3,544
Views
0
CrossRef citations to date
0
Altmetric
Review

The contribution of age-related changes in the gut-brain axis to neurological disorders

, , & ORCID Icon
Article: 2302801 | Received 06 Aug 2023, Accepted 04 Jan 2024, Published online: 18 Jan 2024

ABSTRACT

Trillions of microbes live symbiotically in the host, specifically in mucosal tissues such as the gut. Recent advances in metagenomics and metabolomics have revealed that the gut microbiota plays a critical role in the regulation of host immunity and metabolism, communicating through bidirectional interactions in the microbiota-gut-brain axis (MGBA). The gut microbiota regulates both gut and systemic immunity and contributes to the neurodevelopment and behaviors of the host. With aging, the composition of the microbiota changes, and emerging studies have linked these shifts in microbial populations to age-related neurological diseases (NDs). Preclinical studies have demonstrated that gut microbiota-targeted therapies can improve behavioral outcomes in the host by modulating microbial, metabolomic, and immunological profiles. In this review, we discuss the pathways of brain-to-gut or gut-to-brain signaling and summarize the role of gut microbiota and microbial metabolites across the lifespan and in disease. We highlight recent studies investigating 1) microbial changes with aging; 2) how aging of the maternal microbiome can affect offspring health; and 3) the contribution of the microbiome to both chronic age-related diseases (e.g., Parkinson’s disease, Alzheimer’s disease and cerebral amyloidosis), and acute brain injury, including ischemic stroke and traumatic brain injury.

This article is part of the following collections:
Gut microbiota and aging

1. Introduction

Over the past decade, research has identified a novel role of the gut microbiome in the bidirectional communication between the gut and the brain, termed the microbiota-gut-brain-axis (MGBA).Citation1 Disruption in the balance of gut microbial communities (often referred to as “dysbiosis”) has implicated various pathways along this axis that contribute to the progression of neurodegenerative diseases, such as Alzheimer’s disease (AD) and Parkinson’s disease (PD).Citation1 The MGBA also contributes to outcomes after acute neurological injury, such as stroke and traumatic brain injury (TBI).Citation1 Many of these NDs are diseases that increase in prevalence with aging. Emerging studies have shown that the process of aging directs and changes the composition of the microbiome,Citation2 which leads to chronic systemic inflammation, or “inflammaging”.Citation3 Inflammaging is characterized by an increased level of circulating pro-inflammatory cytokines and breakdown of the barrier integrity of host tissues including in the brain (e.g., blood-brain barrier) and gut (e.g., intestinal epithelium), leading to antigen translocation into the host and heightened systemic inflammation.Citation2,Citation4 Inflammaging may be caused by a variety of processes that accompany aging, such as oxidative stress or cellular senescence, many of which are mediated by recently identified pathways in the MGBA.Citation5 In experimental studies, specific microbially-derived metabolites are altered both with aging and in age-related diseases, which has now been confirmed in clinical populations as well.Citation6 Our understanding of the mechanisms through which these metabolites change host homeostasis is emerging, but detailed mechanistic studies are required if we hope to harness the potential of the microbiome to enhance health.

2. The microbiome as a mediator in gut-to-brain signaling

The human microbiome is comprised of trillions of microorganisms, including bacteria, archaea, viruses, and eukaryotes, which exert a profound effect on all physiological and pathological processes occurring in the host.Citation1,Citation7 Microbial communities vary across distinct body sites and organs, but those residing in the gastrointestinal (GI) tract have, to date, attracted the greatest attention in biomedical research, including gerontology and neurological research.

Multiple large-scale studies, such as the NIH-funded Human Microbiome Project (HMP), have provided extensive information regarding the genetic sequences of most microorganisms residing in the human gut. These datasets revealed that the phyla Bacteroidetes and Firmicutes account for 90% of the gut microbiota, but many others such as Proteobacteria, Actinobacteria, Fusobacteria, Spirochaetes, Verrucomicrobia and Lentisphaerae are also present. The identification of a core microbiome, despite being critical for our understanding of the microbial contribution to health and disease, is challenging due the huge variability in gut microbiome configurations across individuals and within the same subject across the lifespan.Citation8 More recently, multi-omics approaches such as metagenomics and metabolomic sequencing has dramatically expanded our knowledge of the functional role of gut microorganisms in host health and pathological processes.Citation9 The interplay between the gut microbiota and the host is regulated by a complex network of metabolic, immune, and neuroendocrine interactions. When physiological changes within the gut microbial community evolve into a detrimental state, or so-called “dysbiosis”,Citation10 significant alterations in the pool of the metabolites produced and released by these microorganisms occur, with important repercussions for host physiology.

