concentrations, metabolites with potent anti-inflammatory functions [112,263].
Such effects may enhance fetal resilience to hypoxic-ischemic insults, although direct evidence in the context of PA is currently lacking.

6.2.2. Probiotics: Augmenting Beneficial Microbial Populations

Probiotics, live microorganisms that confer health benefits when administered in adequate amounts, provide a more direct strategy to enhance maternal gut composition. Supplementation during gestation with Lactobacillus and Bifidobacterium species lowers proinflammatory cytokines, preserves the BBB, and enhances oligodendrocyte maturation following neonatal challenges [8]. Mechanistically, probiotics increase antioxidant capacity, improve immune response, suppress inflammatory cascades, and produce antimicrobial peptides that inhibit pathogenic bacteria [264,265,266,267].
Strain-specific effects have also been observed. For example, Clostridium butyricum enhances SCFAs production and mitigates ischemia/reperfusion injury in rodent models [179], while Lactobacillus reuteri modulates OXT signalling, increasing hypothalamic oxytocin expression and improving GABA-mediated neuroprotection under hypoxic stress [199,266]. In addition, probiotic interventions have reduced neuronal damage in experimental models of perinatal inflammation [8], and are clinically employed in conditions such as necrotizing enterocolitis (NEC), a pathology that can follow PA [268].
In maternal populations, probiotics have been used to reduce complications, including gestational diabetes, preeclampsia, and infection-related outcomes [111], factors that may indirectly influence PA severity if unmanaged. Nevertheless, while preclinical findings are promising, human data remain limited and inconsistent, highlighting the need for large-scale trials [269].

6.2.3. Postbiotics: Delivering Microbial Metabolites Directly

Postbiotics are non-viable bacterial components or metabolites, such as SCFAs, that offer an alternative route to harness microbial benefits without live organisms. Supplementation with SCFAs has demonstrated safety and efficacy in attenuating hypoxic-ischemic brain lesions, improving BBB function, reducing oxidative stress, and suppressing inflammation in animal studies [137,154,194]. Additional evidence suggests that postbiotics may ameliorate placental dysfunction and immune dysregulation associated with dysbiosis [12,94].

6.2.4. Synbiotics and Other Advanced Strategies

Synbiotics, defined as combinations of probiotics and prebiotics, represent an evolving therapeutic concept. By supplying beneficial microorganisms alongside their preferred substrates, synbiotics may synergistically enhance colonization and functional outputs. While PA-specific studies are absent, synbiotics have improved clinical outcomes in NEC and reduced allergy incidence in infants [270]. These findings suggest potential neuroprotective benefits under hypoxic stress conditions. Maternal synbiotic supplementation during gestation and lactation significantly enhances antioxidant capacity, upregulates mitochondrial function–related genes, and increases the abundance of beneficial intestinal microbiota in offspring, suggesting a synergistic benefit over probiotics alone [271].
Faecal microbiota transplantation (FMT), although still experimental in pregnancy and neonates, is another emerging modality. By transferring processed stool from healthy donors, FMT restores microbial diversity and metabolic function. Evidence supports its efficacy in metabolic syndrome and inflammatory conditions across animal and human studies [272,273,274], but its safety and utility in PA-related contexts require rigorous evaluation.

6.2.5. Addressing Harmful Metabolites and Novel Pharmacological Targets

Not all microbial metabolites are beneficial. Compounds such as trimethylamine (TMA) and trimethylamine N-oxide (TMAO) are implicated in adverse cardiovascular programming and offspring hypertension [275,276]. Inhibitors like 3,3-dimethyl-1-butanol (DMB) suppress TMA/TMAO formation, mitigating these risks [276,277].
Furthermore, pharmacological targeting of the microbial-sensing pathways shows promise. FFARs agonists, such as 4-CMBT, or AhR ligands like indole-3-propionic acid (IPA), have demonstrated anti-inflammatory and neuroprotective effects in preclinical settings [159,278,279]. Such strategies, though untested in PA, may complement microbiome-directed therapies to enhance foetal resilience to hypoxia.
In summary, microbiome-based interventions, including prebiotics, probiotics, postbiotics, synbiotics, and emerging pharmacological approaches, offer exciting opportunities to improve maternal and neonatal outcomes. While animal models provide strong mechanistic evidence, clinical translation remains at an early stage. Addressing safety, timing, and strain-specific effects will be essential to realize the full potential of these therapies in preventing or mitigating the PA-related neurodevelopmental injury.

7. Current Challenges and Future Research Directions

Understanding the maternal microbiome’s influence on PA remains a developing area of investigation. While animal models have provided initial insights into the maternal microbiome–foetal brain axis, human research remains limited. This is compounded by methodological variability, ethical concerns, and the absence of clear clinical guidelines for microbiome-based interventions during pregnancy or neonatal care. Bridging these gaps is crucial for translating preclinical findings into effective and safe perinatal neuroprotective strategies.

7.1. Methodological and Scientific Gaps

Most studies to date have focused on the maternal gut microbiome, excluding other potentially influential microbial niches such as the vaginal, oral, skin, and placental microbiota. These ecosystems may play important and interactive roles in foetal immune and neurodevelopment, particularly in modulating the neonatal response to hypoxic stress. The exclusion of these sites from current studies limits our understanding of the broader maternal microbial landscape and its role in perinatal outcomes.
In addition, this review employed a narrative approach, which, while suitable for conceptual exploration, lacks the methodological rigor of systematic reviews and may not capture the full spectrum of available evidence. The majority of research investigating maternal gut microbiome influences on offspring development originates from rodent models, which enable microbiome manipulation through antibiotic depletion, gnotobiotic designs, and maternal immune activation [9,57,125]. Large animal models have validated the impact of maternal gut microbiome on infant development, both in cattle and swine [280], as well as in non-human primates [281,282,283]. Since perinatal asphyxia models are well-established in rodents, large animals, and primates, these species offer valuable platforms for investigating maternal microbiome influences on hypoxic-ischemic outcomes [142,284,285,286]. However, few studies have directly examined the interplay between maternal gut microbiome manipulation and neonatal hypoxic-ischemic injury, representing a critical research gap that these established models are well-positioned to address.
There is also a lack of longitudinal human studies examining maternal microbiome dynamics across pregnancy and their association with neonatal outcomes following PA. Without such studies, the ability to draw causal links or develop predictive microbial biomarkers remains limited.

7.2. Ethical and Safety Concerns

The use of microbiome-modulating therapies during pregnancy and in neonates introduces several ethical and safety concerns. Although many probiotics contain strains that are naturally present in the human gut or used in fermented foods, such as Lactobacillus, Bifidobacterium, Streptococcus, Saccharomyces, and Bacillus, their safety in vulnerable populations remains incompletely understood [287].
Evidence suggests that maternal probiotic use is relatively safe. A systematic review by Jarde et al. found no increased risk of miscarriage, foetal malformation, premature birth, low birth weight, or Caesarean section associated with probiotic administration during pregnancy. Therefore, targeting the maternal microbiome may represent a safer alternative to direct postnatal probiotic supplementation, particularly in preterm or at-risk neonates [90].
In contrast, neonatal probiotic use is more controversial. Case reports have documented severe adverse outcomes, including bacteraemia, fungemia, and endocarditis, in immunocompromised or preterm infants treated with probiotics [288,289,290,291], while others consider the direct administration rather beneficial [292]. In 2023, the U.S. Food and Drug Administration (FDA) issued a formal warning following reports of infections and deaths in preterm infants who received probiotics to prevent necrotizing enterocolitis [293]. These incidents underscore the need for stringent safety protocols and regulatory oversight when considering microbiota-based interventions in neonatology.
Beyond individual-level risks, public health concerns also emerge. O’Doherty et al. argue that microbial interventions may inadvertently affect others through microbial shedding or environmental transmission. For example, altering the microbiome of a pregnant individual may influence the microbial exposure of her infant, household members, or even broader community contacts. These findings highlight the importance of ethical scrutiny and the development of responsible policies around microbiome modulation, especially during critical developmental windows [294].
Taken together, these findings reinforce the need for internationally accepted standards and clinical guidelines governing the design, testing, and application of microbiome-modulating therapies in perinatal medicine.

7.3. Clinical Evidence and Limitations

While there are many therapeutic microbiome-targeted options, few have been studied in the context of PA and even fewer possess a straight from the bench to bedside pathway that would allow human trials soon.
Several limitations hinder clinical translation, including mechanistic uncertainty regarding how microbiome modulation leads to improved neurodevelopmental outcomes in PA, population-specific variability in maternal microbiome composition, the absence of defined timing and dosing windows for intervention during pregnancy or postpartum, and substantial regulatory and safety hurdles in ensuring maternal–foetal safety for novel therapeutics.
Furthermore, animal models, while invaluable for mechanistic insights, do not fully replicate human perinatal physiology and immune–microbiome interactions, which complicates extrapolation to clinical practice. As a result, while the therapeutic potential of microbiome-based interventions in PA is promising, progress toward clinical application will require rigorous, multi-centre studies employing standardized protocols, validated biomarkers, and long-term neurodevelopmental follow-up.

7.4. Future Research Recommendations

Future research should expand beyond the maternal gut microbiome to explore the contribution of other maternal microbial niches. The vaginal and placental microbiota are likely to play central roles in foetal immune education and inflammatory priming. Their influence on susceptibility to perinatal hypoxia-related brain injury remains underexplored and deserves targeted investigation.
Mechanistic studies must also evolve. Integrating high-throughput multi-omics approaches, such as metagenomics, metabolomics, transcriptomics, and immune profiling, with histological and imaging assessments of the foetal brain will allow a better understanding of how maternal microbiome signals are transduced across the placental barrier. The placenta itself is emerging as a critical intermediary in this signalling network and should be prioritized in future experimental designs.
Human clinical research must shift toward longitudinal, standardized protocols. Prospective cohort studies tracking microbiome profiles across pregnancy and associating them with neurodevelopmental outcomes in infants exposed to PA are essential. These studies should incorporate both structural imaging and behavioural assessments to evaluate the functional significance of microbial influences on neurodevelopment.
Randomized controlled trials are urgently needed to evaluate the safety and efficacy of probiotics and related interventions during pregnancy. Given current safety concerns surrounding neonatal probiotic use, maternal-targeted strategies may offer a safer route to modulate the infant microbiome indirectly. However, rigorous adverse event monitoring and stratification by maternal and infant risk factors will be crucial in any interventional study.
Finally, incorporating microbiome-based screening into routine prenatal care may enhance risk stratification for perinatal brain injury. Identifying microbial signatures predictive of adverse outcomes could support early intervention and inform the development of personalized therapeutic strategies.
In summary, the maternal microbiome represents a promising yet underutilized target for influencing the outcome of perinatal hypoxia/asphyxia. At this moment, no intervention that addresses the microbiome is currently in practice for mitigating PA effects [295]. Addressing current methodological, ethical, and clinical limitations through multidisciplinary research is essential to realize the translational potential of microbiome-based interventions in perinatal neuroprotection.

8. Conclusions

This narrative review shows the critical role of the maternal gut microbiome in shaping fetal immune and neurodevelopmental trajectories, with growing evidence linking its composition to neonatal vulnerability or resilience in the context of perinatal hypoxia/asphyxia. Maternal gut eubiosis may enhance brain resilience to perinatal asphyxia through potent antioxidant and anti-inflammatory stimuli that reverberate across the gut-placenta-brain axis, and through promotion of neurogenesis, neuroplasticity, and oxytocinergic pathways. Conversely, dysbiosis may exacerbate hypoxic-ischemic brain injury by priming a hyperreactive immune system, increasing oxidative stress, and negatively influencing neurodevelopmental trajectories.
The therapeutic landscape offers promising opportunities through dietary modifications, prebiotic, probiotic, and postbiotic therapeutics. However, significant challenges remain, including methodological limitations in current research, ethical considerations surrounding maternal interventions, optimal timing strategies, and the critical need for thorough safety data in pregnancy populations.
While animal studies support these associations, human data remain limited. Further research is needed to establish causality, identify key microbial mediators, and evaluate the safety and efficacy of microbiome-targeted interventions during pregnancy and the perinatal period. Bridging mechanistic insights from animal models to longitudinal human studies will be essential to translate these findings into safe, effective interventions in obstetric and neonatal practice. Therefore, harnessing the maternal microbiome as a therapeutic target could offer a promising avenue to improve perinatal neuroprotection and long-term neurodevelopmental outcomes. Ultimately, integrating maternal microbiome modulation into perinatal care—alongside established approaches such as therapeutic hypothermia—may provide a transformative strategy to reduce the burden of hypoxic-ischemic brain injury.

Author Contributions

Conceptualization, V.-P.M. and M.I.I.; methodology, V.-P.M.; validation, V.-P.M., M.I.I., C.M.D.Z., A.M.C., A.-T.C., I.-A.D., C.C., G.G.P. and A.-M.Z.; formal analysis, V.-P.M.; investigation, V.-P.M.; resources, V.-P.M. and A.-M.Z.; data curation, V.-P.M. and M.I.I.; writing and original draft preparation, V.-P.M., M.I.I., C.M.D.Z., A.M.C., A.R., A.-T.C., I.-A.D., C.C., G.G.P. and A.-M.Z.; writing, review and editing, all authors; visualization, A.R.; supervision, G.G.P. and A.-M.Z.; project administration, V.-P.M. All authors have read and agreed to the published version of the manuscript.

Funding

The APC was funded by Carol Davila University of Medicine and Pharmacy.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors express gratitude to Mihai Moldovan (Department of Neuroscience, University of Copenhagen; Department of Clinical Neurophysiology, Rigshospitalet, Copenhagen, Denmark; Department of Physiology—Neuroscience “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania) for valuable discussions regarding the structural organization and conceptual framework of this manuscript. Figures in this manuscript were created with BioRender.com. The authors used ChatGPT (OpenAI GPT4o, o1-pro and o3-pro) and Claude (Anthropic Claude Sonnet 4.0, Opus 4.0 & Opus 4.1) to assist with text editing (to improve grammar, prose clarity, and stylistic consistency) and manuscript organization (brainstorming ideas for table of contents and better flow for complex sections). AI tools were not used for literature review, data interpretation, scientific reasoning, or generating conclusions. All outputs were reviewed and revised by the authors, who take full responsibility for the scientific accuracy of the final content.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
4-CMBT4-Chloro-α-(1-methylethyl)-N-2-thiazolyl-benzeneacetamide
AhRAryl hydrocarbon receptor
AMPK5′ adenosine monophosphate-activated protein kinase
BBBBlood-brain barrier
BDNFBrain-derived neurotrophic factor
BEVsBacterial extracellular vesicles
BMIBody mass index
COX-2Cyclooxygenase-2
CSFCerebrospinal fluid
DHADocosahexaenoic acid
DMB3,3-dimethyl-1-butanol
F/BFirmicutes/Bacteroidetes ratio
FDAU.S. Food and Drug Administration
FFARFree fatty acid receptor
FFAR2Free fatty acid receptor 2
FFAR3Free fatty acid receptor 3
FMTFaecal microbiota transplantation
FOSFructo-oligosaccharides
GABAGamma-aminobutyric acid
GFAPGlial fibrillary acidic protein
GOSGalacto-oligosaccharides
GPCRsG protein-coupled receptors
H3Histone 3
HDACHistone deacetylase
HIHypoxia-ischaemia
HIEHypoxic-ischaemic encephalopathy
Iba1Ionised calcium-binding adapter molecule 1
IBDInflammatory bowel disease
IDOIndoleamine 2,3-dioxygenase
IgGImmunoglobulin G
IL-1βInterleukin-1 beta
IL-6Interleukin-6
IL-10Interleukin-10
IL-17Interleukin-17
IPAIndole-3-propionic acid
JAK-STATJanus kinase/Signal Transducer and Activator of Transcription
LPSLipopolysaccharide
LRRC19Leucine Rich Repeat Containing 19
MCAOMiddle cerebral artery occlusion
MGMMaternal gut microbiota
MIAMaternal immune activation
NADHNicotinamide adenine dinucleotide
NECNecrotising enterocolitis
NF-κBNuclear factor kappa B
Nrf2Nuclear factor erythroid 2-related factor 2
OXTOxytocin
P7/P9Postnatal day 7/9
PAPerinatal asphyxia
PI3K/AKTPhosphoinositide 3-kinase/Protein kinase B
PKCProtein kinase C
PUFAPolyunsaturated fatty acids
PVLPeriventricular leukomalacia
ROSReactive oxygen species
S100BS100 calcium-binding protein B
SCFAsShort-chain fatty acids
SDSprague-Dawley
SIRT1Sirtuin 1
SODSuperoxide dismutase
TECsThymic epithelial cells
TGF-βTransforming growth factor-beta
TMATrimethylamine
TMAOTrimethylamine N-oxide
TMGTrimethylglycine
TNF-αTumour necrosis factor-alpha
TregsRegulatory T cells