A growing body of evidence has implicated the gut microbiome in the progressive accumulation of molecular and cellular alterations observed with senescence. These changes ultimately increase the susceptibility to chronic diseases in older populations. Aging-associated processes include the progressive accumulation of senescent cells (SCs), which are identified by definitive cell cycle arrest, abnormal mitochondrial reactive oxygen species (ROS) production, metabolic shifts, and the production of senescence-associated secretory phenotype factors (as reviewed in refCitation11). Release of these factors triggers proinflammatory responses from different immune cells that participate in the physiological changes seen with aging. These detrimental changes are potentially reversible, as shown by recent studies in mice that target these SCs, leading to reductions in systemic inflammation, TNFα/NF-κB signaling, and senescence-associated signatures in aged mice.Citation12 Intriguingly, emerging evidence suggests that the gut microbiome may play an important role in modulating the effects of SC. A very recent study using germ-free (GF) mice (raised in total absence of microbial colonization) showed that the aging microbiome was responsible for accumulation of senescent markers in ileal B cells, which in turn further altered gut microbiome composition.Citation13

3. The gut-brain axis: a bidirectional communication

Communication between the brain and microbiota is bidirectional and can occur through multiple pathways. These include neural connections between the brain and the gut through the vagus nerve, hormonal and immune pathways, and metabolite signaling.Citation14 There is a high degree of intercommunication between the gut and the peripheral nervous system (PNS) which participate in the immunological and hormonal responses to gut bacterial biochemical processes. Gut microbial signals can be “sensed” via vagal and spinal neurons, integrated in the brainstem and hypothalamus, and ultimately influence efferent signals to peripheral organs.Citation14 Several recent studies manipulating the gut microbiota composition have illustrated the importance of the interaction between gut microbes and the PNS (), via efferent/afferent pathways, in regulating host physiology, as discussed below.

Figure 1. The bidirectional communication between the gut and central nervous system is regulated by multiple pathways(BBB = blood brain barrier, CRH = corticotropin-releasing hormone, ACTH = adrenocorticotropic hormone, HPA = hypothalamic-pituitary-adrenal, GABA = gamma-aminobutyric acid, SCFA = short chain fatty acid, 5-HT = 5-hydroxytryptamine). Created in Biorender.com.

Figure 1. The bidirectional communication between the gut and central nervous system is regulated by multiple pathways(BBB = blood brain barrier, CRH = corticotropin-releasing hormone, ACTH = adrenocorticotropic hormone, HPA = hypothalamic-pituitary-adrenal, GABA = gamma-aminobutyric acid, SCFA = short chain fatty acid, 5-HT = 5-hydroxytryptamine). Created in Biorender.com.

3.1 Neuroanatomic pathways: vagal mechanisms/vagus nerve

The two-way neuroanatomic communication between the gut and brain occurs through afferent or efferent signaling along two main directional pathways: (1) the autonomic nervous system (ANS) including the vagus nerve (VN) and the enteric nervous system (ENS). The VN is the tenth cranial nerve and one of the main components of the parasympathetic nervous system, which forms the ANS together with the sympathetic nervous system. The ANS has a primary role in regulating multiple physiological processes, including heart rate, immune response, and digestion.Citation15 Signals from the gut are conveyed to the central nervous system (CNS) through the ANS in a bottom-up manner, and responses from the CNS are then sent to the gut by the ANS in a top-down manner.Citation16 In this context, the VN represents the most direct connection between the gut and the brain, participating in both bottom-up and top-down signaling via both afferent (sensory) and efferent (motor) nerves.Citation17 Vagal terminals reach the gut in the mucosal layer, the smooth muscle layer, and synapse with enteroendocrine cells (EECs), without direct contact with the gut microbiota in the lumen. A recent study by Bohórquez and colleaguesCitation18 showed the presence of a specialized group of EECs, defined as neuropods, which provide a direct connection between the gut lumen and brain stem by synapsing with the VN through glutamatergic transmission. The neuropod-vagal terminal circuit is activated in response to sugar, thereby transducing fast sensory input from the gut lumen. Additionally, vagal fibers, which express receptors for multiple metabolites produced by the microbiome, can sense changes in microbial populations.Citation19 Similarly, EECs express receptors for microbial metabolites such as short-chain fatty acids (SCFAs), indoles, bile acids, and lipopolysaccharide (LPS).Citation20 Both human and animal studies have highlighted the crucial role of the VN in regulating brain activity. Either partial or total vagotomy in rodents led to changes in brain circuits and behavioral functions implicated in various neuropsychiatric disorders,Citation21 such as anxiety, fear-related phenotypes,Citation22 learning and memory,Citation23 locomotionCitation24 and sensorimotor gating.Citation25 Similarly, direct stimulation of the VN modulates stress-induced depressive phenotypes via regulation of serotonergic circuitry in the hippocampus,Citation26 in anxiety and post-traumatic stress disordersCitation27,Citation28 and the reward system involved in affective disorders through effects on dopaminergic circuitry in the substantia nigra.Citation29 Recent studies have identified specific microorganisms residing in the gut that can modulate brain function via vagal fibers. For example, administration of Lactobacillus rhamnosus was effective in decreasing both depressive- and anxiety-like phenotypes in mice, an effect that was mediated by an increased firing rate of VN terminals.Citation11 Similarly, supplementation with Campylobacter jejuni increased VN c-Fos expression in the vagal ganglia, leading to activation of neurons in the solitary nucleus of the brainstem.Citation30 Additionally, another study showed that C. jejuni treatment in mice promoted anxiety-related behavior.Citation31 The interactions between the vagus nerve, the gut epithelium and the ENS are summarized in .

Figure 2. Vagus nerve interactions with the gut epithelium and the ENS. Created in Biorender.com.

Figure 2. Vagus nerve interactions with the gut epithelium and the ENS. Created in Biorender.com.

3.2 Neuroanatomic pathways: the enteric nervous system

The ENS, a part of the PNS, is at a critical intersection between the host and the gut microbiome. Anatomically, the ENS is organized as a web of motor, sensor, and interneurons that are embedded in the inner and outer layers of the muscularis externa and in the submucosa of the digestive system. By integrating peripheral sensory information with input from the ANS, the ENS controls the muscular and secretory functions of the GI tract, including peristalsis and the production and release of enzymes and hormones, such as gastrin and secretin.Citation28 Sensory neurons in the ENS form synapses with both enteric motor neurons and vagal fibers, and express receptors for multiple microbial metabolites and components, including SCFAsCitation32 and LPS through toll-like receptor 4 (TLR4).Citation33 Studies in GF mice have shed light on the influence of the microbiome in controlling the electrophysiology of ENS neurons. Maturation of the ENS begins during postnatal development as microbial strains colonize the infant gut through the activation of pattern recognition receptors, such as TLRs, on ENS terminals that bind to microbial products, including LPS.Citation34 Microbial reconstitution of GF mice induced upregulation of the expression of 5-hydroxytryptamine (5-HT) and its receptors in enteric neurons.Citation34,Citation35 Antibiotic-mediated depletion of gut bacteria in mice altered both the morpho-functional structure and the neurochemistry of the ENS, including the loss of neurons in the myenteric plexus, increased TLR2 expression in neuromuscular and mucosal layers in the ileum, and a reduction in glial cells.Citation36 Different microorganisms may exert different effects on ENS neuronal activity through distinct mechanisms.Citation37 Recent studies suggest that changes in the ENS might be a driving factor in determining dysbiosis of the gut microbiome through the regulation of intestinal transit, gut barrier permeability, and luminal pH.Citation38

3.3 Systemic and mucosal immune regulation: immunological pathways

Studies in both animal models and humans have shown that the gut microbiome is essential for the regulation of the host immune system. Microbial colonization of the host’s mucosa during the early postnatal period profoundly shapes the development and maturation of the host immune system.Citation39 Beyond infancy, the gut microbiome is intricately involved in maintaining immune homeostasis through complex interactions with the mucosal immune system. Immune cells also play important roles in the gutCitation40 including: (1) tolerance toward a multitude of microorganisms in the normal, healthy gut ecology; (2) surveillance of potential pathogenic strains; and (3) inhibition of commensal overgrowth and prevention of translocation of bacteria from the intestinal lumen into the host, a process that requires the integrity of the intestinal mucosal barrier.Citation41

Crosstalk between the gut microbiota and host immune system regulates the production and release of neurotransmitters and neuropeptides, cytokines, and other signaling mediators that influence brain function by multiple mechanisms, including actions on vagal and spinal afferent fibers.Citation42 In this context, one of the mechanisms of regulation of the MGBA involves the maturation and function of microglia, the resident immune cells of the brain.Citation42,Citation43 This process starts during fetal and early postnatal development and is mediated by the composition of the maternal gut microbiome.Citation44

During prenatal development, maternal gut microbial communities contribute to fetal microglia programming, which in turn, directly affect the formation of cortical cytoarchitecture and neural circuits.Citation45 In GF mice, substantial alterations in gene expression occur in fetal microglial at mid and late gestation. Microglia programming continued postnatally (P20) leading to increased microglial density (Iba1+ cells) in the somatosensory neocortex of GF females compared to SPF females, which was not seen in males.Citation44 The impaired microglia maturation seen in GF mice was rescued by early-life colonization with a complex microbiome.Citation46 Migration of CD4 T cells into the brain around the time of birth is also critical for microglial maturation, and is potentially regulated by the gut microbiome.Citation47 Therefore, further studies are needed to investigate how alterations in the gut microbiome composition and immune cell populations in early development could lead to increased predisposition for neurological disorders later in life.

Microglia not only modulate inflammatory processes in the brain, but are also involved in synaptic plasticity and remodeling, maturation of the CNS, and debris and aggregate clearance.Citation48 Microglia activation and function can be mediated by several factors produced from host-bacteria interactions,Citation49 including cytokines, tryptophan metabolites, bacterial-derived cell components (e.g., peptidoglycans and LPS)Citation50 and bacterial-derived metabolites (SCFAs).Citation51 These gut-derived signals can reach the brain through the bloodstream, through the VNCitation52 and potentially through the newly discovered meningeal lymphatic system through actions on γδ T cells.Citation53 Several studies have proposed epigenetic regulation and chromatin remodeling as a potential mechanism for microbiota-dependent modulation of microglia function.Citation43,Citation54 A recent investigation in the context of neurodegenerative processesCitation55 identified SCFAs, specifically acetate, as a crucial regulator of microglial metabolic function through histone methylation modification on genes related to microglial proliferation, morphology, activation, and metabolism. Epigenetic regulation of immune function via bacterial metabolites has also been observed in the gut immune system. For example, SCFAs, particularly propionate, decreased IL-17-producing γδ T cells in humans (e.g., peripheral blood mononuclear cells) and in mice (e.g., intestinal lamina propria) in a histone deacetylase-dependent manner.Citation56 Further research is warranted to unravel the precise mechanisms by which the gut microbiome modulates microglia and, as such, brain function. However, these studies have provided mechanistic insights that might lead to the identification of new microbiome-based therapeutic strategies that target microglial function in brain disorders.