References

  1. Liu, L.; Oza, S.; Hogan, D.; Chu, Y.; Perin, J.; Zhu, J.; Lawn, J.E.; Cousens, S.; Mathers, C.; Black, R.E. Global, Regional, and National Causes of under-5 Mortality in 2000–15: An Updated Systematic Analysis with Implications for the Sustainable Development Goals. Lancet 2016, 388, 3027–3035, Erratum in Lancet 2017, 389, 1884. [Google Scholar] [CrossRef]
  2. Kurinczuk, J.J.; White-Koning, M.; Badawi, N. Epidemiology of Neonatal Encephalopathy and Hypoxic-Ischaemic Encephalopathy. Early Hum. Dev. 2010, 86, 329–338. [Google Scholar] [CrossRef]
  3. Oza, S.; Lawn, J.E.; Hogan, D.R.; Mathers, C.; Cousens, S.N. Neonatal Cause-of-Death Estimates for the Early and Late Neonatal Periods for 194 Countries: 2000–2013. Bull. World Health Organ. 2015, 93, 19–28. [Google Scholar] [CrossRef]
  4. Cannavò, L.; Perrone, S.; Gitto, E. Brain-Oriented Strategies for Neuroprotection of Asphyxiated Newborns in the First Hours of Life. Pediatr. Neurol. 2023, 143, 44–49. [Google Scholar] [CrossRef]
  5. Lear, B.A.; Zhou, K.Q.; Dhillon, S.K.; Lear, C.A.; Bennet, L.; Gunn, A.J. Preventive, Rescue and Reparative Neuroprotective Strategies for the Fetus and Neonate. Semin. Fetal Neonatal Med. 2024, 29, 101542. [Google Scholar] [CrossRef]
  6. Wassink, G.; Davidson, J.O.; Dhillon, S.K.; Zhou, K.; Bennet, L.; Thoresen, M.; Gunn, A.J. Therapeutic Hypothermia in Neonatal Hypoxic-Ischemic Encephalopathy. Curr. Neurol. Neurosci. Rep. 2019, 19, 2. [Google Scholar] [CrossRef] [PubMed]
  7. Drobyshevsky, A.; Synowiec, S.; Goussakov, I.; Fabres, R.; Lu, J.; Caplan, M. Intestinal Microbiota Modulates Neuroinflammatory Response and Brain Injury after Neonatal Hypoxia-Ischemia. Gut Microbes 2024, 16, 2333808. [Google Scholar] [CrossRef] [PubMed]
  8. Lu, J.; Lu, L.; Yu, Y.; Baranowski, J.; Claud, E.C. Maternal Administration of Probiotics Promotes Brain Development and Protects Offspring’s Brain from Postnatal Inflammatory Insults in C57/BL6J Mice. Sci. Rep. 2020, 10, 8178. [Google Scholar] [CrossRef] [PubMed]
  9. Ionescu, M.I.; Maria Catrina, A.; Dogaru, I.A.; Catalina Barbalata, D.; Ciotei, C.; Haidoiu, C.; Suhaianu, V.; Gradisteanu Pircalabioru, G.; O’Mahony, S.M.; Zagrean, A.-M. MICROBIOME: The Trials and Errors of Developing an Experimental Model to Study the Impact of Maternal Gut Microbiome Disruption on Perinatal Asphyxia. Reprod. Fertil. 2024, 5, e240050. [Google Scholar] [CrossRef]
  10. Husso, A.; Pessa-Morikawa, T.; Koistinen, V.M.; Kärkkäinen, O.; Kwon, H.N.; Lahti, L.; Iivanainen, A.; Hanhineva, K.; Niku, M. Impacts of Maternal Microbiota and Microbial Metabolites on Fetal Intestine, Brain, and Placenta. BMC Biol. 2023, 21, 207. [Google Scholar] [CrossRef]
  11. Abdel-Haq, R.; Schlachetzki, J.C.M.; Glass, C.K.; Mazmanian, S.K. Microbiome-Microglia Connections via the Gut-Brain Axis. J. Exp. Med. 2019, 216, 41–59. [Google Scholar] [CrossRef] [PubMed]
  12. Qin, X.; Zhang, M.; Chen, S.; Tang, Y.; Cui, J.; Ding, G. Short-Chain Fatty Acids in Fetal Development and Metabolism. Trends Mol. Med. 2024, 31, 625–639. [Google Scholar] [CrossRef]
  13. Lu, X.; Shi, Z.; Jiang, L.; Zhang, S. Maternal Gut Microbiota in the Health of Mothers and Offspring: From the Perspective of Immunology. Front. Immunol. 2024, 15, 1362784. [Google Scholar] [CrossRef]
  14. Sajdel-Sulkowska, E.M. The Impact of Maternal Gut Microbiota during Pregnancy on Fetal Gut-Brain Axis Development and Life-Long Health Outcomes. Microorganisms 2023, 11, 2199. [Google Scholar] [CrossRef]
  15. Bennet, L. Sex, Drugs and Rock and Roll: Tales from Preterm Fetal Life. J. Physiol. 2017, 595, 1865–1881. [Google Scholar] [CrossRef]
  16. Giussani, D.A. The Fetal Brain Sparing Response to Hypoxia: Physiological Mechanisms: Fetal Brain Sparing. J. Physiol. 2016, 594, 1215–1230. [Google Scholar] [CrossRef] [PubMed]
  17. Jensen, A.; Berger, R. Fetal Circulatory Responses to Oxygen Lack. J. Dev. Physiol. 1991, 16, 181–207. [Google Scholar]
  18. Okazaki, K.; Nakamura, S.; Koyano, K.; Konishi, Y.; Kondo, M.; Kusaka, T. Neonatal Asphyxia as an Inflammatory Disease: Reactive Oxygen Species and Cytokines. Front. Pediatr. 2023, 11, 1070743. [Google Scholar] [CrossRef]
  19. Szydlowska, K.; Tymianski, M. Calcium, Ischemia and Excitotoxicity. Cell Calcium 2010, 47, 122–129. [Google Scholar] [CrossRef]
  20. Hartings, J.A.; Shuttleworth, C.W.; Kirov, S.A.; Ayata, C.; Hinzman, J.M.; Foreman, B.; Andrew, R.D.; Boutelle, M.G.; Brennan, K.C.; Carlson, A.P.; et al. The Continuum of Spreading Depolarizations in Acute Cortical Lesion Development: Examining Leão’s Legacy. J. Cereb. Blood Flow Metab. 2017, 37, 1571–1594. [Google Scholar] [CrossRef] [PubMed]
  21. Wassink, G.; Gunn, E.R.; Drury, P.P.; Bennet, L.; Gunn, A.J. The Mechanisms and Treatment of Asphyxial Encephalopathy. Front. Neurosci. 2014, 8, 40. [Google Scholar] [CrossRef]
  22. Iwata, O.; Iwata, S.; Thornton, J.S.; De Vita, E.; Bainbridge, A.; Herbert, L.; Scaravilli, F.; Peebles, D.; Wyatt, J.S.; Cady, E.B.; et al. “Therapeutic Time Window” Duration Decreases with Increasing Severity of Cerebral Hypoxia-Ischaemia under Normothermia and Delayed Hypothermia in Newborn Piglets. Brain Res. 2007, 1154, 173–180. [Google Scholar] [PubMed]
  23. Hostetter, M.K. Society for Pediatric Research Presidential Address 1994: Yeast as Metaphor. Pediatr. Res. 1994, 36, 692–698. [Google Scholar] [CrossRef]
  24. Thornton, C.; Leaw, B.; Mallard, C.; Nair, S.; Jinnai, M.; Hagberg, H. Cell Death in the Developing Brain after Hypoxia-Ischemia. Front. Cell. Neurosci. 2017, 11, 248. [Google Scholar] [CrossRef]
  25. Rodriguez, J.; Xie, C.; Li, T. Inhibiting the Interaction between Apoptosisinducing Factor and Cyclophilin A Prevents Brain Injury in Neonatal Mice after Hypoxia-Ischemia. Neuropharmacology 2020, 171, 108088. [Google Scholar] [CrossRef]
  26. Shaw, J.C.; Crombie, G.K.; Zakar, T.; Palliser, H.K.; Hirst, J.J. Perinatal Compromise Contributes to Programming of GABAergic and Glutamatergic Systems Leading to Long-Term Effects on Offspring Behaviour. J. Neuroendocrinol. 2020, 32, e12814. [Google Scholar] [CrossRef]
  27. Shaw, J.C.; Berry, M.J.; Dyson, R.M.; Crombie, G.K.; Hirst, J.J.; Palliser, H.K. Reduced Neurosteroid Exposure Following Preterm Birth and Its’ Contribution to Neurological Impairment: A Novel Avenue for Preventative Therapies. Front. Physiol. 2019, 10, 599. [Google Scholar] [CrossRef]
  28. Fleiss, B.; Gressens, P. Tertiary Mechanisms of Brain Damage: A New Hope for Treatment of Cerebral Palsy? Lancet Neurol. 2012, 11, 556–566. [Google Scholar] [CrossRef]
  29. Bennet, L.; Dhillon, S.; Lear, C.A.; van den Heuij, L.; King, V.; Dean, J.M.; Wassink, G.; Davidson, J.O.; Gunn, A.J. Chronic Inflammation and Impaired Development of the Preterm Brain. J. Reprod. Immunol. 2018, 125, 45–55. [Google Scholar] [CrossRef] [PubMed]
  30. Dhillon, S.K.; Gunn, E.R.; Lear, B.A.; King, V.J.; Lear, C.A.; Wassink, G.; Davidson, J.O.; Bennet, L.; Gunn, A.J. Cerebral Oxygenation and Metabolism after Hypoxia-Ischemia. Front. Pediatr. 2022, 10, 925951. [Google Scholar] [CrossRef] [PubMed]
  31. Buser, J.R.; Maire, J.; Riddle, A.; Gong, X.; Nguyen, T.; Nelson, K.; Luo, N.L.; Ren, J.; Struve, J.; Sherman, L.S.; et al. Arrested Preoligodendrocyte Maturation Contributes to Myelination Failure in Premature Infants. Ann. Neurol. 2012, 71, 93–109. [Google Scholar] [CrossRef]
  32. Dean, J.M.; Mcclendon, E.; Hansen, K.; Azimi-Zonooz, A.; Chen, K.; Riddle, A.; Gong, X.; Sharifnia, E.; Hagen, M.; Ahmad, T.; et al. Prenatal Cerebral Ischemia Disrupts MRIdefined Cortical Microstructure through Disturbances in Neuronal Arborization. Sci. Transl. Med. 2013, 5, 168–175. [Google Scholar] [CrossRef] [PubMed]
  33. Mota-Rojas, D.; Villanueva-García, D.; Solimano, A.; Muns, R.; Ibarra-Ríos, D.; Mota-Reyes, A. Pathophysiology of Perinatal Asphyxia in Humans and Animal Models. Biomedicines 2022, 10, 347. [Google Scholar] [CrossRef] [PubMed]
  34. Hinojosa-Rodríguez, M.; Harmony, T.; Carrillo-Prado, C.; Van Horn, J.D.; Irimia, A.; Torgerson, C.; Jacokes, Z. Clinical Neuroimaging in the Preterm Infant: Diagnosis and Prognosis. NeuroImage Clin. 2017, 16, 355–368. [Google Scholar] [CrossRef]
  35. Banker, B.Q.; Larroche, J.C. Periventricular Leukomalacia of Infancy. A Form of Neonatal Anoxic Encephalopathy. Arch. Neurol. 1962, 7, 386–410. [Google Scholar] [CrossRef]
  36. Schneider, J.; Miller, S.P. Preterm Brain Injury: White Matter Injury. Handb. Clin. Neurol. 2019, 162, 155–172. [Google Scholar]
  37. Volpe, J.J.; Kinney, H.C.; Jensen, F.E.; Rosenberg, P.A. The Developing Oligodendrocyte: Key Cellular Target in Brain Injury in the Premature Infant. Int. J. Dev. Neurosci. 2011, 29, 423–440. [Google Scholar] [CrossRef] [PubMed]
  38. Nikas, I.; Dermentzoglou, V.; Theofanopoulou, M.; Theodoropoulos, V. Parasagittal Lesions and Ulegyria in Hypoxic-Ischemic Encephalopathy: Neuroimaging Findings and Review of the Pathogenesis. J. Child Neurol. 2008, 23, 51–58. [Google Scholar] [CrossRef]
  39. Rivkin, M.J. Hypoxic-Ischemic Brain Injury in the Term Newborn. Neuropathology, Clinical Aspects, and Neuroimaging. Clin. Perinatol. 1997, 24, 607–625. [Google Scholar] [CrossRef]
  40. Groenendaal, F.; de Vries, L.S. Fifty Years of Brain Imaging in Neonatal Encephalopathy Following Perinatal Asphyxia. Pediatr. Res. 2017, 81, 150–155. [Google Scholar] [CrossRef]
  41. Shah, P.; Anvekar, A.; McMichael, J.; Rao, S. Outcomes of Infants with Apgar Score of Zero at 10 Min: The West Australian Experience. Arch. Dis. Child. Fetal Neonatal Ed. 2015, 100, F492–F494. [Google Scholar] [CrossRef]
  42. Milner, K.M.; Neal, E.F.G.; Roberts, G.; Steer, A.C.; Duke, T. Long-Term Neurodevelopmental Outcome in High-Risk Newborns in Resource-Limited Settings: A Systematic Review of the Literature. Paediatr. Int. Child Health 2015, 35, 227–242. [Google Scholar] [CrossRef]
  43. Omizzolo, C.; Scratch, S.E.; Stargatt, R.; Kidokoro, H.; Thompson, D.K.; Lee, K.J.; Cheong, J.; Neil, J.; Inder, T.E.; Doyle, L.W.; et al. Neonatal Brain Abnormalities and Memory and Learning Outcomes at 7 Years in Children Born Very Preterm. Memory 2014, 22, 605–615. [Google Scholar] [CrossRef]
  44. Stipdonk, L.W.; Franken, M.-C.J.P.; Dudink, J. Language Outcome Related to Brain Structures in School-Aged Preterm Children: A Systematic Review. PLoS ONE 2018, 13, e0196607, Erratum in PLoS ONE 2018, 13, e0203298. [Google Scholar]
  45. Wheelock, M.D.; Austin, N.C.; Bora, S.; Eggebrecht, A.T.; Melzer, T.R.; Woodward, L.J.; Smyser, C.D. Altered Functional Network Connectivity Relates to Motor Development in Children Born Very Preterm. NeuroImage 2018, 183, 574–583. [Google Scholar] [CrossRef]
  46. Lee, B.L.; Gano, D.; Rogers, E.E.; Xu, D.; Cox, S.; James Barkovich, A.; Li, Y.; Ferriero, D.M.; Glass, H.C. Long-Term Cognitive Outcomes in Term Newborns with Watershed Injury Caused by Neonatal Encephalopathy. Pediatr. Res. 2022, 92, 505–512. [Google Scholar] [CrossRef]
  47. Ahearne, C.E.; Boylan, G.B.; Murray, D.M. Short and Long Term Prognosis in Perinatal Asphyxia: An Update. World J. Clin. Pediatr. 2016, 5, 67–74. [Google Scholar] [CrossRef] [PubMed]
  48. de Haan, M.; Wyatt, J.S.; Roth, S.; Vargha-Khadem, F.; Gadian, D.; Mishkin, M. Brain and Cognitive-Behavioural Development after Asphyxia at Term Birth. Dev. Sci. 2006, 9, 350–358. [Google Scholar] [CrossRef]
  49. Matara, D.-I.; Pouliakis, A.; Xanthos, T.; Sokou, R.; Kafalidis, G.; Iliodromiti, Z.; Boutsikou, T.; Iacovidou, N.; Salakos, C. Microbial Translocation and Perinatal Asphyxia/Hypoxia: A Systematic Review. Diagnostics 2022, 12, 214. [Google Scholar] [CrossRef] [PubMed]
  50. Ferretti, P.; Pasolli, E.; Tett, A.; Asnicar, F.; Gorfer, V.; Fedi, S.; Armanini, F.; Truong, D.T.; Manara, S.; Zolfo, M.; et al. Mother-to-Infant Microbial Transmission from Different Body Sites Shapes the Developing Infant Gut Microbiome. Cell Host Microbe 2018, 24, 133–145.e5. [Google Scholar] [CrossRef] [PubMed]
  51. Yang, H.; Guo, R.; Li, S.; Liang, F.; Tian, C.; Zhao, X.; Long, Y.; Liu, F.; Jiang, M.; Zhang, Y.; et al. Systematic Analysis of Gut Microbiota in Pregnant Women and Its Correlations with Individual Heterogeneity. NPJ Biofilms Microbiomes 2020, 6, 32. [Google Scholar] [CrossRef] [PubMed]
  52. Smid, M.C.; Ricks, N.M.; Panzer, A.; Mccoy, A.N.; Azcarate-Peril, M.A.; Keku, T.O.; Boggess, K.A. Maternal Gut Microbiome Biodiversity in Pregnancy. Am. J. Perinatol. 2018, 35, 24–30. [Google Scholar]
  53. Koren, O.; Goodrich, J.K.; Cullender, T.C.; Spor, A.; Laitinen, K.; Bäckhed, H.K.; Gonzalez, A.; Werner, J.J.; Angenent, L.T.; Knight, R.; et al. Host Remodeling of the Gut Microbiome and Metabolic Changes during Pregnancy. Cell 2012, 150, 470–480. [Google Scholar] [CrossRef]
  54. Parks, D.H.; Chuvochina, M.; Chaumeil, P.-A.; Rinke, C.; Mussig, A.J.; Hugenholtz, P. A Complete Domain-to-Species Taxonomy for Bacteria and Archaea. Nat. Biotechnol. 2020, 38, 1079–1086, Erratum in Nat. Biotechnol. 2020, 38, 1098. [Google Scholar] [CrossRef]
  55. Buttó, L.F.; Haller, D. Dysbiosis in Intestinal Inflammation: Cause or Consequence. Int. J. Med. Microbiol. 2016, 306, 302–309. [Google Scholar] [CrossRef]
  56. DeGruttola, A.K.; Low, D.; Mizoguchi, A.; Mizoguchi, E. Current Understanding of Dysbiosis in Disease in Human and Animal Models. Inflamm. Bowel Dis. 2016, 22, 1137–1150. [Google Scholar] [CrossRef]
  57. Vuong, H.E.; Pronovost, G.N.; Williams, D.W.; Coley, E.J.L.; Siegler, E.L.; Qiu, A.; Kazantsev, M.; Wilson, C.J.; Rendon, T.; Hsiao, E.Y. The Maternal Microbiome Modulates Fetal Neurodevelopment in Mice. Nature 2020, 586, 281–286. [Google Scholar] [CrossRef]
  58. Ruiz-Triviño, J.; Álvarez, D.; Cadavid J, Á.P.; Alvarez, A.M. From Gut to Placenta: Understanding How the Maternal Microbiome Models Life-Long Conditions. Front. Endocrinol. 2023, 14, 1304727. [Google Scholar] [CrossRef] [PubMed]
  59. Faienza, M.F.; Urbano, F.; Anaclerio, F.; Moscogiuri, L.A.; Konstantinidou, F.; Stuppia, L.; Gatta, V. Exploring Maternal Diet-Epigenetic-Gut Microbiome Crosstalk as an Intervention Strategy to Counter Early Obesity Programming. Curr. Issues Mol. Biol. 2024, 46, 4358–4378. [Google Scholar] [CrossRef] [PubMed]
  60. Ziętek, M.; Celewicz, Z.; Szczuko, M. Short-Chain Fatty Acids, Maternal Microbiota and Metabolism in Pregnancy. Nutrients 2021, 13, 1244. [Google Scholar] [CrossRef]
  61. Wu, J.; Wang, K.; Wang, X.; Pang, Y.; Jiang, C. The Role of the Gut Microbiome and Its Metabolites in Metabolic Diseases. Protein Cell 2021, 12, 360–373. [Google Scholar] [CrossRef]
  62. Beckers, K.F.; Sones, J.L. Maternal Microbiome and the Hypertensive Disorder of Pregnancy, Preeclampsia. Am. J. Physiol. Heart Circ. Physiol. 2020, 318, H1–H10. [Google Scholar] [CrossRef] [PubMed]
  63. Deady, C.; FitzGerard, J.; Kara, N.; Mazzocchi, M.; O’Mahony, A.; Ionescu, M.I.; Shanahan, R.; Kelly, S.; Crispie, F.; Cotter, P.D.; et al. Maternal Immune Activation and Antibiotics Affect Offspring Neurodevelopment, Behaviour, and Microbiome. Brain Behav. Immun. Health 2025, 48, 101065. [Google Scholar] [CrossRef]
  64. Mann, P.E.; Huynh, K.; Widmer, G. Maternal High Fat Diet and Its Consequence on the Gut Microbiome: A Rat Model. Gut Microbes 2018, 9, 143–154. [Google Scholar] [CrossRef] [PubMed]
  65. Gohir, W.; Whelan, F.J.; Surette, M.G.; Moore, C.; Schertzer, J.D.; Sloboda, D.M. Pregnancy-Related Changes in the Maternal Gut Microbiota Are Dependent upon the Mother’s Periconceptional Diet. Gut Microbes 2015, 6, 310–320. [Google Scholar] [CrossRef]
  66. Grant, E.T.; Boudaud, M.; Muller, A.; Macpherson, A.J.; Desai, M.S. Maternal Diet and Gut Microbiome Composition Modulate Early-Life Immune Development. EMBO Mol. Med. 2023, 15, e17241. [Google Scholar] [CrossRef]
  67. Barrientos, G.; Ronchi, F.; Conrad, M.L. Nutrition during Pregnancy: Influence on the Gut Microbiome and Fetal Development. Am. J. Reprod. Immunol. 2024, 91, e13802. [Google Scholar] [CrossRef]
  68. García-Mantrana, I.; Selma-Royo, M.; González, S.; Parra-Llorca, A.; Martínez-Costa, C.; Collado, M.C. Distinct Maternal Microbiota Clusters Are Associated with Diet during Pregnancy: Impact on Neonatal Microbiota and Infant Growth during the First 18 Months of Life. Gut Microbes 2020, 11, 962–978. [Google Scholar] [CrossRef]
  69. Wang, Y.; Xie, T.; Wu, Y.; Liu, Y.; Zou, Z.; Bai, J. Impacts of Maternal Diet and Alcohol Consumption during Pregnancy on Maternal and Infant Gut Microbiota. Biomolecules 2021, 11, 369. [Google Scholar] [CrossRef]
  70. Degroote, S.; Hunting, D.J.; Baccarelli, A.A.; Takser, L. Maternal Gut and Fetal Brain Connection: Increased Anxiety and Reduced Social Interactions in Wistar Rat Offspring Following Peri-Conceptional Antibiotic Exposure. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2016, 71, 76–82. [Google Scholar] [CrossRef] [PubMed]
  71. Chen, Y.; Li, Z.; Tye, K.D.; Luo, H.; Tang, X.; Liao, Y.; Wang, D.; Zhou, J.; Yang, P.; Li, Y.; et al. Probiotic Supplementation during Human Pregnancy Affects the Gut Microbiota and Immune Status. Front. Cell. Infect. Microbiol. 2019, 9, 254. [Google Scholar] [CrossRef] [PubMed]
  72. Dekker Nitert, M.; Gomez-Arango, L.F.; Barrett, H.L.; McIntyre, H.D.; Anderson, G.J.; Frazer, D.M.; Callaway, L.K. Iron Supplementation Has Minor Effects on Gut Microbiota Composition in Overweight and Obese Women in Early Pregnancy. Br. J. Nutr. 2018, 120, 283–289. [Google Scholar] [CrossRef] [PubMed]
  73. Nuriel-Ohayon, M.; Neuman, H.; Koren, O. Microbial Changes during Pregnancy, Birth, and Infancy. Front. Microbiol. 2016, 7, 1031. [Google Scholar] [CrossRef] [PubMed]
  74. Li, M.; Zhang, G.; Cui, L.; Zhang, L.; Zhou, Q.; Mu, C.; Chi, R.; Zhang, N.; Ma, G. Dynamic Changes in Gut Microbiota during Pregnancy among Chinese Women and Influencing Factors: A Prospective Cohort Study. Front. Microbiol. 2023, 14, 1114228. [Google Scholar] [CrossRef]
  75. Gomez-Arango, L.F.; Barrett, H.L.; McIntyre, H.D.; Callaway, L.K.; Morrison, M.; Dekker Nitert, M.; SPRING Trial Group. Connections between the Gut Microbiome and Metabolic Hormones in Early Pregnancy in Overweight and Obese Women. Diabetes 2016, 65, 2214–2223. [Google Scholar] [CrossRef]
  76. Torres, J.; Hu, J.; Seki, A.; Eisele, C.; Nair, N.; Huang, R.; Tarassishin, L.; Jharap, B.; Cote-Daigneault, J.; Mao, Q.; et al. Infants Born to Mothers with IBD Present with Altered Gut Microbiome That Transfers Abnormalities of the Adaptive Immune System to Germ-Free Mice. Gut 2020, 69, 42–51. [Google Scholar] [CrossRef]
  77. Moossavi, S.; Sepehri, S.; Robertson, B.; Bode, L.; Goruk, S.; Field, C.J.; Lix, L.M.; de Souza, R.J.; Becker, A.B.; Mandhane, P.J.; et al. Composition and Variation of the Human Milk Microbiota Are Influenced by Maternal and Early-Life Factors. Cell Host Microbe 2019, 25, 324–335.e4. [Google Scholar] [CrossRef]
  78. Peng, Y.; Tun, H.M.; Ng, S.C.; Wai, H.K.-F.; Zhang, X.; Parks, J.; Field, C.J.; Mandhane, P.; Moraes, T.J.; Simons, E.; et al. Maternal Smoking during Pregnancy Increases the Risk of Gut Microbiome-Associated Childhood Overweight and Obesity. Gut Microbes 2024, 16, 2323234. [Google Scholar] [CrossRef]
  79. Santarossa, S.; Sitarik, A.R.; Cassidy-Bushrow, A.E.; Comstock, S.S. Prenatal Physical Activity and the Gut Microbiota of Pregnant Women: Results from a Preliminary Investigation. Phys. Act. Nutr. 2023, 27, 1–7. [Google Scholar] [CrossRef]
  80. Bressa, C.; Bailén-Andrino, M.; Pérez-Santiago, J.; González-Soltero, R.; Pérez, M.; Montalvo-Lominchar, M.G.; Maté-Muñoz, J.L.; Domínguez, R.; Moreno, D.; Larrosa, M. Differences in Gut Microbiota Profile between Women with Active Lifestyle and Sedentary Women. PLoS ONE 2017, 12, e0171352. [Google Scholar] [CrossRef]
  81. Ryan, N.; O’Mahony, S.; Leahy-Warren, P.; Philpott, L.; Mulcahy, H. The Impact of Perinatal Maternal Stress on the Maternal and Infant Gut and Human Milk Microbiomes: A Scoping Review. PLoS ONE 2025, 20, e0318237. [Google Scholar] [CrossRef]
  82. Baker, M. Pregnancy Alters Resident Gut Microbes. Nature 2012, 488, 123. [Google Scholar] [CrossRef]
  83. Kennedy, K.M.; Plagemann, A.; Sommer, J.; Hofmann, M.; Henrich, W.; Barrett, J.F.R.; Surette, M.G.; Atkinson, S.; Braun, T.; Sloboda, D.M. Parity Modulates Impact of BMI and Gestational Weight Gain on Gut Microbiota in Human Pregnancy. Gut Microbes 2023, 15, 2259316. [Google Scholar] [CrossRef]
  84. Qi, X.; Yun, C.; Pang, Y.; Qiao, J. The Impact of the Gut Microbiota on the Reproductive and Metabolic Endocrine System. Gut Microbes 2021, 13, 1894070. [Google Scholar] [CrossRef]
  85. Amato, K.R.; Pradhan, P.; Mallott, E.K.; Shirola, W.; Lu, A. Host-Gut Microbiota Interactions during Pregnancy. Evol. Med. Public Health 2024, 12, 7–23. [Google Scholar] [CrossRef]
  86. DuPont, H.L.; Salge, M.M.H. The Importance of a Healthy Microbiome in Pregnancy and Infancy and Microbiota Treatment to Reverse Dysbiosis for Improved Health. Antibiotics 2023, 12, 1617. [Google Scholar] [CrossRef]
  87. Chen, X.; Li, P.; Liu, M.; Zheng, H.; He, Y.; Chen, M.-X.; Tang, W.; Yue, X.; Huang, Y.; Zhuang, L.; et al. Gut Dysbiosis Induces the Development of Pre-Eclampsia through Bacterial Translocation. Gut 2020, 69, 513–522. [Google Scholar] [CrossRef]
  88. Dunlop, A.L.; Mulle, J.G.; Ferranti, E.P.; Edwards, S.; Dunn, A.B.; Corwin, E.J. Maternal Microbiome and Pregnancy Outcomes That Impact Infant Health: A Review: A Review. Adv. Neonatal Care 2015, 15, 377–385. [Google Scholar] [CrossRef] [PubMed]
  89. Bauman, M.D.; Iosif, A.-M.; Smith, S.E.P.; Bregere, C.; Amaral, D.G.; Patterson, P.H. Activation of the Maternal Immune System during Pregnancy Alters Behavioral Development of Rhesus Monkey Offspring. Biol. Psychiatry 2014, 75, 332–341. [Google Scholar] [CrossRef] [PubMed]
  90. Jarde, A.; Lewis-Mikhael, A.-M.; Moayyedi, P.; Stearns, J.C.; Collins, S.M.; Beyene, J.; McDonald, S.D. Pregnancy Outcomes in Women Taking Probiotics or Prebiotics: A Systematic Review and Meta-Analysis. BMC Pregnancy Childbirth 2018, 18, 14. [Google Scholar] [CrossRef]
  91. Rao, S.C.; Athalye-Jape, G.K.; Deshpande, G.C.; Simmer, K.N.; Patole, S.K. Probiotic Supplementation and Late-Onset Sepsis in Preterm Infants: A Meta-Analysis. Pediatrics 2016, 137, e20153684. [Google Scholar] [CrossRef] [PubMed]
  92. Aceti, A.; Maggio, L.; Beghetti, I.; Gori, D.; Barone, G.; Callegari, M.L.; Fantini, M.P.; Indrio, F.; Meneghin, F.; Morelli, L.; et al. Probiotics Prevent Late-Onset Sepsis in Human Milk-Fed, Very Low Birth Weight Preterm Infants: Systematic Review and Meta-Analysis. Nutrients 2017, 9, 904. [Google Scholar] [CrossRef] [PubMed]
  93. Buggio, L.; Somigliana, E.; Borghi, A.; Vercellini, P. Probiotics and Vaginal Microecology: Fact or Fancy? BMC Womens Health 2019, 19, 25. [Google Scholar] [CrossRef]
  94. Pronovost, G.N.; Yu, K.B.; Coley-O’Rourke, E.J.L.; Telang, S.S.; Chen, A.S.; Vuong, H.E.; Williams, D.W.; Chandra, A.; Rendon, T.K.; Paramo, J.; et al. The Maternal Microbiome Promotes Placental Development in Mice. Sci. Adv. 2023, 9, eadk1887. [Google Scholar] [CrossRef]
  95. Lopez-Tello, J.; Schofield, Z.; Kiu, R.; Dalby, M.J.; van Sinderen, D.; Le Gall, G.; Sferruzzi-Perri, A.N.; Hall, L.J. Maternal Gut Microbiota Bifidobacterium Promotes Placental Morphogenesis, Nutrient Transport and Fetal Growth in Mice. Cell. Mol. Life Sci. 2022, 79, 386. [Google Scholar] [CrossRef]
  96. Perez-Muñoz, M.E.; Arrieta, M.-C.; Ramer-Tait, A.E.; Walter, J. A Critical Assessment of the “Sterile Womb” and “in Utero Colonization” Hypotheses: Implications for Research on the Pioneer Infant Microbiome. Microbiome 2017, 5, 48. [Google Scholar] [CrossRef]
  97. Kennedy, K.M.; de Goffau, M.C.; Perez-Muñoz, M.E.; Arrieta, M.-C.; Bäckhed, F.; Bork, P.; Braun, T.; Bushman, F.D.; Dore, J.; de Vos, W.M.; et al. Questioning the Fetal Microbiome Illustrates Pitfalls of Low-Biomass Microbial Studies. Nature 2023, 613, 639–649. [Google Scholar] [CrossRef]
  98. Xie, Z.; Chen, Z.; Chai, Y.; Yao, W.; Ma, G. Unveiling the Placental Bacterial Microbiota: Implications for Maternal and Infant Health. Front. Physiol. 2025, 16, 1544216. [Google Scholar] [CrossRef]
  99. Ygberg, S.; Nilsson, A. The Developing Immune System—From Foetus to Toddler. Acta Paediatr. 2012, 101, 120–127. [Google Scholar] [CrossRef]
  100. McDavid, A.; Laniewski, N.; Grier, A.; Gill, A.L.; Kessler, H.A.; Huyck, H.; Carbonell, E.; Holden-Wiltse, J.; Bandyopadhyay, S.; Carnahan, J.; et al. Aberrant Newborn T Cell and Microbiota Developmental Trajectories Predict Respiratory Compromise during Infancy. iScience 2022, 25, 104007. [Google Scholar] [CrossRef] [PubMed]
  101. Gao, Y.; O’Hely, M.; Quinn, T.P.; Ponsonby, A.-L.; Harrison, L.C.; Frøkiær, H.; Tang, M.L.K.; Brix, S.; Kristiansen, K.; Burgner, D.; et al. Maternal Gut Microbiota during Pregnancy and the Composition of Immune Cells in Infancy. Front. Immunol. 2022, 13, 986340. [Google Scholar] [CrossRef] [PubMed]
  102. Gonzalez-Perez, G.; Hicks, A.L.; Tekieli, T.M.; Radens, C.M.; Williams, B.L.; Lamousé-Smith, E.S.N. Maternal Antibiotic Treatment Impacts Development of the Neonatal Intestinal Microbiome and Antiviral Immunity. J. Immunol. 2016, 196, 3768–3779. [Google Scholar] [CrossRef]
  103. Levan, S.R.; Stamnes, K.A.; Lin, D.L.; Panzer, A.R.; Fukui, E.; McCauley, K.; Fujimura, K.E.; McKean, M.; Ownby, D.R.; Zoratti, E.M.; et al. Elevated Faecal 12,13-DiHOME Concentration in Neonates at High Risk for Asthma Is Produced by Gut Bacteria and Impedes Immune Tolerance. Nat. Microbiol. 2019, 4, 1851–1861, Erratum in Nat. Microbiol. 2019, 4, 2020. [Google Scholar] [CrossRef]
  104. Gao, Y.; Nanan, R.; Macia, L.; Tan, J.; Sominsky, L.; Quinn, T.P.; O’Hely, M.; Ponsonby, A.-L.; Tang, M.L.K.; Collier, F.; et al. The Maternal Gut Microbiome during Pregnancy and Offspring Allergy and Asthma. J. Allergy Clin. Immunol. 2021, 148, 669–678. [Google Scholar] [CrossRef]
  105. de Moraes-Pinto, M.I.; Suano-Souza, F.; Aranda, C.S. Immune System: Development and Acquisition of Immunological Competence. J. Pediatr. (Rio J.) 2021, 97 (Suppl. 1), S59–S66. [Google Scholar] [CrossRef]
  106. Timm, S.; Schlünssen, V.; Olsen, J.; Ramlau-Hansen, C.H. Prenatal Antibiotics and Atopic Dermatitis among 18-Month-Old Children in the Danish National Birth Cohort. Clin. Exp. Allergy 2017, 47, 929–936. [Google Scholar] [CrossRef]
  107. Faas, M.M.; Smink, A.M. Shaping Immunity: The Influence of the Maternal Gut Bacteria on Fetal Immune Development. Semin. Immunopathol. 2025, 47, 13. [Google Scholar] [CrossRef]
  108. Sun, Y.; Huang, S.; Li, M.; Yang, Y.; Ma, J.; Xie, R.; Wang, J.; Zhao, Q.; Qin, S.; He, L.; et al. Maternal High-Fat Diet Disrupts Intestinal Mucus Barrier of Offspring by Regulating Gut Immune Receptor LRRC19. Commun. Biol. 2025, 8, 402. [Google Scholar] [CrossRef] [PubMed]
  109. Babu, S.T.; Niu, X.; Raetz, M.; Savani, R.C.; Hooper, L.V.; Mirpuri, J. Maternal High-Fat Diet Results in Microbiota-Dependent Expansion of ILC3s in Mice Offspring. JCI Insight 2018, 3, e99223. [Google Scholar] [CrossRef]
  110. Wallace, J.G.; Bellissimo, C.J.; Yeo, E.; Fei Xia, Y.; Petrik, J.J.; Surette, M.G.; Bowdish, D.M.E.; Sloboda, D.M. Obesity during Pregnancy Results in Maternal Intestinal Inflammation, Placental Hypoxia, and Alters Fetal Glucose Metabolism at Mid-Gestation. Sci. Rep. 2019, 9, 17621. [Google Scholar] [CrossRef]
  111. Liang, W.; Feng, Y.; Yang, D.; Qin, J.; Zhi, X.; Wu, W.; Jie, Q. Oral Probiotics Increased the Proportion of Treg, Tfr, and Breg Cells to Inhibit the Inflammatory Response and Impede Gestational Diabetes Mellitus. Mol. Med. 2023, 29, 122. [Google Scholar] [CrossRef] [PubMed]
  112. Brosseau, C.; Selle, A.; Duval, A.; Misme-Aucouturier, B.; Chesneau, M.; Brouard, S.; Cherbuy, C.; Cariou, V.; Bouchaud, G.; Mincham, K.T.; et al. Prebiotic Supplementation during Pregnancy Modifies the Gut Microbiota and Increases Metabolites in Amniotic Fluid, Driving a Tolerogenic Environment in Utero. Front. Immunol. 2021, 12, 712614. [Google Scholar] [CrossRef]
  113. Hu, M.; Eviston, D.; Hsu, P.; Mariño, 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]
  114. Tan, J.; McKenzie, C.; Potamitis, M.; Thorburn, A.N.; Mackay, C.R.; Macia, L. The Role of Short-Chain Fatty Acids in Health and Disease. Adv. Immunol. 2014, 121, 91–119. [Google Scholar]
  115. Thorburn, A.N.; McKenzie, C.I.; Shen, S.; Stanley, D.; Macia, L.; Mason, L.J.; Roberts, L.K.; Wong, C.H.Y.; 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]
  116. Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T.; et al. Commensal Microbe-Derived Butyrate Induces the Differentiation of Colonic Regulatory T Cells. Nature 2013, 504, 446–450, Erratum in Nature 2014, 506, 254. [Google Scholar] [CrossRef]