3.4 Neuroendocrine-hypothalamic-pituitary-adrenal axis pathway

The neuroendocrine system regulates many processes in the human body and plays a critical role in organ development and function. Among the primary neuroendocrine pathways, the hypothalamic – pituitary – adrenal (HPA) axis is thought to be tightly connected to the gut microbiome in the context of the MGBA. The HPA includes the hypothalamus, the pituitary gland, and adrenal glands.Citation57 In the context of the stress response, the hypothalamus receives stimuli to produce and release corticotrophin-releasing hormone (CRH), which induces the secretion of adrenocorticotropic hormone (ACTH) from the pituitary gland. ACTH stimulates the adrenal cortex to release glucocorticoids, mineralocorticoids, and catecholamines, which then modulate other downstream processes to produce appropriate responses to the stressor.Citation58 Bidirectional communication between the neuroendocrine-HPA axis system and the gut microbiota involves multiple other components of the MGBA, including the immune system, gut hormones and microbially-derived products, as well as receptors expressed on both the intestinal and the blood-brain barriers as reviewed in ref.Citation58 Several studies have suggested that microbially-derived compounds, such as precursors of neurotransmitters and gut hormones, and SCFAs, can regulate the neuroendocrine system.Citation58,Citation59 Similarly, the microbiome may participate in HPA axis modulation through the immune system.Citation60 Hyperactivation of the HPA axis is associated with disorders affecting both the neuroendocrine system and the gut microbiome, such as irritable bowel syndrome (IBS) and depression. One intriguing hypothesisCitation61 suggests that HPA axis dysregulation might lead to gut dysbiosis and alterations in the integrity of the intestinal barrier, which in turn could promote chronic low-grade inflammation that is seen in both IBSCitation62,Citation63 and depression.Citation64,Citation65

The microbiome is involved in the bidirectional communications between the neuroendocrine system, including the HPA axis, and the immune responseCitation66,Citation67 as reviewed in ref.Citation68 The integrity of the intestinal barrier can be altered by neuroendocrine mediators released in the context of stress response, thus facilitating the release of microbially-derived molecules, which subsequently activate immune pathways.Citation69 For instance, levels of bacterial LPS increase the production of cytokine colony-stimulating factor 1 in muscularis macrophages in the myenteric plexus, which leads to regulation of gut motility, such as increased colonic transit time through the ENS.Citation70 Similarly, alterations in the gut microbiome and its metabolites can induce activation of the HPA axis and associated immune responses.Citation71 Studies in GF mice showed that the absence of gut microorganisms is linked to neuroendocrine and behavioral alterations.Citation72,Citation73 GF mice showed exaggerated HPA axis activation in response to stress, with increased levels of circulating corticosterone, and independent of cytokine-mediated pathways, as shown by unaltered IL-1β and IL-6 levels in the plasma. Intriguingly, colonization with specific strains, such as B. infantis or E. coli, modulated the HPA axis by either increasing or decreasing its activity, respectively.Citation73

3.5 Microbially-derived neuroactive metabolites

The MGBA is regulated by a large number of different neurotransmitters, neuropeptides, and microbially derived products.Citation74 While neurotransmitters have multiple effects on gut ecology, the microbiome itself produces and releases neurotransmitters. Studies showing a microbial origin for dopamine, norepinephrine, gamma-aminobutyric acid (GABA), and serotonin, among others, point toward a potential role of microbiota-produced neurotransmitters in influencing brain function, as discussed below.Citation75 Additionally, neuropeptide synthesis is influenced by hormones and amino acid availability, which is controlled by the microbiome.Citation74 However, neuropeptides can also modulate the composition and function of the gut microbiota, reflecting the complex bidirectional crosstalk that integrates these systems.

Bacteria have unique structural components known as microorganism-associated molecular patterns (MAMPs), such as LPS. MAMPs play an important role in host development and immune function.Citation76 Transformation of host-derived components involves the production of secondary bile acids and steroid hormones, which have neuroactive properties.Citation77 The gut microbiota also plays an important role in the transformation of dietary substrates. Many microbially-derived metabolites of amino acids, carbohydrates, and other plant-derived molecules exert pleiotropic effects on the MGBA and ultimately, on brain function and behavior. The microbial metabolism of tryptophan, tyrosine, and phenylalanine influences the production of neurotransmitters such as serotonin, noradrenaline, and dopamine.Citation78 Tryptophan is particularly relevant to the brain, including tryptophan derivatives, indoles and kynurenine, which can modulate glutamate signaling and have been implicated in anxiety-like behavior and cognitive dysfunction.Citation79,Citation80 The microbiome is also implicated in the production of the major inhibitory neurotransmitter GABA, and alterations in glutamate/GABA circuits in the brain have been associated with the development of autism spectrum disorders, schizophrenia, major depressive disorder, and other neuropsychiatric disorders.Citation11 For instance, a wide analysis of GABA production levels from several commercially available probiotics highlighted the strain-specific ability of Levilactobacillus brevis and Lactiplantibacillus plantarum in secreting GABA both in vitro and in vivo.Citation81 Additionally, expression of GABA receptors can be modulated by specific bacterial strains such as Lactobacillus rhamnosus in mice, with data indicating a decrease in GABAB1b mRNA in the hippocampus and amygdala, and concomitant decrease in GABAAα2 mRNA expression in the prefrontal cortex. These changes were associated with reduced anxiety- and depression-like behaviors.Citation11

Complex plant polysaccharides, or dietary fibers, are fermented by the gut microbiome to produce SCFAs, a class of compounds that has recently gained a great deal of attention because of their ability to influence multiple processes in the host, including behavior.Citation82,Citation83 SCFAs, such as butyrate, propionate, and acetate, are used as an energy source for colonic epithelial cells and can enter the systemic circulation and modulate the immune system through the regulation of gene expression.Citation84 SCFAs can also cross the BBB via monocarboxylate transporter-expressed endothelial cells, where they can directly act on both neurons and microglia (as extensively reviewed in refCitation84)in the neurological and neuropsychiatric disorders investigated. Recent findings on the role of neuroactive metabolites and their effects on the MGBACitation21,Citation85–93 are summarized in . In the following section, we will focus on the link between gut dysbiosis and neurological disorders in the context of the MGBA.