  117. Jain, N. The Early Life Education of the Immune System: Moms, Microbes and (Missed) Opportunities. Gut Microbes 2020, 12, 1824564. [Google Scholar] [CrossRef]
  118. Humann, J.; Mann, B.; Gao, G.; Moresco, P.; Ramahi, J.; Loh, L.N.; Farr, A.; Hu, Y.; Durick-Eder, K.; Fillon, S.A.; et al. Bacterial Peptidoglycan Traverses the Placenta to Induce Fetal Neuroproliferation and Aberrant Postnatal Behavior. Cell Host Microbe 2016, 19, 388–399. [Google Scholar] [CrossRef]
  119. Gomez de Agüero, M.; Ganal-Vonarburg, S.C.; Fuhrer, T.; Rupp, S.; Uchimura, Y.; Li, H.; Steinert, A.; Heikenwalder, M.; Hapfelmeier, S.; Sauer, U.; et al. The Maternal Microbiota Drives Early Postnatal Innate Immune Development. Science 2016, 351, 1296–1302. [Google Scholar] [CrossRef] [PubMed]
  120. Kaisanlahti, A.; Turunen, J.; Byts, N.; Samoylenko, A.; Bart, G.; Virtanen, N.; Tejesvi, M.V.; Zhyvolozhnyi, A.; Sarfraz, S.; Kumpula, S.; et al. Maternal Microbiota Communicates with the Fetus through Microbiota-Derived Extracellular Vesicles. Microbiome 2023, 11, 249. [Google Scholar] [CrossRef]
  121. Li, Q.; Barres, B.A. Microglia and Macrophages in Brain Homeostasis and Disease. Nat. Rev. Immunol. 2018, 18, 225–242. [Google Scholar] [CrossRef]
  122. Zengeler, K.E.; Lukens, J.R. Innate Immunity at the Crossroads of Healthy Brain Maturation and Neurodevelopmental Disorders. Nat. Rev. Immunol. 2021, 21, 454–468. [Google Scholar] [CrossRef]
  123. Yeo, X.Y.; Choi, Y.; Hong, Y.; Kwon, H.N.; Jung, S. Contemporary Insights into Neuroimmune Interactions across Development and Aging. Front. Neurol. 2025, 16, 1611124. [Google Scholar] [CrossRef] [PubMed]
  124. Gohlke, J.M.; Stockton, P.S.; Sieber, S.; Foley, J.; Portier, C.J. AhR-Mediated Gene Expression in the Developing Mouse Telencephalon. Reprod. Toxicol. 2009, 28, 321–328. [Google Scholar] [CrossRef]
  125. Kim, S.; Kim, H.; Yim, Y.S.; Ha, S.; Atarashi, K.; Tan, T.G.; Longman, R.S.; Honda, K.; Littman, D.R.; Choi, G.B.; et al. Maternal Gut Bacteria Promote Neurodevelopmental Abnormalities in Mouse Offspring. Nature 2017, 549, 528–532. [Google Scholar] [CrossRef]
  126. Kim, E.; Paik, D.; Ramirez, R.N.; Biggs, D.G.; Park, Y.; Kwon, H.-K.; Choi, G.B.; Huh, J.R. Maternal Gut Bacteria Drive Intestinal Inflammation in Offspring with Neurodevelopmental Disorders by Altering the Chromatin Landscape of CD4+ T Cells. Immunity 2022, 55, 145–158.e7. [Google Scholar] [CrossRef]
  127. Choi, G.B.; Yim, Y.S.; Wong, H.; Kim, S.; Kim, H.; Kim, S.V.; Hoeffer, C.A.; Littman, D.R.; Huh, J.R. The Maternal Interleukin-17a Pathway in Mice Promotes Autism-like Phenotypes in Offspring. Science 2016, 351, 933–939. [Google Scholar] [CrossRef]
  128. Sun, Z.; Lee-Sarwar, K.; Kelly, R.S.; Lasky-Su, J.A.; Litonjua, A.A.; Weiss, S.T.; Liu, Y.-Y. Revealing the Importance of Prenatal Gut Microbiome in Offspring Neurodevelopment in Humans. EBioMedicine 2023, 90, 104491. [Google Scholar] [CrossRef]
  129. Surzenko, N.; Pjetri, E.; Munson, C.A.; Friday, W.B.; Hauser, J.; Mitchell, E.S. Prenatal Exposure to the Probiotic Lactococcus Lactis Decreases Anxiety-like Behavior and Modulates Cortical Cytoarchitecture in a Sex Specific Manner. PLoS ONE 2020, 15, e0223395. [Google Scholar] [CrossRef]
  130. Sarker, G.; Peleg-Raibstein, D. Maternal Overnutrition Induces Long-Term Cognitive Deficits across Several Generations. Nutrients 2018, 11, 7. [Google Scholar] [CrossRef] [PubMed]
  131. Liu, X.; Li, X.; Xia, B.; Jin, X.; Zou, Q.; Zeng, Z.; Zhao, W.; Yan, S.; Li, L.; Yuan, S.; et al. High-Fiber Diet Mitigates Maternal Obesity-Induced Cognitive and Social Dysfunction in the Offspring via Gut-Brain Axis. Cell Metab. 2021, 33, 923–938.e6. [Google Scholar] [CrossRef]
  132. Falsaperla, R.; Sciuto, S.; Gioè, D.; Sciuto, L.; Pisani, F.; Pavone, P.; Ruggieri, M. Mild Hypoxic-Ischemic Encephalopathy: Can Neurophysiological Monitoring Predict Unfavorable Neurological Outcome? A Systematic Review and Meta-Analysis. Am. J. Perinatol. 2023, 40, 833–838. [Google Scholar] [CrossRef]
  133. Hayes, B.C.; Ryan, S.; McGarvey, C.; Mulvany, S.; Doherty, E.; Grehan, A.; Madigan, C.; Matthews, T.; King, M.D. Brain Magnetic Resonance Imaging and Outcome after Hypoxic Ischaemic Encephalopathy. J. Matern. Fetal. Neonatal Med. 2016, 29, 777–782. [Google Scholar] [CrossRef] [PubMed]
  134. Törn, A.E.; Hesselman, S.; Johansen, K.; Ågren, J.; Wikström, A.-K.; Jonsson, M. Outcomes in Children after Mild Neonatal Hypoxic Ischaemic Encephalopathy: A Population-Based Cohort Study. BJOG 2023, 130, 1602–1609. [Google Scholar] [CrossRef]
  135. DuPont, T.L.; Chalak, L.F.; Morriss, M.C.; Burchfield, P.J.; Christie, L.; Sánchez, P.J. Short-Term Outcomes of Newborns with Perinatal Acidemia Who Are Not Eligible for Systemic Hypothermia Therapy. J. Pediatr. 2013, 162, 35–41. [Google Scholar] [CrossRef]
  136. Chen, A.; Teng, C.; Wei, J.; Wu, X.; Zhang, H.; Chen, P.; Cai, D.; Qian, H.; Zhu, H.; Zheng, X.; et al. Gut Microbial Dysbiosis Exacerbates Long-Term Cognitive Impairments by Promoting Intestinal Dysfunction and Neuroinflammation Following Neonatal Hypoxia-Ischemia. Gut Microbes 2025, 17, 2471015. [Google Scholar] [CrossRef]
  137. Hsu, C.-Y.; Khachatryan, L.G.; Younis, N.K.; Mustafa, M.A.; Ahmad, N.; Athab, Z.H.; Polyanskaya, A.V.; Kasanave, E.V.; Mirzaei, R.; Karampoor, S. Microbiota-Derived Short Chain Fatty Acids in Pediatric Health and Diseases: From Gut Development to Neuroprotection. Front. Microbiol. 2024, 15, 1456793. [Google Scholar] [CrossRef]
  138. Chakkarapani, A.A.; Aly, H.; Benders, M.; Cotten, C.M.; El-Dib, M.; Gressens, P.; Hagberg, H.; Sabir, H.; Wintermark, P.; Robertson, N.J.; et al. Therapies for Neonatal Encephalopathy: Targeting the Latent, Secondary and Tertiary Phases of Evolving Brain Injury. Semin. Fetal Neonatal Med. 2021, 26, 101256. [Google Scholar] [CrossRef] [PubMed]
  139. Perlman, J.M. Intervention Strategies for Neonatal Hypoxic-Ischemic Cerebral Injury. Clin. Ther. 2006, 28, 1353–1365. [Google Scholar] [CrossRef]
  140. Aly, H.; Khashaba, M.T.; El-Ayouty, M.; El-Sayed, O.; Hasanein, B.M. IL-1beta, IL-6 and TNF-Alpha and Outcomes of Neonatal Hypoxic Ischemic Encephalopathy. Brain Dev. 2006, 28, 178–182. [Google Scholar] [CrossRef]
  141. O’Dea, M.I.; Kelly, L.A.; McKenna, E.; Strickland, T.; Hurley, T.P.; Butler, J.; Vavasseur, C.; El-Khuffash, A.F.; Miletin, J.; Fallah, L.; et al. Altered Cytokine Endotoxin Responses in Neonatal Encephalopathy Predict MRI Outcomes. Front. Pediatr. 2021, 9, 734540. [Google Scholar] [CrossRef]
  142. Ziemka-Nalecz, M.; Jaworska, J.; Sypecka, J.; Polowy, R.; Filipkowski, R.K.; Zalewska, T. Sodium Butyrate, a Histone Deacetylase Inhibitor, Exhibits Neuroprotective/Neurogenic Effects in a Rat Model of Neonatal Hypoxia-Ischemia. Mol. Neurobiol. 2017, 54, 5300–5318. [Google Scholar] [CrossRef]
  143. Lewis, K.; Lutgendorff, F.; Phan, V.; Söderholm, J.D.; Sherman, P.M.; McKay, D.M. Enhanced Translocation of Bacteria across Metabolically Stressed Epithelia Is Reduced by Butyrate. Inflamm. Bowel Dis. 2010, 16, 1138–1148. [Google Scholar] [CrossRef] [PubMed]
  144. Knox, E.G.; Aburto, M.R.; Tessier, C.; Nagpal, J.; Clarke, G.; O’Driscoll, C.M.; Cryan, J.F. Microbial-Derived Metabolites Induce Actin Cytoskeletal Rearrangement and Protect Blood-Brain Barrier Function. iScience 2022, 25, 105648. [Google Scholar] [CrossRef]
  145. Wojciech, L.; Tan, K.S.W.; Gascoigne, N.R.J. Taming the Sentinels: Microbiome-Derived Metabolites and Polarization of T Cells. Int. J. Mol. Sci. 2020, 21, 7740. [Google Scholar] [CrossRef]
  146. Mancino, S.; Boraso, M.; Galmozzi, A.; Serafini, M.M.; De Fabiani, E.; Crestani, M.; Viviani, B. Dose-Dependent Dual Effects of HDAC Inhibitors on Glial Inflammatory Response. Sci. Rep. 2025, 15, 12262. [Google Scholar] [CrossRef] [PubMed]
  147. Leus, N.G.; Zwinderman, M.R.; Dekker, F.J. Histone Deacetylase 3 (HDAC 3) as Emerging Drug Target in NF-ΚB-Mediated Inflammation. Curr. Opin. Chem. Biol. 2016, 33, 160–168. [Google Scholar] [CrossRef]
  148. Caetano-Silva, M.E.; Rund, L.; Hutchinson, N.T.; Woods, J.A.; Steelman, A.J.; Johnson, R.W. Inhibition of Inflammatory Microglia by Dietary Fiber and Short-Chain Fatty Acids. Sci. Rep. 2023, 13, 2819. [Google Scholar] [CrossRef]
  149. Zhou, Z.; Xu, N.; Matei, N.; McBride, D.W.; Ding, Y.; Liang, H.; Tang, J.; Zhang, J.H. Sodium Butyrate Attenuated Neuronal Apoptosis via GPR41/Gβγ/PI3K/Akt Pathway after MCAO in Rats. J. Cereb. Blood Flow Metab. 2021, 41, 267–281. [Google Scholar] [CrossRef]
  150. Sun, J.; Lu, L.; Lian, Y.; Xu, S.; Zhu, Y.; Wu, Y.; Lin, Q.; Hou, J.; Li, Y.; Yu, Z. Sodium Butyrate Attenuates Microglia-Mediated Neuroinflammation by Modulating the TLR4/MyD88/NF-ΚB Pathway and Microbiome-Gut-Brain Axis in Cardiac Arrest Mice. Mol. Brain 2025, 18, 13. [Google Scholar] [CrossRef] [PubMed]
  151. Zhang, R.; Chen, X.; Shen, Q.; Liu, L.; Hou, X.; Liu, N. Sodium Butyrate Improves the Effects of Brain Injury in a Small-for-Gestational-Age Rat Model by Activating the JAK1/STAT3 Pathway. J. Neuropathol. Exp. Neurol. 2025, nlaf085. [Google Scholar] [CrossRef]
  152. Dietz, R.M.; Wright, C.J. Oxidative Stress Diseases Unique to the Perinatal Period: A Window into the Developing Innate Immune Response. Am. J. Reprod. Immunol. 2018, 79, e12787. [Google Scholar] [CrossRef] [PubMed]
  153. Silva, Y.P.; Bernardi, A.; Frozza, R.L. The Role of Short-Chain Fatty Acids from Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. 2020, 11, 25. [Google Scholar] [CrossRef]
  154. Hoyles, L.; Snelling, T.; Umlai, U.-K.; Nicholson, J.K.; Carding, S.R.; Glen, R.C.; McArthur, S. Microbiome-Host Systems Interactions: Protective Effects of Propionate upon the Blood-Brain Barrier. Microbiome 2018, 6, 55. [Google Scholar] [CrossRef] [PubMed]
  155. Wei, J.; Chen, A.; Huang, D.; Teng, C.; Cai, D.; Wu, X.; Wang, T.; Hu, W.; Huang, Z.; Wang, P.; et al. Gut Microbiome-Derived Lipopolysaccharides Aggravate Cognitive Impairment via TLR4-Mediated Inflammatory Signaling in Neonatal Rats Following Hypoxic-Ischemic Brain Damage. Brain Behav. Immun. 2025, 127, 4–24. [Google Scholar] [CrossRef] [PubMed]
  156. Kaisar, M.M.M.; Pelgrom, L.R.; van der Ham, A.J.; Yazdanbakhsh, M.; Everts, B. Butyrate Conditions Human Dendritic Cells to Prime Type 1 Regulatory T Cells via Both Histone Deacetylase Inhibition and G Protein-Coupled Receptor 109A Signaling. Front. Immunol. 2017, 8, 1429. [Google Scholar] [CrossRef]
  157. Tao, R.; de Zoeten, E.F.; Ozkaynak, E.; Chen, C.; Wang, L.; Porrett, P.M.; Li, B.; Turka, L.A.; Olson, E.N.; Greene, M.I.; et al. Deacetylase Inhibition Promotes the Generation and Function of Regulatory T Cells. Nat. Med. 2007, 13, 1299–1307. [Google Scholar] [CrossRef]
  158. Pessa-Morikawa, T.; Husso, A.; Kärkkäinen, O.; Koistinen, V.; Hanhineva, K.; Iivanainen, A.; Niku, M. Maternal Microbiota-Derived Metabolic Profile in Fetal Murine Intestine, Brain and Placenta. BMC Microbiol. 2022, 22, 46. [Google Scholar] [CrossRef]
  159. Peesh, P.; Blasco-Conesa, M.P.; El Hamamy, A.; Khan, R.; Guzman, G.U.; Honarpisheh, P.; Mohan, E.C.; Goodman, G.W.; Nguyen, J.N.; Banerjee, A.; et al. Benefits of Equilibrium between Microbiota- and Host-Derived Ligands of the Aryl Hydrocarbon Receptor after Stroke in Aged Male Mice. Nat. Commun. 2025, 16, 1767. [Google Scholar] [CrossRef]
  160. Miyamoto, K.; Sujino, T.; Kanai, T. The Tryptophan Metabolic Pathway of the Microbiome and Host Cells in Health and Disease. Int. Immunol. 2024, 36, 601–616. [Google Scholar] [CrossRef]
  161. Honarpisheh, P.; Bryan, R.M.; McCullough, L.D. Aging Microbiota-Gut-Brain Axis in Stroke Risk and Outcome. Circ. Res. 2022, 130, 1112–1144. [Google Scholar] [CrossRef] [PubMed]
  162. Wang, T.; Chen, B.; Luo, M.; Xie, L.; Lu, M.; Lu, X.; Zhang, S.; Wei, L.; Zhou, X.; Yao, B.; et al. Microbiota-Indole 3-Propionic Acid-Brain Axis Mediates Abnormal Synaptic Pruning of Hippocampal Microglia and Susceptibility to ASD in IUGR Offspring. Microbiome 2023, 11, 245. [Google Scholar] [CrossRef]
  163. Li, Y.; Wang, T.; Sun, P.; Zhu, W.; Chen, Y.; Chen, M.; Yang, X.; Du, X.; Zhao, Y. Farrerol Alleviates Hypoxic-Ischemic Encephalopathy by Inhibiting Ferroptosis in Neonatal Rats via the Nrf2 Pathway. Physiol. Res. 2023, 72, 511–520. [Google Scholar] [CrossRef] [PubMed]
  164. Peeples, E.S.; Genaro-Mattos, T.C. Ferroptosis: A Promising Therapeutic Target for Neonatal Hypoxic-Ischemic Brain Injury. Int. J. Mol. Sci. 2022, 23, 7420. [Google Scholar] [CrossRef] [PubMed]
  165. Fang, Y.; Zheng, Y.; Gao, Q.; Pang, M.; Wu, Y.; Feng, X.; Tao, X.; Hu, Y.; Lin, Z.; Lin, W. Activation of the Nrf2/Keap1 Signaling Pathway Mediates the Neuroprotective Effect of Perillyl Alcohol against Cerebral Hypoxic-Ischemic Damage in Neonatal Rats. Redox Rep. 2024, 29, 2394714. [Google Scholar] [CrossRef]
  166. Liu, J.-X.; Zheng, D.; Chen, L.; Chen, S.; Min, J.-W. Nuclear Factor Erythroid 2-Related Factor 2 as a Potential Therapeutic Target in Neonatal Hypoxic-Ischemic Encephalopathy. J. Integr. Neurosci. 2024, 23, 103. [Google Scholar] [CrossRef]
  167. Li, Y.; Liu, H.; Zhang, L.; Yang, Y.; Lin, Y.; Zhuo, Y.; Fang, Z.; Che, L.; Feng, B.; Xu, S.; et al. Maternal Dietary Fiber Composition during Gestation Induces Changes in Offspring Antioxidative Capacity, Inflammatory Response, and Gut Microbiota in a Sow Model. Int. J. Mol. Sci. 2019, 21, 31. [Google Scholar] [CrossRef]
  168. Zhang, J.; Wang, J.; Ma, Z.; Fu, Z.; Zhao, Y.; Zeng, X.; Lin, G.; Zhang, S.; Guan, W.; Chen, F. Enhanced Antioxidative Capacity Transfer between Sow and Fetus via the Gut-Placenta Axis with Dietary Selenium Yeast and Glycerol Monolaurate Supplementation during Pregnancy. Antioxidants 2024, 13, 141. [Google Scholar] [CrossRef]
  169. Singh, R.; Chandrashekharappa, S.; Bodduluri, S.R.; Baby, B.V.; Hegde, B.; Kotla, N.G.; Hiwale, A.A.; Saiyed, T.; Patel, P.; Vijay-Kumar, M.; et al. Enhancement of the Gut Barrier Integrity by a Microbial Metabolite through the Nrf2 Pathway. Nat. Commun. 2019, 10, 89. [Google Scholar] [CrossRef]
  170. Kunst, C.; Schmid, S.; Michalski, M.; Tümen, D.; Buttenschön, J.; Müller, M.; Gülow, K. The Influence of Gut Microbiota on Oxidative Stress and the Immune System. Biomedicines 2023, 11, 1388. [Google Scholar] [CrossRef]