Table 1. Neuroactive metabolites and their effects.

4. Microbiome changes with aging

Microbial colonization of the GI tract begins at birth, with mother-to-infant vertical transfer of skin and vaginal microbial strains, which are then replaced by species typically seen in the adult gut. The first 1,000 days of life are characterized by a relatively low bacterial diversity, with genus Bifidobacterium representing up to 50% of the infants’ gut microbial community, followed by an intense remodeling of the foundational gut microbiota.Citation94 Fluctuations in the infant microbiome configuration follow the transition from a breast milk-based diet to an adult-like diet at the time of introduction of solid food and are also influenced by the surrounding environment.Citation95 Given the instability of the infant gut microbiota, exposure to detrimental environmental factors can disrupt this highly orchestrated microbial succession, leading to gut dysbiosis that persists well beyond the early developmental period and serves as a risk factor for disease later in life.Citation96 This first period of microbial colonization is characterized by rapid changes determined by either environmental factors or intrinsic ecological drifts which continue throughout adolescence. In contrast, relative stability in gut microbial communities is reached during adulthood. Even though environmental factors, such as antibiotic treatment and changes in diet, can alter gut microbiome composition, key species are thought to regulate the integrity and stability of the ecosystem in adult individuals.

Aging is a complex, time-dependent decline of the physiological, immunological, metabolic, and genomic functions in the host. Many studiesCitation97 have attempted to describe the molecular and cellular hallmarks of aging, which include cellular senescence, telomere dysfunction and damage, alterations in protein synthesis and epigenetic regulation, mitochondrial and nutrient-sensing dysfunction, and depletion of stem cell reserves. More recently, chronic inflammation and gut dysbiosis have emerged as additional factors associated with aging.Citation98 Immune aging is characterized by “immunosenescence,” a progressive decline in the ability of both innate and acquired immunity to induce an effective response to both infection and vaccination.Citation99 Changes in the gut microbiome are implicated in both age-related pathologies and potentially act as mediators of the aging process.Citation100 A major change seen in the gastrointestinal tract with aging is a decrease in the barrier integrity of the intestinal epithelium.Citation2 The function of the intestinal barrier can be modulated by commensal resident microbiota. Akkermansia, for example, given orally to mice has been shown to alleviate senescence-related phenotypes in the intestines of aged mice.Citation101 In young mice, higher amounts of Parabacteroides and Akkermansia were found, whereas these two bacterial taxa decreased in the gut of aged mice.Citation101 Akkermansia induces mucus production in the gut, which may help to restore and maintain barrier integrity.Citation102 The effects of aging on the microbiome also contribute to changes in pathways controlled by the gut microbiome, including those involved in the biosynthesis of GABA and SCFA production, which are less enriched in aged mice than in young mice.Citation101 In humans, Bifidobacterium are found at higher levels in infants, while Lachnospiracae levels are higher in adults.Citation103 However, some consistencies remain throughout a healthy lifespan, evident through data that show how certain bacterial species, such as Enterobacteriaceae, are found at similar levels in both infants and the elderly.Citation103 Some researchers have suggested that changes in the microbiota that occur with aging may be better assessed using biological age, rather than chronological age, as the contributing factor. Although methods for determining biological age can produce varying results, researchers have found that the Frailty Index (FI34), a method to calculate biological age, is better correlated with changes in microbiota in humans than chronological age.Citation104 One method of identifying and characterizing biological age may be through characterization of the gut microbiome composition. As implicated in many studiesCitation6,Citation101,Citation103,Citation105,Citation106(), the gut microbiome composition is significantly altered with increasing age, even in the absence of an ND, which further exacerbates gut dysbiosis.

Table 2. Gut microbiota alterations in aging.

5. The role of an aging maternal gut microbiome in offspring neurodevelopment

The maternal exposomeCitation107 in particular, plays a crucial role in early life development.Citation108 Detrimental alterations in the maternal exposome can trigger fetal programming events that predispose offspring to chronic health conditions, including brain disorders, later in life (‘Developmental Origins of Health and Disease’Citation109) (). Multiple studies in the context of maternal obesity, infections, and antibiotic use during pregnancy have confirmed the crucial role of maternal gut dysbiosis as a mediator of offspring’s neurodevelopment.Citation110–112 Interestingly, aging-associated alterations in gut microbiome,Citation113 which are transmitted to the offspring, also result in chronic dysbiosis and increased disease risk in adult offspring. Additional epigenetic programming during early development can also be passed on to subsequent generations.

Figure 3. Environmental factors causing detrimental alterations in the maternal exposome. Toxin and pollutant exposure, infection during pregnancy, diet and metabolic status, smoking, psychosocial stressors such as low socioeconomic status, major life events, and pregnancy-related stressors, can determine broad changes in the maternal environment, thereby jeopardizing pregnancy outcomes and fetal developmental programming. Created in Biorender.com.