  171. Uchiyama, J.; Akiyama, M.; Hase, K.; Kumagai, Y.; Kim, Y.-G. Gut Microbiota Reinforce Host Antioxidant Capacity via the Generation of Reactive Sulfur Species. Cell Rep. 2022, 38, 110479. [Google Scholar] [CrossRef]
  172. Xu, K.; Liu, G.; Fu, C. The Tryptophan Pathway Targeting Antioxidant Capacity in the Placenta. Oxidative Medicine and Cellular Longevity 2018, 2018, 1054797. [Google Scholar] [CrossRef]
  173. Cömert, T.K.; Akpinar, F.; Erkaya, S.; Durmaz, B.; Durmaz, R. The Effect of Gestational Weight Gain on Serum Total Oxidative Stress, Total Antioxidant Capacity and Gut Microbiota. Biosci. Microbiota Food Health 2022, 41, 160–167. [Google Scholar] [CrossRef]
  174. Du, X.; Elsabagh, M.; He, F.; Wu, H.; Zhang, B.; Fan, K.; Wang, M.; Zhang, H. Gut Microbiota and Its Metabolites Modulate Pregnancy Outcomes by Regulating Placental Autophagy and Ferroptosis. Antioxidants 2025, 14, 970. [Google Scholar] [CrossRef]
  175. Jithoo, A.; Penny, T.R.; Pham, Y.; Sutherland, A.E.; Smith, M.J.; Petraki, M.; Fahey, M.C.; Jenkin, G.; Malhotra, A.; Miller, S.L.; et al. The Temporal Relationship between Blood-Brain Barrier Integrity and Microglial Response Following Neonatal Hypoxia Ischemia. Cells 2024, 13, 660. [Google Scholar] [CrossRef]
  176. Braniste, V.; Al-Asmakh, M.; Kowal, C.; Anuar, F.; Abbaspour, A.; Tóth, M.; Korecka, A.; Bakocevic, N.; Ng, L.G.; Kundu, P.; et al. The Gut Microbiota Influences Blood-Brain Barrier Permeability in Mice. Sci. Transl. Med. 2014, 6, 263ra158, Erratum in Sci. Transl. Med. 2014, 6, 266er7. [Google Scholar] [CrossRef]
  177. González-Bosch, C.; Boorman, E.; Zunszain, P.A.; Mann, G.E. Short-Chain Fatty Acids as Modulators of Redox Signaling in Health and Disease. Redox Biol. 2021, 47, 102165. [Google Scholar] [CrossRef]
  178. Sun, J.; Wang, F.; Li, H.; Zhang, H.; Jin, J.; Chen, W.; Pang, M.; Yu, J.; He, Y.; Liu, J.; et al. Neuroprotective Effect of Sodium Butyrate against Cerebral Ischemia/Reperfusion Injury in Mice. BioMed Res. Int. 2015, 2015, 395895. [Google Scholar] [CrossRef]
  179. Sun, J.; Ling, Z.; Wang, F.; Chen, W.; Li, H.; Jin, J.; Zhang, H.; Pang, M.; Yu, J.; Liu, J. Clostridium Butyricum Pretreatment Attenuates Cerebral Ischemia/Reperfusion Injury in Mice via Anti-Oxidation and Anti-Apoptosis. Neurosci. Lett. 2016, 613, 30–35. [Google Scholar] [CrossRef] [PubMed]
  180. Park, M.J.; Sohrabji, F. The Histone Deacetylase Inhibitor, Sodium Butyrate, Exhibits Neuroprotective Effects for Ischemic Stroke in Middle-Aged Female Rats. J. Neuroinflamm. 2016, 13, 300. [Google Scholar] [CrossRef] [PubMed]
  181. Liu, X.; Tao, X.; Xiong, Z.; Wang, H.; Zeng, L. Mechanism of HDAC2 Regulating Nrf2 Acetylation Level in Neuronal Ferroptosis of Neonatal Rats with Hypoxic-Ischemic Brain Injury. Brain Inj. 2025, 39, 635–645. [Google Scholar] [CrossRef]
  182. Hwang, I.K.; Yoo, K.-Y.; Li, H.; Park, O.K.; Lee, C.H.; Choi, J.H.; Jeong, Y.-G.; Lee, Y.L.; Kim, Y.-M.; Kwon, Y.-G.; et al. Indole-3-Propionic Acid Attenuates Neuronal Damage and Oxidative Stress in the Ischemic Hippocampus. J. Neurosci. Res. 2009, 87, 2126–2137. [Google Scholar] [CrossRef] [PubMed]
  183. Basak, S.; Das, R.K.; Banerjee, A.; Paul, S.; Pathak, S.; Duttaroy, A.K. Maternal Obesity and Gut Microbiota Are Associated with Fetal Brain Development. Nutrients 2022, 14, 4515. [Google Scholar] [CrossRef]
  184. Radford-Smith, D.E.; Probert, F.; Burnet, P.W.J.; Anthony, D.C. Modifying the Maternal Microbiota Alters the Gut-Brain Metabolome and Prevents Emotional Dysfunction in the Adult Offspring of Obese Dams. Proc. Natl. Acad. Sci. USA 2022, 119, e2108581119. [Google Scholar] [CrossRef] [PubMed]
  185. He, X.; Zhang, T.; Zeng, Y.; Pei, P.; Liu, Y.; Jia, W.; Zhao, H.; Bi, M.; Wang, S. Sodium Butyrate Mediates Histone Crotonylation and Alleviated Neonatal Rats Hypoxic-Ischemic Brain Injury through Gut-Brain Axis. Front. Microbiol. 2022, 13, 993146. [Google Scholar] [CrossRef]
  186. Chen, A.; Xiong, L.-J.; Tong, Y.; Mao, M. The Neuroprotective Roles of BDNF in Hypoxic Ischemic Brain Injury. Biomed. Rep. 2013, 1, 167–176. [Google Scholar] [CrossRef]
  187. Urbonaite, G.; Knyzeliene, A.; Bunn, F.S.; Smalskys, A.; Neniskyte, U. The Impact of Maternal High-Fat Diet on Offspring Neurodevelopment. Front. Neurosci. 2022, 16, 909762. [Google Scholar] [CrossRef]
  188. Fusco, S.; Spinelli, M.; Cocco, S.; Ripoli, C.; Mastrodonato, A.; Natale, F.; Rinaudo, M.; Livrizzi, G.; Grassi, C. Maternal Insulin Resistance Multigenerationally Impairs Synaptic Plasticity and Memory via Gametic Mechanisms. Nat. Commun. 2019, 10, 4799. [Google Scholar] [CrossRef] [PubMed]
  189. Page, K.C.; Jones, E.K.; Anday, E.K. Maternal and Postweaning High-Fat Diets Disturb Hippocampal Gene Expression, Learning, and Memory Function. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2014, 306, R527–R537. [Google Scholar] [CrossRef]
  190. Niculescu, M.D.; Lupu, D.S. High Fat Diet-Induced Maternal Obesity Alters Fetal Hippocampal Development. Int. J. Dev. Neurosci. 2009, 27, 627–633. [Google Scholar] [CrossRef]
  191. Mastenbroek, L.J.M.; Kooistra, S.M.; Eggen, B.J.L.; Prins, J.R. The Role of Microglia in Early Neurodevelopment and the Effects of Maternal Immune Activation. Semin. Immunopathol. 2024, 46, 1. [Google Scholar] [CrossRef]
  192. Cavaliere, G.; Catapano, A.; Trinchese, G.; Cimmino, F.; Penna, E.; Pizzella, A.; Cristiano, C.; Lama, A.; Crispino, M.; Mollica, M.P. Butyrate Improves Neuroinflammation and Mitochondrial Impairment in Cerebral Cortex and Synaptic Fraction in an Animal Model of Diet-Induced Obesity. Antioxidants 2022, 12, 4. [Google Scholar] [CrossRef]
  193. Jiang, Y.; Li, K.; Li, X.; Xu, L.; Yang, Z. Sodium Butyrate Ameliorates the Impairment of Synaptic Plasticity by Inhibiting the Neuroinflammation in 5XFAD Mice. Chem. Biol. Interact. 2021, 341, 109452. [Google Scholar] [CrossRef]
  194. Jaworska, J.; Ziemka-Nalecz, M.; Sypecka, J.; Zalewska, T. The Potential Neuroprotective Role of a Histone Deacetylase Inhibitor, Sodium Butyrate, after Neonatal Hypoxia-Ischemia. J. Neuroinflamm. 2017, 14, 34. [Google Scholar] [CrossRef]
  195. Jaworska, J.; Zalewska, T.; Sypecka, J.; Ziemka-Nalecz, M. Effect of the HDAC Inhibitor, Sodium Butyrate, on Neurogenesis in a Rat Model of Neonatal Hypoxia-Ischemia: Potential Mechanism of Action. Mol. Neurobiol. 2019, 56, 6341–6370. [Google Scholar] [CrossRef]
  196. Wang, Y.; Zhu, J.; Zou, N.; Zhang, L.; Wang, Y.; Zhang, M.; Wang, C.; Yang, L. Pathogenesis from the Microbial-Gut-Brain Axis in White Matter Injury in Preterm Infants: A Review. Front. Integr. Neurosci. 2023, 17, 1051689. [Google Scholar] [CrossRef]
  197. Buffington, S.A.; Di Prisco, G.V.; Auchtung, T.A.; Ajami, N.J.; Petrosino, J.F.; Costa-Mattioli, M. Microbial Reconstitution Reverses Maternal Diet-Induced Social and Synaptic Deficits in Offspring. Cell 2016, 165, 1762–1775. [Google Scholar] [CrossRef]
  198. Poutahidis, T.; Kearney, S.M.; Levkovich, T.; Qi, P.; Varian, B.J.; Lakritz, J.R.; Ibrahim, Y.M.; Chatzigiagkos, A.; Alm, E.J.; Erdman, S.E. Microbial Symbionts Accelerate Wound Healing via the Neuropeptide Hormone Oxytocin. PLoS ONE 2013, 8, e78898. [Google Scholar] [CrossRef] [PubMed]
  199. Sgritta, M.; Dooling, S.W.; Buffington, S.A.; Momin, E.N.; Francis, M.B.; Britton, R.A.; Costa-Mattioli, M. Mechanisms Underlying Microbial-Mediated Changes in Social Behavior in Mouse Models of Autism Spectrum Disorder. Neuron 2019, 101, 246–259.e6. [Google Scholar] [CrossRef]
  200. Ibrahim, Y.M.; Kearney, S.M.; Levkovich, T.; Springer, A.; Mirabal, S.; Poutahidis, T.; Varian, B.J.; Lakritz, J.R.; Alm, E.J.; Erdman, S.E. Maternal Gut Microbes Control Offspring Sex and Survival. J. Probiotics Health 2014, 2, 1. [Google Scholar]
  201. Tyzio, R.; Cossart, R.; Khalilov, I.; Minlebaev, M.; Hübner, C.A.; Represa, A.; Ben-Ari, Y.; Khazipov, R. Maternal Oxytocin Triggers a Transient Inhibitory Switch in GABA Signaling in the Fetal Brain during Delivery. Science 2006, 314, 1788–1792. [Google Scholar] [CrossRef]
  202. Ceanga, M.; Spataru, A.; Zagrean, A.-M. Oxytocin Is Neuroprotective against Oxygen-Glucose Deprivation and Reoxygenation in Immature Hippocampal Cultures. Neurosci. Lett. 2010, 477, 15–18. [Google Scholar] [CrossRef]
  203. Panaitescu, A.M.; Isac, S.; Pavel, B.; Ilie, A.S.; Ceanga, M.; Totan, A.; Zagrean, L.; Peltecu, G.; Zagrean, A.M. Oxytocin Reduces Seizure Burden and Hippocampal Injury in a Rat Model of Perinatal Asphyxia. Acta Endocrinol. 2018, 14, 315–319. [Google Scholar] [CrossRef]
  204. Pâslaru, A.-C.; Călin, A.; Morozan, V.-P.; Stancu, M.; Tofan, L.; Panaitescu, A.M.; Zăgrean, A.-M.; Zăgrean, L.; Moldovan, M. Burst-Suppression EEG Reactivity to Photic Stimulation—A Translational Biomarker in Hypoxic–Ischemic Brain Injury. Biomolecules 2024, 14, 953. [Google Scholar] [CrossRef]
  205. Ionescu, M.I.; Grigoras, I.-F.; Ionescu, R.-B.; Chitimus, D.M.; Haret, R.M.; Ianosi, B.; Ceanga, M.; Zagrean, A.-M. Oxytocin Exhibits Neuroprotective Effects on Hippocampal Cultures under Severe Oxygen-Glucose Deprivation Conditions. Curr. Issues Mol. Biol. 2024, 46, 6223–6236. [Google Scholar] [CrossRef]
  206. Spoljaric, A.; Seja, P.; Spoljaric, I.; Virtanen, M.A.; Lindfors, J.; Uvarov, P.; Summanen, M.; Crow, A.K.; Hsueh, B.; Puskarjov, M.; et al. Vasopressin Excites Interneurons to Suppress Hippocampal Network Activity across a Broad Span of Brain Maturity at Birth. Proc. Natl. Acad. Sci. USA 2017, 114, E10819–E10828. [Google Scholar] [CrossRef] [PubMed]
  207. Chen, J.; Aguilera, G. Vasopressin Protects Hippocampal Neurones in Culture against Nutrient Deprivation or Glutamate-Induced Apoptosis: Vasopressin Is Anti-Apoptotic in Hippocampal Neurone Cultures. J. Neuroendocrinol. 2010, 22, 1072–1081. [Google Scholar] [CrossRef]
  208. Wellmann, S.; Benzing, J.; Cippà, G.; Admaty, D.; Creutzfeldt, R.; Mieth, R.A.; Beinder, E.; Lapaire, O.; Morgenthaler, N.G.; Haagen, U.; et al. High Copeptin Concentrations in Umbilical Cord Blood after Vaginal Delivery and Birth Acidosis. J. Clin. Endocrinol. Metab. 2010, 95, 5091–5096. [Google Scholar] [CrossRef] [PubMed]
  209. Desbonnet, L.; Clarke, G.; Traplin, A.; O’Sullivan, O.; Crispie, F.; Moloney, R.D.; Cotter, P.D.; Dinan, T.G.; Cryan, J.F. Gut Microbiota Depletion from Early Adolescence in Mice: Implications for Brain and Behaviour. Brain Behav. Immun. 2015, 48, 165–173. [Google Scholar]
  210. Stigger, F.; Lovatel, G.; Marques, M.; Bertoldi, K.; Moysés, F.; Elsner, V.; Siqueira, I.R.; Achaval, M.; Marcuzzo, S. Inflammatory Response and Oxidative Stress in Developing Rat Brain and Its Consequences on Motor Behavior Following Maternal Administration of LPS and Perinatal Anoxia. Int. J. Dev. Neurosci. 2013, 31, 820–827. [Google Scholar] [CrossRef] [PubMed]
  211. Eklind, S.; Mallard, C.; Arvidsson, P.; Hagberg, H. Lipopolysaccharide Induces Both a Primary and a Secondary Phase of Sensitization in the Developing Rat Brain. Pediatr. Res. 2005, 58, 112–116. [Google Scholar] [CrossRef]
  212. Osredkar, D.; Thoresen, M.; Maes, E.; Flatebø, T.; Elstad, M.; Sabir, H. Hypothermia Is Not Neuroprotective after Infection-Sensitized Neonatal Hypoxic-Ischemic Brain Injury. Resuscitation 2014, 85, 567–572. [Google Scholar] [CrossRef]
  213. Beckers, K.F.; Flanagan, J.P.; Sones, J.L. Microbiome and Pregnancy: Focus on Microbial Dysbiosis Coupled with Maternal Obesity. Int. J. Obes. 2024, 48, 439–448, Erratum in Int. J. Obes. 2025, 49, 1421. [Google Scholar] [CrossRef] [PubMed]
  214. Kalyan, M.; Tousif, A.H.; Sonali, S.; Vichitra, C.; Sunanda, T.; Praveenraj, S.S.; Ray, B.; Gorantla, V.R.; Rungratanawanich, W.; Mahalakshmi, A.M.; et al. Role of Endogenous Lipopolysaccharides in Neurological Disorders. Cells 2022, 11, 4038. [Google Scholar] [CrossRef]
  215. Banks, W.A.; Gray, A.M.; Erickson, M.A.; Salameh, T.S.; Damodarasamy, M.; Sheibani, N.; Meabon, J.S.; Wing, E.E.; Morofuji, Y.; Cook, D.G.; et al. Lipopolysaccharide-Induced Blood-Brain Barrier Disruption: Roles of Cyclooxygenase, Oxidative Stress, Neuroinflammation, and Elements of the Neurovascular Unit. J. Neuroinflamm. 2015, 12, 223. [Google Scholar] [CrossRef] [PubMed]
  216. Zhao, J.; Bi, W.; Xiao, S.; Lan, X.; Cheng, X.; Zhang, J.; Lu, D.; Wei, W.; Wang, Y.; Li, H.; et al. Neuroinflammation Induced by Lipopolysaccharide Causes Cognitive Impairment in Mice. Sci. Rep. 2019, 9, 5790. [Google Scholar] [CrossRef] [PubMed]
  217. Ostrem, B.E.L.; Domínguez-Iturza, N.; Stogsdill, J.A.; Faits, T.; Kim, K.; Levin, J.Z.; Arlotta, P. Fetal Brain Response to Maternal Inflammation Requires Microglia. Development 2024, 151, dev.202252. [Google Scholar] [CrossRef]
  218. Yeh, H.; Ikezu, T. Transcriptional and Epigenetic Regulation of Microglia in Health and Disease. Trends Mol. Med. 2019, 25, 96–111. [Google Scholar] [CrossRef]
  219. Isac, S.; Panaitescu, A.M.; Iesanu, M.; Grigoras, I.F.; Totan, A.; Udriste, A.; Cucu, N.; Peltecu, G.; Zagrean, L.; Zagrean, A.-M. Maternal High-Fat Diet Modifies the Immature Hippocampus Vulnerability to Perinatal Asphyxia in Rats. Neonatology 2018, 114, 355–361. [Google Scholar] [CrossRef]
  220. Isac, S.; Panaitescu, A.M.; Spataru, A.; Iesanu, M.; Totan, A.; Udriste, A.; Cucu, N.; Peltecu, G.; Zagrean, L.; Zagrean, A.-M. Trans-Resveratrol Enriched Maternal Diet Protects the Immature Hippocampus from Perinatal Asphyxia in Rats. Neurosci. Lett. 2017, 653, 308–313. [Google Scholar] [CrossRef]
  221. Isac, S.; Panaitescu, A.M.; Iesanu, M.I.; Zeca, V.; Cucu, N.; Zagrean, L.; Peltecu, G.; Zagrean, A.-M. Maternal Citicoline-Supplemented Diet Improves the Response of the Immature Hippocampus to Perinatal Asphyxia in Rats. Neonatology 2020, 117, 729–735. [Google Scholar] [CrossRef]
  222. Zhang, W.; Hu, X.; Yang, W.; Gao, Y.; Chen, J. Omega-3 Polyunsaturated Fatty Acid Supplementation Confers Long-Term Neuroprotection against Neonatal Hypoxic-Ischemic Brain Injury through Anti-Inflammatory Actions. Stroke 2010, 41, 2341–2347. [Google Scholar] [CrossRef]
  223. Guo, L.; Chen, Q.; Gao, Y.; Jiang, H.; Zhou, F.; Zhang, F.; Xu, M. CDP-Choline Modulates Cholinergic Signaling and Gut Microbiota to Alleviate DSS-Induced Inflammatory Bowel Disease. Biochem. Pharmacol. 2023, 217, 115845. [Google Scholar] [CrossRef] [PubMed]
  224. Prakash, V.; Bose, C.; Sunilkumar, D.; Cherian, R.M.; Thomas, S.S.; Nair, B.G. Resveratrol as a Promising Nutraceutical: Implications in Gut Microbiota Modulation, Inflammatory Disorders, and Colorectal Cancer. Int. J. Mol. Sci. 2024, 25, 3370. [Google Scholar] [CrossRef] [PubMed]
  225. Robertson, R.C.; Kaliannan, K.; Strain, C.R.; Ross, R.P.; Stanton, C.; Kang, J.X. Maternal Omega-3 Fatty Acids Regulate Offspring Obesity through Persistent Modulation of Gut Microbiota. Microbiome 2018, 6, 95. [Google Scholar] [CrossRef]
  226. Carvalho, A.V.S.; Sanches, E.F.; Ribeiro, R.T.; Durán-Carabali, L.E.; Júnior, O.R.; Muniz, B.D.; Wajner, M.; Wyse, A.T.; Netto, C.A.; Sizonenko, S.V. Maternal Lactoferrin Supplementation Prevents Mitochondrial and Redox Homeostasis Dysfunction, and Improves Antioxidant Defenses through Nrf2 and UCP2 Signaling after Neonatal Hypoxia-Ischemia. Free Radic. Biol. Med. 2025, 231, 68–79. [Google Scholar] [CrossRef]
  227. Zhao, C.; Chen, N.; Ashaolu, T.J. Prebiotic and Modulatory Evidence of Lactoferrin on Gut Health and Function. J. Funct. Foods 2023, 108, 105741. [Google Scholar] [CrossRef]
  228. Coscia, A.; Bardanzellu, F.; Caboni, E.; Fanos, V.; Peroni, D.G. When a Neonate Is Born, so Is a Microbiota. Life 2021, 11, 148. [Google Scholar] [CrossRef] [PubMed]
  229. Suárez-Martínez, C.; Santaella-Pascual, M.; Yagüe-Guirao, G.; Martínez-Graciá, C. Infant Gut Microbiota Colonization: Influence of Prenatal and Postnatal Factors, Focusing on Diet. Front. Microbiol. 2023, 14, 1236254. [Google Scholar] [CrossRef]
  230. Iribarren, I.; Hilario, E.; Álvarez, A.; Alonso-Alconada, D. Neonatal Multiple Organ Failure after Perinatal Asphyxia. An. Pediatr. (Engl. Ed.) 2022, 97, 280.e1–280.e8. [Google Scholar] [CrossRef]
  231. Gamsu, H.R.; Kempley, S.T. Enteral Hypoxia/Ischaemia and Necrotizing Enterocolitis. Semin. Neonatol. 1997, 2, 245–254. [Google Scholar] [CrossRef]
  232. Nikiforou, M.; Willburger, C.; de Jong, A.E.; Kloosterboer, N.; Jellema, R.K.; Ophelders, D.R.M.G.; Steinbusch, H.W.M.; Kramer, B.W.; Wolfs, T. Global Hypoxia-Ischemia Induced Inflammation and Structural Changes in the Preterm Ovine Gut Which Were Not Ameliorated by Mesenchymal Stem Cell Treatment. Mol. Med. 2016, 22, 244–257. [Google Scholar] [CrossRef]
  233. Guzmán-De La Garza, F.J.; Cámara-Lemarroy, C.R.; Ballesteros-Elizondo, R.G.; Alarcón-Galván, G.; Cordero-Pérez, P.; Fernández-Garza, N.E. Ketamine Reduces Intestinal Injury and Inflammatory Cell Infiltration after Ischemia/Reperfusion in Rats. Surg. Today 2010, 40, 1055–1062. [Google Scholar] [CrossRef] [PubMed]
  234. Moreno-Indias, I.; Torres, M.; Sanchez-Alcoholado, L.; Cardona, F.; Almendros, I.; Gozal, D.; Montserrat, J.M.; Queipo-Ortuño, M.I.; Farré, R. Normoxic Recovery Mimicking Treatment of Sleep Apnea Does Not Reverse Intermittent Hypoxia-Induced Bacterial Dysbiosis and Low-Grade Endotoxemia in Mice. Sleep 2016, 39, 1891–1897. [Google Scholar] [CrossRef]
  235. Moreno-Indias, I.; Torres, M.; Montserrat, J.M.; Sanchez-Alcoholado, L.; Cardona, F.; Tinahones, F.J.; Gozal, D.; Poroyko, V.A.; Navajas, D.; Queipo-Ortuño, M.I.; et al. Intermittent Hypoxia Alters Gut Microbiota Diversity in a Mouse Model of Sleep Apnoea. Eur. Respir. J. 2015, 45, 1055–1065. [Google Scholar] [CrossRef]
  236. Chen, G.; Li, F.; Du, J. Change of Gut Microbiome Structure in Preterm Infants with Hypoxic Ischemic Encephalopathy Induced by Apnea. Pediatr. Neonatol. 2023, 64, 455–464. [Google Scholar] [CrossRef] [PubMed]
  237. Lundgren, S.N.; Madan, J.C.; Emond, J.A.; Morrison, H.G.; Christensen, B.C.; Karagas, M.R.; Hoen, A.G. Maternal Diet during Pregnancy Is Related with the Infant Stool Microbiome in a Delivery Mode-Dependent Manner. Microbiome 2018, 6, 109. [Google Scholar] [CrossRef]
  238. Ionescu, M.I.; Zahiu, C.D.M.; Vlad, A.; Galos, F.; Gradisteanu Pircalabioru, G.; Zagrean, A.-M.; O’Mahony, S.M. Nurturing Development: How a Mother’s Nutrition Shapes Offspring’s Brain through the Gut. Nutr. Neurosci. 2025, 28, 50–72. [Google Scholar] [CrossRef]
  239. Nakajima, A.; Kaga, N.; Nakanishi, Y.; Ohno, H.; Miyamoto, J.; Kimura, I.; Hori, S.; Sasaki, T.; Hiramatsu, K.; Okumura, K.; et al. Maternal High Fiber Diet during Pregnancy and Lactation Influences Regulatory T Cell Differentiation in Offspring in Mice. J. Immunol. 2017, 199, 3516–3524. [Google Scholar] [CrossRef]
  240. Nakajima, A.; Habu, S.; Kasai, M.; Okumura, K.; Ishikawa, D.; Shibuya, T.; Kobayashi, O.; Osada, T.; Ohkusa, T.; Watanabe, S.; et al. Impact of Maternal Dietary Gut Microbial Metabolites on an Offspring’s Systemic Immune Response in Mouse Models. Biosci. Microbiota Food Health 2020, 39, 33–38. [Google Scholar] [CrossRef] [PubMed]
  241. Fu, J.; Zheng, Y.; Gao, Y.; Xu, W. Dietary Fiber Intake and Gut Microbiota in Human Health. Microorganisms 2022, 10, 2507. [Google Scholar] [CrossRef]
  242. Strobel, K.M.; Juul, S.E.; Hendrixson, D.T. Maternal Nutritional Status and the Microbiome across the Pregnancy and the Post-Partum Period. Microorganisms 2023, 11, 1569. [Google Scholar] [CrossRef]
  243. Miller, C.B.; Benny, P.; Riel, J.; Boushey, C.; Perez, R.; Khadka, V.; Qin, Y.; Maunakea, A.K.; Lee, M.-J. Adherence to Mediterranean Diet Impacts Gastrointestinal Microbial Diversity throughout Pregnancy. BMC Pregnancy Childbirth 2021, 21, 558. [Google Scholar] [CrossRef]
  244. Crovetto, F.; Nakaki, A.; Arranz, A.; Borras, R.; Vellvé, K.; Paules, C.; Boutet, M.L.; Castro-Barquero, S.; Freitas, T.; Casas, R.; et al. Effect of a Mediterranean Diet or Mindfulness-Based Stress Reduction during Pregnancy on Child Neurodevelopment: A Prespecified Analysis of the IMPACT BCN Randomized Clinical Trial. JAMA Netw. Open 2023, 6, e2330255. [Google Scholar] [CrossRef] [PubMed]
  245. Duda-Chodak, A.; Tarko, T.; Satora, P.; Sroka, P. Interaction of Dietary Compounds, Especially Polyphenols, with the Intestinal Microbiota: A Review. Eur. J. Nutr. 2015, 54, 325–341. [Google Scholar] [CrossRef]
  246. Roumes, H.; Sanchez, S.; Benkhaled, I.; Fernandez, V.; Goudeneche, P.; Perrin, F.; Pellerin, L.; Guillard, J.; Bouzier-Sore, A.-K. Neuroprotective Effect of Eco-Sustainably Extracted Grape Polyphenols in Neonatal Hypoxia-Ischemia. Nutrients 2022, 14, 773. [Google Scholar] [CrossRef]
  247. Pontes, P.B.; Toscano, A.E.; Lacerda, D.C.; da Silva Araújo, E.R.; da Costa, P.C.T.; Alves, S.M.; de Brito Alves, J.L.; Manhães-de-Castro, R. Effectiveness of Polyphenols on Perinatal Brain Damage: A Systematic Review of Preclinical Studies. Foods 2023, 12, 2278. [Google Scholar] [CrossRef] [PubMed]
  248. Loren, D.J.; Seeram, N.P.; Schulman, R.N.; Holtzman, D.M. Maternal Dietary Supplementation with Pomegranate Juice Is Neuroprotective in an Animal Model of Neonatal Hypoxic-Ischemic Brain Injury. Pediatr. Res. 2005, 57, 858–864. [Google Scholar] [CrossRef] [PubMed]
  249. Dumont, U.; Sanchez, S.; Olivier, B.; Chateil, J.-F.; Deffieux, D.; Quideau, S.; Pellerin, L.; Beauvieux, M.-C.; Bouzier-Sore, A.-K.; Roumes, H. Maternal Alcoholism and Neonatal Hypoxia-Ischemia: Neuroprotection by Stilbenoid Polyphenols. Brain Res. 2020, 1738, 146798. [Google Scholar] [CrossRef]
  250. Ray, S.K.; Mukherjee, S. Evolving Interplay between Dietary Polyphenols and Gut Microbiota-an Emerging Importance in Healthcare. Front. Nutr. 2021, 8, 634944. [Google Scholar] [CrossRef]
  251. Gowd, V.; Karim, N.; Shishir, M.R.I.; Xie, L.; Chen, W. Dietary Polyphenols to Combat the Metabolic Diseases via Altering Gut Microbiota. Trends Food Sci. Technol. 2019, 93, 81–93. [Google Scholar] [CrossRef]
  252. Wang, X.; Qi, Y.; Zheng, H. Dietary Polyphenol, Gut Microbiota, and Health Benefits. Antioxidants 2022, 11, 1212. [Google Scholar] [CrossRef]
  253. Leng, P.; Wang, Y.; Xie, M. Ellagic Acid and Gut Microbiota: Interactions, and Implications for Health. Food Sci. Nutr. 2025, 13, e70133. [Google Scholar] [CrossRef]
  254. Wang, P.; Wang, R.; Zhao, W.; Zhao, Y.; Wang, D.; Zhao, S.; Ge, Z.; Ma, Y.; Zhao, X. Gut Microbiota-Derived 4-Hydroxyphenylacetic Acid from Resveratrol Supplementation Prevents Obesity through SIRT1 Signaling Activation. Gut Microbes 2025, 17, 2446391. [Google Scholar] [CrossRef] [PubMed]
  255. Suganuma, H.; Arai, Y.; Kitamura, Y.; Hayashi, M.; Okumura, A.; Shimizu, T. Maternal Docosahexaenoic Acid-Enriched Diet Prevents Neonatal Brain Injury: Effect of Docosahexaenoic Acid Diet. Neuropathology 2010, 30, 597–605. [Google Scholar] [CrossRef] [PubMed]
  256. Middleton, P.; Gomersall, J.C.; Gould, J.F.; Shepherd, E.; Olsen, S.F.; Makrides, M. Omega-3 Fatty Acid Addition during Pregnancy. Cochrane Libr. 2018, 11, CD003402. [Google Scholar] [CrossRef]
  257. Costantini, L.; Molinari, R.; Farinon, B.; Merendino, N. Impact of Omega-3 Fatty Acids on the Gut Microbiota. Int. J. Mol. Sci. 2017, 18, 2645. [Google Scholar] [CrossRef]
  258. Wang, C.; Wei, S.; Liu, B.; Wang, F.; Lu, Z.; Jin, M.; Wang, Y. Maternal Consumption of a Fermented Diet Protects Offspring against Intestinal Inflammation by Regulating the Gut Microbiota. Gut Microbes 2022, 14, 2057779. [Google Scholar] [CrossRef]
  259. Erçelik, H.C.; Kaya, V. The Effects of Fermented Food Consumption in Pregnancy on Neonatal and Infant Health: An Integrative Review. J. Pediatr. Nurs. 2024, 75, 173–179. [Google Scholar] [CrossRef]
  260. Rezac, S.; Kok, C.R.; Heermann, M.; Hutkins, R. Fermented Foods as a Dietary Source of Live Organisms. Front. Microbiol. 2018, 9, 1785. [Google Scholar] [CrossRef]
  261. Pandiyan, A.; Gurung, M.; Mulakala, B.K.; Ponniah, S.K.; Yeruva, L. The Role of Fermented Foods in Maternal Health during Pregnancy and Infant Health during the First 1,000 Days of Life. Front. Nutr. 2025, 12, 1581723. [Google Scholar] [CrossRef]
  262. 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]
  263. Hogenkamp, A.; Thijssen, S.; van Vlies, N.; Garssen, J. Supplementing Pregnant Mice with a Specific Mixture of Nondigestible Oligosaccharides Reduces Symptoms of Allergic Asthma in Male Offspring. J. Nutr. 2015, 145, 640–646. [Google Scholar] [CrossRef]
  264. Cuinat, C.; Stinson, S.E.; Ward, W.E.; Comelli, E.M. Maternal Intake of Probiotics to Program Offspring Health. Curr. Nutr. Rep. 2022, 11, 537–562. [Google Scholar] [CrossRef]
  265. Musazadeh, V.; Faghfouri, A.H.; Zarezadeh, M.; Pakmehr, A.; Moghaddam, P.T.; Hamedi-Kalajahi, F.; Jahandideh, A.; Ghoreishi, Z. Remarkable Impacts of Probiotics Supplementation in Enhancing of the Antioxidant Status: Results of an Umbrella Meta-Analysis. Front. Nutr. 2023, 10, 1117387, Erratum in Front. Nutr. 2024, 11, 1371746. [Google Scholar] [CrossRef] [PubMed]
  266. Jones, S.E.; Versalovic, J. Probiotic Lactobacillus Reuteri Biofilms Produce Antimicrobial and Anti-Inflammatory Factors. BMC Microbiol. 2009, 9, 35. [Google Scholar] [CrossRef]
  267. Latif, A.; Shehzad, A.; Niazi, S.; Zahid, A.; Ashraf, W.; Iqbal, M.W.; Rehman, A.; Riaz, T.; Aadil, R.M.; Khan, I.M.; et al. Probiotics: Mechanism of Action, Health Benefits and Their Application in Food Industries. Front. Microbiol. 2023, 14, 1216674, Erratum in Front. Microbiol. 2024, 15, 1378225. [Google Scholar] [CrossRef]
  268. Grev, J.; Berg, M.; Soll, R. Maternal Probiotic Supplementation for Prevention of Morbidity and Mortality in Preterm Infants. Cochrane Database Syst. Rev. 2018, 12, CD012519. [Google Scholar] [CrossRef] [PubMed]
  269. Keunen, K.; van Elburg, R.M.; van Bel, F.; Benders, M.J.N.L. Impact of Nutrition on Brain Development and Its Neuroprotective Implications Following Preterm Birth. Pediatr. Res. 2015, 77, 148–155. [Google Scholar] [CrossRef]
  270. Sha, C.; Jin, Z.; Ku, S.Y.; Kogosov, A.S.; Yu, S.; Bergese, S.D.; Hsieh, H. Necrotizing Enterocolitis and Neurodevelopmental Impairments: Microbiome, Gut, and Brain Entanglements. Biomolecules 2024, 14, 1254. [Google Scholar] [CrossRef] [PubMed]
  271. Wang, K.; Kong, X.; Azad, M.A.K.; Zhu, Q.; Xiong, L.; Zheng, Y.; Hu, Z.; Yin, Y.; He, Q. Maternal Probiotic or Synbiotic Supplementation Modulates Jejunal and Colonic Antioxidant Capacity, Mitochondrial Function, and Microbial Abundance in Bama Mini-Piglets. Oxid. Med. Cell. Longev. 2021, 2021, 6618874. [Google Scholar] [CrossRef] [PubMed]
  272. Zhang, L.; Zhou, W.; Zhan, L.; Hou, S.; Zhao, C.; Bi, T.; Lu, X. Fecal Microbiota Transplantation Alters the Susceptibility of Obese Rats to Type 2 Diabetes Mellitus. Aging 2020, 12, 17480–17502. [Google Scholar] [CrossRef] [PubMed]
  273. Vrieze, A.; Van Nood, E.; Holleman, F.; Salojärvi, J.; Kootte, R.S.; Bartelsman, J.F.W.M.; Dallinga-Thie, G.M.; Ackermans, M.T.; Serlie, M.J.; Oozeer, R.; et al. Transfer of Intestinal Microbiota from Lean Donors Increases Insulin Sensitivity in Individuals with Metabolic Syndrome. Gastroenterology 2012, 143, 913–916.e7, Erratum in Gastroenterology 2013, 144, 250. [Google Scholar] [CrossRef]
  274. Proença, I.M.; Allegretti, J.R.; Bernardo, W.M.; de Moura, D.T.H.; Ponte Neto, A.M.; Matsubayashi, C.O.; Flor, M.M.; Kotinda, A.P.S.T.; de Moura, E.G.H. Fecal Microbiota Transplantation Improves Metabolic Syndrome Parameters: Systematic Review with Meta-Analysis Based on Randomized Clinical Trials. Nutr. Res. 2020, 83, 1–14. [Google Scholar] [CrossRef]
  275. Huang, Y.-H.; Tain, Y.-L.; Hsu, C.-N. Maternal Supplementation of Probiotics, Prebiotics or Postbiotics to Prevent Offspring Metabolic Syndrome: The Gap between Preclinical Results and Clinical Translation. Int. J. Mol. Sci. 2022, 23, 10173. [Google Scholar] [CrossRef]
  276. Wang, Z.; Roberts, A.B.; Buffa, J.A.; Levison, B.S.; Zhu, W.; Org, E.; Gu, X.; Huang, Y.; Zamanian-Daryoush, M.; Culley, M.K.; et al. Non-Lethal Inhibition of Gut Microbial Trimethylamine Production for the Treatment of Atherosclerosis. Cell 2015, 163, 1585–1595. [Google Scholar] [CrossRef]
  277. Hsu, C.-N.; Chang-Chien, G.-P.; Lin, S.; Hou, C.-Y.; Tain, Y.-L. Targeting on Gut Microbial Metabolite Trimethylamine-N-Oxide and Short-Chain Fatty Acid to Prevent Maternal High-Fructose-Diet-Induced Developmental Programming of Hypertension in Adult Male Offspring. Mol. Nutr. Food Res. 2019, 63, e1900073. [Google Scholar] [CrossRef]
  278. Milligan, G.; Barki, N.; Tobin, A.B. Chemogenetic Approaches to Explore the Functions of Free Fatty Acid Receptor 2. Trends Pharmacol. Sci. 2021, 42, 191–202. [Google Scholar] [CrossRef] [PubMed]
  279. Zhao, Q.; Chen, T.; Ni, C.; Hu, Y.; Nan, Y.; Lin, W.; Liu, Y.; Zheng, F.; Shi, X.; Lin, Z.; et al. Indole-3-Propionic Acid Attenuates HI-Related Blood-Brain Barrier Injury in Neonatal Rats by Modulating the PXR Signaling Pathway. ACS Chem. Neurosci. 2022, 13, 2897–2912. [Google Scholar] [CrossRef]