Figure 3. Environmental factors causing detrimental alterations in the maternal exposome. Toxin and pollutant exposure, infection during pregnancy, diet and metabolic status, smoking, psychosocial stressors such as low socioeconomic status, major life events, and pregnancy-related stressors, can determine broad changes in the maternal environment, thereby jeopardizing pregnancy outcomes and fetal developmental programming. Created in Biorender.com.

Epidemiological studies have linked advanced maternal age (AMA; ≥35 years) to adverse pregnancy outcomes, such as diabetes and preeclampsia,Citation114 and an increased risk for metabolic and brain disorders in the offspring.Citation115–119 This may involve epigenetic reprogramming in either the oocyteCitation120 or the fetus, or direct effects of inherited dysbiosis. The precise mechanisms by which AMA affects brain development are unclear.Citation114 Over the past three decades in the United States, there has been a steady increase in the birth rate for women aged 35–39 years from 45.9 per 1000 women in 2010, to 52.7 in 2019.Citation121 Similarly, the birth rate for women aged 40–44 years rose by 5% from 2020 to 2021.Citation122 Although a multitude of factors contribute to the increased risk of complications seen in older mothers and their offspring, recent studies have implicated changes in the maternal microbiome that may contribute to these poor outcomes.Citation123,Citation124

Despite the established association between aging, gut dysbiosis, and increased inflammation, few studies have focused on the effects of maternal age-related gut dysbiosis on fetal development and brain health outcomes. A recent study in humans showed that both the vaginal and the gut microbiomes of women displayed significant differences in microbial composition based on age and pregnancy status.Citation125 Given that aging is characterized by gut dysbiosis, an altered intestinal metabolome, increased barrier permeability, and chronic, low-grade inflammation, it is possible that advanced maternal age could alter both the maternal gut microbiome and gut mucosal and systemic immune system, similar to what is observed in maternal obesity. This can then disrupt physiological adaptations to pregnancy and impair placental function, leading to increased brain and systemic inflammation in the fetus and alterations in neurodevelopment (). These effects might be mediated by gut dysbiosis-mediated alterations in the abundance of microbiome-derived metabolites, such as SCFAs,Citation126–128 which can (1) actively modulate maternal immune cells, leading to enhanced systemic and placental inflammationCitation129 and (2) cross the placenta and directly influence epigenetic programming of fetal brain cells, neuroinflammation through microglial activation, and neural circuit formation. After birth, maternally inherited dysbiosis can sustain systemic and neuroinflammatory events in offspring, leading to detrimental effects on brain function and behavior. It is unknown how long these detrimental changes last in the offspring, or if these early developmental events can alter the risk for neurodegenerative diseases later in life. Longitudinal studies are required to address this question.

Figure 4. Proposed mechanism for AMA-related fetal programming and increased risk for brain disorders in offspring. AMA-associated gut dysbiosis and increased inflammation may drive abnormal immune activation in both the placenta and the fetal brain, specifically in microglial cells. Therefore, increased brain inflammation could alter neurodevelopment through multiple mechanisms, including epigenetic modifications in neuronal and glial cells. At birth, vertical transmission of a dysbiotic gut microbiome sustains this systemic neuroinflammation in the neonate, jeopardizing postnatal neurodevelopment and adult health. Created in Biorender.com.

Figure 4. Proposed mechanism for AMA-related fetal programming and increased risk for brain disorders in offspring. AMA-associated gut dysbiosis and increased inflammation may drive abnormal immune activation in both the placenta and the fetal brain, specifically in microglial cells. Therefore, increased brain inflammation could alter neurodevelopment through multiple mechanisms, including epigenetic modifications in neuronal and glial cells. At birth, vertical transmission of a dysbiotic gut microbiome sustains this systemic neuroinflammation in the neonate, jeopardizing postnatal neurodevelopment and adult health. Created in Biorender.com.

6. Neurodegenerative diseases

6.1 Parkinson’s disease (PD)

PD is a neurodegenerative disease characterized by a loss of dopaminergic neurons in the substantia nigra pars compacta and the deposition of insoluble alpha-synuclein polymers in neurons, forming Lewy bodies.Citation130 Approximately 80% of PD patients suffer from GI dysfunction.Citation131 PD patients commonly suffer from symptoms such as constipation, which precede the clinical diagnosis of PD and its other hallmark symptoms such as bradykinesia and dementia,Citation131 indicating that gut dysfunction may play a role in the pathogenesis of PD. Recent studies have attempted to identify specific microbiota changes that may lead to PD. An MPTP-induced mouse model of PD showed very distinct changes in the gut microbiome, including a significant decrease in the levels of Prevotella and Faecalibacterium and an increase in Ralstonia bacteria, compared to control mice.Citation132 Additionally, Enterobacteriaceae is increased in both humans and rodent models of PD.Citation133 The specific mechanisms behind these microbiota changes and their contribution to PD are yet to be understood. However, certain bacterial species present in the gut or fecal matter of patients with PD have been well-described (). Citrobacter rodentium, which is enriched in patients with PD, has also been shown to aggravate motor symptoms in mouse models. Increases in Proteus mirabilis are also linked to PD symptoms and have been shown to promote motor deficits in mouse models of PD.Citation139 Researchers have linked the changes in the expression of these specific bacterial genes to mechanisms that regulate lipid biosynthesis and secretory pathways, including dopamine (DA) regulation and production, as many PD symptoms can be traced back to a decrease in DA levels.Citation140 Studies have shown that some of the bacterial enzymes residing in the gut produce DA,Citation141 further solidifying the link between the roles of microbial-derived metabolites in the progression of PD. This direct correlation between bacterial metabolites and the onset of PD symptoms implies there a role of the MGBA in the pathogenesis of PD ()Citation134–138; however, the specific metabolites involved remain to be studied in-depth.