  280. Amat, S.; Dahlen, C.R.; Swanson, K.C.; Ward, A.K.; Reynolds, L.P.; Caton, J.S. Bovine Animal Model for Studying the Maternal Microbiome, in Utero Microbial Colonization and Their Role in Offspring Development and Fetal Programming. Front. Microbiol. 2022, 13, 854453. [Google Scholar] [CrossRef] [PubMed]
  281. Daiy, K.; Wiley, K.; Allen, J.; Bailey, M.T.; Dettmer, A.M. Associations among Rearing Environment and the Infant Gut Microbiome with Early-Life Neurodevelopment and Cognitive Development in a Nonhuman Primate Model (Macaca Mulatta). J. Dev. Orig. Health Dis. 2025, 16, e1. [Google Scholar] [CrossRef]
  282. Ma, J.; Prince, A.L.; Bader, D.; Hu, M.; Ganu, R.; Baquero, K.; Blundell, P.; Alan Harris, R.; Frias, A.E.; Grove, K.L.; et al. High-Fat Maternal Diet during Pregnancy Persistently Alters the Offspring Microbiome in a Primate Model. Nat. Commun. 2014, 5, 3889. [Google Scholar] [CrossRef]
  283. Li, M.; Brokaw, A.; Furuta, A.M.; Coler, B.; Obregon-Perko, V.; Chahroudi, A.; Wang, H.-Y.; Permar, S.R.; Hotchkiss, C.E.; Golos, T.G.; et al. Non-Human Primate Models to Investigate Mechanisms of Infection-Associated Fetal and Pediatric Injury, Teratogenesis and Stillbirth. Front. Genet. 2021, 12, 680342. [Google Scholar] [CrossRef] [PubMed]
  284. Collins, B.; Lemanski, E.A.; Wright-Jin, E. The Importance of Including Maternal Immune Activation in Animal Models of Hypoxic-Ischemic Encephalopathy. Biomedicines 2024, 12, 2559. [Google Scholar] [CrossRef]
  285. Koehler, R.C.; Yang, Z.-J.; Lee, J.K.; Martin, L.J. Perinatal Hypoxic-Ischemic Brain Injury in Large Animal Models: Relevance to Human Neonatal Encephalopathy. J. Cereb. Blood Flow Metab. 2018, 38, 2092–2111. [Google Scholar] [CrossRef]
  286. Jacobson Misbe, E.N.; Richards, T.L.; McPherson, R.J.; Burbacher, T.M.; Juul, S.E. Perinatal Asphyxia in a Nonhuman Primate Model. Dev. Neurosci. 2011, 33, 210–221. [Google Scholar] [CrossRef]
  287. Doron, S.; Snydman, D.R. Risk and Safety of Probiotics. Clin. Infect. Dis. 2015, 60 (Suppl. 2), S129–S134. [Google Scholar] [CrossRef] [PubMed]
  288. Bertelli, C.; Pillonel, T.; Torregrossa, A.; Prod’hom, G.; Fischer, C.J.; Greub, G.; Giannoni, E. Bifidobacterium Longum Bacteremia in Preterm Infants Receiving Probiotics. Clin. Infect. Dis. 2015, 60, 924–927. [Google Scholar] [CrossRef] [PubMed]
  289. Pillai, A.; Tan, J.; Paquette, V.; Panczuk, J. Does Probiotic Bacteremia in Premature Infants Impact Clinically Relevant Outcomes? A Case Report and Updated Review of Literature. Clin. Nutr. ESPEN 2020, 39, 255–259. [Google Scholar] [CrossRef]
  290. Roy, U.; Jessani, L.G.; Rudramurthy, S.M.; Gopalakrishnan, R.; Dutta, S.; Chakravarty, C.; Jillwin, J.; Chakrabarti, A. Seven Cases of Saccharomyces Fungaemia Related to Use of Probiotics. Mycoses 2017, 60, 375–380. [Google Scholar] [CrossRef]
  291. Farella, I.; Fortunato, M.; Martinelli, D.; De Carlo, C.; Sparapano, E.; Stolfa, S.; Romanelli, F.; De Laurentiis, V.; Martinotti, S.; Capozzi, L.; et al. Lactobacillus Rhamnosus Sepsis in a Preterm Infant Following Probiotic Administration: Challenges in Diagnosis. Microorganisms 2025, 13, 265. [Google Scholar] [CrossRef] [PubMed]
  292. Underwood, M.A.; Umberger, E.; Patel, R.M. Safety and Efficacy of Probiotic Administration to Preterm Infants: Ten Common Questions. Pediatr. Res. 2020, 88, 48–55. [Google Scholar] [CrossRef] [PubMed]
  293. Available online: https://www.fda.gov/media/172606/download?attachment (accessed on 3 May 2025).
  294. O’Doherty, K.C.; Virani, A.; Wilcox, E.S. The Human Microbiome and Public Health: Social and Ethical Considerations. Am. J. Public Health 2016, 106, 414–420. [Google Scholar] [CrossRef] [PubMed]
  295. Chan, N.H.; Hawkins, C.C.; Rodrigues, B.V.; Cornet, M.-C.; Gonzalez, F.F.; Wu, Y.W. Neuroprotection for Neonatal Hypoxic-Ischemic Encephalopathy: A Review of Novel Therapies Evaluated in Clinical Studies. Dev. Med. Child Neurol. 2025, 67, 591–599. [Google Scholar] [CrossRef]
Figure 1. Influence of the maternal gut microbiome on the metabolic function, immune, and placental health. Left: A balanced maternal gut microbiome (eubiosis) is associated with improved glucose tolerance, reduced insulin levels, and lower markers of gestational diabetes. It is characterized by low systemic inflammation, with decreased pro-inflammatory cytokines and controlled populations of granulocytes and T-regulatory cells, as well as optimal placental development. Right: In contrast, dysbiosis is associated with impaired metabolic function (gestational diabetes, insulin resistance, altered oestrogen metabolism), heightened immune activation (elevated levels of M1 macrophages, Tumour Necrosis Factor alpha (TNF-α), Interleukin-1 beta (IL-1β), Interleukin-6 (IL-6), Interleukin-17 (IL-17); reduced levels of Interleukin-10 (IL-10); maternal immune activation), and compromised placental function (lower placental weight and volume; increased risk of preeclampsia and hypertension). Created in BioRender. Racovita, A. (2025) https://BioRender.com/75vubaj.
Figure 1. Influence of the maternal gut microbiome on the metabolic function, immune, and placental health. Left: A balanced maternal gut microbiome (eubiosis) is associated with improved glucose tolerance, reduced insulin levels, and lower markers of gestational diabetes. It is characterized by low systemic inflammation, with decreased pro-inflammatory cytokines and controlled populations of granulocytes and T-regulatory cells, as well as optimal placental development. Right: In contrast, dysbiosis is associated with impaired metabolic function (gestational diabetes, insulin resistance, altered oestrogen metabolism), heightened immune activation (elevated levels of M1 macrophages, Tumour Necrosis Factor alpha (TNF-α), Interleukin-1 beta (IL-1β), Interleukin-6 (IL-6), Interleukin-17 (IL-17); reduced levels of Interleukin-10 (IL-10); maternal immune activation), and compromised placental function (lower placental weight and volume; increased risk of preeclampsia and hypertension). Created in BioRender. Racovita, A. (2025) https://BioRender.com/75vubaj.
Antioxidants 14 01134 g001
Figure 2. Impact of the maternal gut microbiome-derived signals on fetal immune and neurodevelopment. Bioactive microbial metabolites, namely short-chain fatty acids (SCFAs), aryl hydrocarbon receptor ligands, bile salts, trimethylated compounds, and peptidoglycan fragments, together with IgG-bound microbial antigens, microbiota-derived extracellular vesicles, and trace amounts of bacteria, cross the placental barrier and modulate fetal neuroimmune development. These signals regulate the balance between pro-inflammatory cytokines (TNF-α, IL-17, IL-1β, IL-6) and anti-inflammatory cytokines (IL-10, TGF-β), promoting microglial maturation, synaptogenesis, axonogenesis, and myelination. Created in BioRender. Racovita, A. (2025) https://BioRender.com/ye58bcx.
Figure 2. Impact of the maternal gut microbiome-derived signals on fetal immune and neurodevelopment. Bioactive microbial metabolites, namely short-chain fatty acids (SCFAs), aryl hydrocarbon receptor ligands, bile salts, trimethylated compounds, and peptidoglycan fragments, together with IgG-bound microbial antigens, microbiota-derived extracellular vesicles, and trace amounts of bacteria, cross the placental barrier and modulate fetal neuroimmune development. These signals regulate the balance between pro-inflammatory cytokines (TNF-α, IL-17, IL-1β, IL-6) and anti-inflammatory cytokines (IL-10, TGF-β), promoting microglial maturation, synaptogenesis, axonogenesis, and myelination. Created in BioRender. Racovita, A. (2025) https://BioRender.com/ye58bcx.
Antioxidants 14 01134 g002
Figure 3. Neuroprotective mechanisms mediated by the maternal gut microbiome. Maternal gut microbiome-derived metabolites confer neuroprotection against perinatal hypoxic-ischemic injury through four integrated pathways spanning the maternal gut-placental-fetal brain axis. (1) Anti-inflammatory mechanisms: Short-chain fatty acids (SCFAs) inhibit histone deacetylase (HDAC) activity, promote M2 microglial polarization, reduce neuroinflammation, and strengthen gut-placental-blood-brain barriers. SCFAs expand regulatory T cells (Tregs) by priming thymic epithelial cells (TECs) to select Treg precursor thymocytes, dendritic cells to promote differentiation into Tregs, and, consequently, suppress inflammatory cascades. Tryptophan-derived indole metabolites activate aryl hydrocarbon receptor (AhR), suppressing NF-κB p65 phosphorylation in microglia and decreasing IL-1β and TNF-α expression; (2) Antioxidant defense: eubiosis prevents ROS leaks into the circulation and contributes to upregulation of junction proteins. At the placental level, microbiota-derived metabolites directly neutralise ROS and lower oxidative stress. Through FFAR activation and Nrf2 signaling, the antioxidant capacity of the fetal BBB is increased, leading to better integrity and protection against ROS activity. In neurons, by HDAC inhibition, SCFAs induce Nrf2 activation that enhances antioxidant enzyme activity and lowers ferroptosis. AhR ligands also directly buffer some of the ROS species. Consecutive to NF-κB inhibition, M2 microglia produce less ROS, mitigating the oxidative stress; (3) Neural repair and neurogenesis: Microbiome metabolites maintain brain-derived neurotrophic factor (BDNF) expression through H3 crotonylation via HDAC inhibition. This promotes neurogenesis, oligodendrocyte maturation, and protective glial responses that enhance tissue preservation, myelination, and adequate pruning during the recovery phase; (4) Oxytocin system modulation: Specific maternal gut bacteria (particularly Lactobacillus reuteri) enhance hypothalamic oxytocin expression. During hypoxic stress, oxytocin facilitates the critical gamma-aminobutyric acid (GABA) switch from excitatory to inhibitory signaling, reducing excitotoxicity and providing direct neuroprotection against neuronal loss. Created in BioRender. Racovita, A. (2025) https://BioRender.com/wc5q5uf.
Figure 3. Neuroprotective mechanisms mediated by the maternal gut microbiome. Maternal gut microbiome-derived metabolites confer neuroprotection against perinatal hypoxic-ischemic injury through four integrated pathways spanning the maternal gut-placental-fetal brain axis. (1) Anti-inflammatory mechanisms: Short-chain fatty acids (SCFAs) inhibit histone deacetylase (HDAC) activity, promote M2 microglial polarization, reduce neuroinflammation, and strengthen gut-placental-blood-brain barriers. SCFAs expand regulatory T cells (Tregs) by priming thymic epithelial cells (TECs) to select Treg precursor thymocytes, dendritic cells to promote differentiation into Tregs, and, consequently, suppress inflammatory cascades. Tryptophan-derived indole metabolites activate aryl hydrocarbon receptor (AhR), suppressing NF-κB p65 phosphorylation in microglia and decreasing IL-1β and TNF-α expression; (2) Antioxidant defense: eubiosis prevents ROS leaks into the circulation and contributes to upregulation of junction proteins. At the placental level, microbiota-derived metabolites directly neutralise ROS and lower oxidative stress. Through FFAR activation and Nrf2 signaling, the antioxidant capacity of the fetal BBB is increased, leading to better integrity and protection against ROS activity. In neurons, by HDAC inhibition, SCFAs induce Nrf2 activation that enhances antioxidant enzyme activity and lowers ferroptosis. AhR ligands also directly buffer some of the ROS species. Consecutive to NF-κB inhibition, M2 microglia produce less ROS, mitigating the oxidative stress; (3) Neural repair and neurogenesis: Microbiome metabolites maintain brain-derived neurotrophic factor (BDNF) expression through H3 crotonylation via HDAC inhibition. This promotes neurogenesis, oligodendrocyte maturation, and protective glial responses that enhance tissue preservation, myelination, and adequate pruning during the recovery phase; (4) Oxytocin system modulation: Specific maternal gut bacteria (particularly Lactobacillus reuteri) enhance hypothalamic oxytocin expression. During hypoxic stress, oxytocin facilitates the critical gamma-aminobutyric acid (GABA) switch from excitatory to inhibitory signaling, reducing excitotoxicity and providing direct neuroprotection against neuronal loss. Created in BioRender. Racovita, A. (2025) https://BioRender.com/wc5q5uf.
Antioxidants 14 01134 g003
Table 1. Factors Influencing Maternal Microbiome Composition During Pregnancy.
Table 1. Factors Influencing Maternal Microbiome Composition During Pregnancy.
FactorEffect on MicrobiomeImpact TypeReferences
High-fat diet↓ Bacteroidetes, ↑ Firmicutes, ↑ pro-inflammatory taxaDysbiotic[64,65]
High-fiber diet↑ short-chain fatty acids (SCFA)-producers, ↑ Bifidobacterium, ↑ diversityBeneficial[66,67,68]
Alcohol consumption↓ beneficial bacteria, altered metabolic pathwaysDysbiotic[69]
Antibiotic use↓ diversity, loss of key commensals, ↑ resistant strainsDysbiotic[63,70]
Probiotic supplementation↑ specific beneficial strains, ↑ barrier functionBeneficial[71]
Iron supplementationAltered Bacteroidetes to Firmicutes ratio, ↑ pathobiontsVariable[72]
Elevated body mass index (BMI)/Obesity↓ diversity, ↑ Firmicutes, altered SCFA productionDysbiotic[73,74]
Insulin resistanceAltered glucose metabolism pathways, ↓ butyrate producersDysbiotic[75]
Pre-existing gastrointestinalconditions: inflammatory bowel disease (IBD)↓ diversity, ↑ Proteobacteria, ↑ inflammatory markersDysbiotic[76]
Smoking↓ beneficial anaerobes, ↑ opportunistic pathogensDysbiotic[77,78]
Regular exercise↑ diversity, ↑ SCFA production, ↑ AkkermansiaBeneficial[79,80]
Psychological stress↑ Proteobacteria, ↓ Lactobacillus, altered barrier functionDysbiotic[13,81]
Pregnancy progression↑ Proteobacteria (with pregnancy progression), ↓ diversity (adaptive)Adaptive[51,52,82]
Previous pregnanciesEnhanced microbial stability, faster adaptationBeneficial[83]
Maternal ageAge-dependent diversity changes, altered metabolic capacityVariable[51]
Residential environmentUrban vs. rural differences in diversity and compositionVariable[51]
↑ increased/higher levels; ↓ decreased/lower levels.
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

Morozan, V.-P.; Ionescu, M.I.; Zahiu, C.M.D.; Catrina, A.M.; Racoviță, A.; Chirilă, A.-T.; Dogaru, I.-A.; Ciotei, C.; Pircalabioru, G.G.; Zăgrean, A.-M. Does the Maternal Gut Microbiome Influence the Outcome of Perinatal Asphyxia? Antioxidants 2025, 14, 1134. https://doi.org/10.3390/antiox14091134

AMA Style

Morozan V-P, Ionescu MI, Zahiu CMD, Catrina AM, Racoviță A, Chirilă A-T, Dogaru I-A, Ciotei C, Pircalabioru GG, Zăgrean A-M. Does the Maternal Gut Microbiome Influence the Outcome of Perinatal Asphyxia? Antioxidants. 2025; 14(9):1134. https://doi.org/10.3390/antiox14091134

Chicago/Turabian Style

Morozan, Vlad-Petru, Mara I. Ionescu, Carmen M. D. Zahiu, Ana Maria Catrina, Andreea Racoviță, Ana-Teodora Chirilă, Ioana-Alexandra Dogaru, Cristian Ciotei, Gratiela Gradisteanu Pircalabioru, and Ana-Maria Zăgrean. 2025. "Does the Maternal Gut Microbiome Influence the Outcome of Perinatal Asphyxia?" Antioxidants 14, no. 9: 1134. https://doi.org/10.3390/antiox14091134

APA Style

Morozan, V.-P., Ionescu, M. I., Zahiu, C. M. D., Catrina, A. M., Racoviță, A., Chirilă, A.-T., Dogaru, I.-A., Ciotei, C., Pircalabioru, G. G., & Zăgrean, A.-M. (2025). Does the Maternal Gut Microbiome Influence the Outcome of Perinatal Asphyxia? Antioxidants, 14(9), 1134. https://doi.org/10.3390/antiox14091134

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