Table 3. Gut microbiota alterations in Parkinson’s disease (PD).

6.2 Alzheimer’s disease (AD)

One of the most common diseases associated with aging is AD; increasing age is the greatest risk factor for late-onset AD.Citation142 Although some treatments targeting amyloid clearance in AD patients have emerged, the availability of therapeutics targeting the prevention of amyloid development is limited.Citation143 Interestingly, recent studies have demonstrated that amyloid-beta (Aβ) plaque deposition is linked to the composition of the gut microbiota. Studies by our research group have shown that in a Tg2576 transgenic mouse model of AD, gut inflammation and dysbiosis precede the accumulation of amyloid plaques in the brain,Citation144 indicating that gut dysbiosis may play a role in the development of amyloid pathology, although these findings require further validation in additional animal models and in AD patients. In the same study, we found Aβ deposition in postmortem gut samples from patients with AD pathology, which suggests that gut-derived Aβ is associated with AD pathophysiology in some ways. Ongoing studies are exploring specific changes in the microbiome to discover a link between AD and the MGBA. Several studies have demonstrated that the composition of gut microbiota is altered in patients with AD. Vogt et al. found that Firmicutes and Bifidobacterium are decreased and Bacteroidetes are increased in elderly AD patients (age:71.3 ± 7.3 years) compared to age-matched controls (age:69.3 ± 7.5 years).Citation145 Liu et al. also observed a decrease in Firmicutes and an increase in Proteobacteria in the elderly patients.Citation146 In addition, Cattaneo et al. showed that the abundance of pro-inflammatory bacteria such as Escherichia/Shigella is increased in patients, whereas that of an anti-inflammatory bacterium (e.g., E. rectale) is increased.Citation147 These findings indicate that gut microbiota may be associated with AD pathophysiology.Citation148 Other studies ()Citation145,Citation146,Citation149–152 show that among different species and disease models of AD, many of the mechanisms or changes governing microbiota-driven alterations in AD development are similar. Some of these changes, however, appear to be species-dependent, that is, they are observed differently in humans and animal models.

Table 4. Gut microbiota alterations in Alzheimer’s disease (AD).

To understand the role of gut microbiota in AD, microbiota-targeted interventions, including fecal microbiota transplantation (FMT), have recently been employed in animal models of AD. Sun et al. performed FMT from naïve WT mice (6 months old) into age-matched APPswe/PSEN1dE9 transgenic mice. These transgenic mice exhibit occasional Aβ deposits by six months and abundant plaques by nine months.Citation153 Cognitive impairment is seen at 12–13 months in this mouse model.Citation153,Citation154 They found that FMT improves cognitive function and synaptic plasticity, and decreases levels of Aβ40, Aβ42, and p-Tau231 in the brain of recipient mice.Citation155 The beneficial effects of FMT seen in the recipient mice were associated with higher levels of fecal SCFAs, such as butyrate. More recently, Kim et al. transplanted the fecal microbiome of 5×FAD mice into WT mice.Citation156 Compared with many other models, 5×FAD mice show more rapid Aβ deposits in the brain (<3 months) and cognitive impairment (<6 months).Citation157,Citation158 They showed that reconstitution of the 5×FAD microbiome reduced spatial learning and memory in recipient WT mice, compared with recipient mice treated with the biome from WT mice. In addition, recipient mice with the 5×FAD microbiome showed decreased neurogenesis, increased neuroinflammation including microglial activation, and elevated pro-inflammatory cytokines (e.g., TNF-α and IL-1β) in the brain. Interestingly, the recipient mice had increased levels of both pro-inflammatory cytokines (e.g., TNF-α, IL-1β, and IL-6) and anti-inflammatory cytokines (e.g., IL-10) in the colon, whereas only IL-1β, but not the other tested cytokines, was increased in the plasma. This indicates that the reconstitution of the gut microbiota with healthy microbiota can ameliorate memory dysfunction by regulating inflammation in AD mice through the MGBA. It has also been reported that transferring healthy microbiota into ADLPAPT mice, a mouse AD model with both amyloid and neurofibrillary tangle pathology, significantly reduces the formation of amyloid plaques and tangles, resulting in cognitive improvement.Citation151 Taken together, these findings indicate that the restoration of a healthy biome can delay the symptoms and progression of AD in animal models. Thus, future investigations of the role of gut microbiota as new therapeutic targets for AD are warranted.

6.3. Cerebral amyloidosis and cerebral amyloid angiopathy (CAA)

Amyloidosis, one of the most significant pathologies found in AD, is also affected by gut dysbiosis in related neurodegenerative disorders, including CAA. CAA is a small vessel disease characterized by amyloid deposition in the basement membrane of the brain vasculature.Citation159 Aging is a major risk factor for CAA; CAA leads to progressive cognitive impairment in elderly patients, and also contributes to ischemic small vessel disease and intracerebral hemorrhage.Citation160 In a mouse model of APP and PS1 mutations (APP/PS1 mice), Chen et al. showed that the microbiota composition between APP/PS1 and WT mice diverged significantly at 1–3 months of age, prior to the onset of cognitive symptoms, amyloid deposition, and neuroinflammation (e.g., microglial activation) in the brain.Citation161 This study, consistent with many others, demonstrated that higher levels of Enterobacteriaceae, as well as Verrucomicrobia were present in the gut of mice that developed amyloid plaques when compared to control mice.Citation161,Citation162 There are very limited studies demonstrating the regulatory role of gut microbiota in CAA or cerebral amyloidosis. Therefore, the investigation of how vascular Aβ versus parenchymal Aβ affects the gut microbiota and gut dysbiosis-associated cognitive impairment in the context of CAA or cerebral amyloidosis will be an important future direction.

7. Acute neurological injuries

7.1 Stroke

Stroke is a leading cause of mortality and morbidity in elderly patients. Options for acute treatment such as recombinant tissue plasminogen activator and endovascular thrombectomy are available,Citation163 however post-stroke treatment is critical as chronic disability and other long-term health consequences of stroke persist for decades.Citation164 The majority of strokes are caused by occlusion of an artery, either by an embolus or an in-situ thrombosis, leading to an area of brain ischemia.Citation165 Interestingly, recent advances in metagenomics have revealed that stroke remarkably alters the composition of the microbiome, and in turn, this stroke-induced “gut dysbiosis” can exacerbate neuroinflammation and behavioral deficits in a mouse model of stroke.Citation16,Citation166–168 In a study comparing young, stroke mice to uninjured aged controls, we found that the gut microbiome of stroke mice is altered and resembles the microbiome composition of uninjured aged mice.Citation169 It was previously reported that post-stroke translocation of gut microbes into the lung leads to sepsis in mice.Citation170 In other mouse model studies of stroke, some of the more specific microbiota changes have been characterized.Citation126,Citation166–168 Using mouse models of ischemic stroke, such as middle cerebral artery occlusion (MCAO), Singh et al. found that stroke can cause gut dysbiosis, as assessed by reduced bacterial diversity and Bacteroidetes overgrowth, which were associated with impaired gut integrity and motility.Citation166 They subsequently transplanted post-stroke microbiome into GF mice. Interestingly, recipient GF mice had larger infarct volumes and worse behavioral deficits, along with increased pro-inflammatory T cells in both the intestines and the ischemic brain. Furthermore, Benakis et al. revealed that gut microbiota can regulate T cell trafficking from the gut into the leptomeninges after stroke and that specific types of T cells, such as regulatory T (Treg) cells and IL-17+ γδ T cells, are critical in regulating neuroprotection by modulating gut-to-brain signaling following stroke.Citation168

As the elderly are more prone to stroke than younger populations, our research group has focused on the regulatory role of MGBA and the underlying mechanisms of stroke in aged mice. We first examined whether aged mice were more susceptible to stroke-induced gut permeability and bacterial translocation than young mice. Aged mice had increased gut permeability after stroke and higher mortality compared to young mice. When we orally gavaged young and aged mice with GFP-tagged E. coli, aged mice exhibited increased bacterial translocation into peripheral tissues, such as the mesenteric lymph nodes, compared with young mice,Citation171 indicating the direct effect of aging on gut dysbiosis and bacterial translocation after stroke.

To profile the effect of aging on the composition of the gut microbiota, we performed 16S rRNA-seq on fecal samples from young and aged mice. We found that the composition of the gut microbiota is distinct between young and aged mice; the Firmicutes: Bacteroidetes ratio was higher in the aged biome compared to the young biome, indicating age-induced gut dysbiosis. Next, we transplanted the aged microbiome and young microbiome into young and aged mice, respectively, using FMT prior to stroke (transient MCAO). Interestingly, young recipient mice with aged microbiome showed increased post-stroke mortality and functional deficits. Conversely, aged recipient mice transplanted with young microbiome showed better post-stroke outcomes.Citation169 Although we found that preconditioning of the aged gut using a young microbiome prior to stroke can contribute to post-stroke recovery, stroke is not predictable. Therefore, in a separate study, we transplanted young microbiome into aged mice several days after stroke (as a treatment) to determine whether post-stroke FMT can improve post-stroke recovery.Citation126 We found that post-stroke reconstitution of young microbiome significantly improved functional outcomes (e.g., increased spontaneous locomotor activity and cognitive functions, and reduced depressive-like phenotype) along with decreased inflammation in both the brain and gut. Post-stroke, young FMT increased Treg cells in the small intestine and enhanced protective mucus production in the large intestine. Moreover, aged stroke mice with young FMT had higher Treg cells and lower IL-17+ γδ T cells in the ischemic hemisphere than aged stroke mice with aged FMT. Using metabolomic analysis, we revealed that the young microbiome contains higher levels of SCFAs, such as acetate, butyrate, and propionate. Based on our metagenomic data, we selected four SCFA producers (Bifidobacterium longum, Clostridium symbiosum, Faecalibacterium prausnitzii and Lactobacillus fermentum) and orally gavaged aged mice with these, and the pre-biotic inulin, after stroke. Interestingly, post-stroke bacteriotherapy using SCFA-producers and inulin increased SCFA levels in the gut, plasma, and brain, and synergistically improved post-stroke recovery by reducing IL-17 production in γδ T cells in the brains of aged stroke mice. In a follow-up study, we found that the aged microbiome alone is sufficient to produce cognitive decline in young GF mice compared to the young microbiome. In conclusion, our findings suggest that aging should also be considered as a detrimental factor regulating the MGBA in stroke. Several other studiesCitation166,Citation172–175() suggest that the gut microbiome is an essential regulator of post-stroke recovery, and the identification of specific bacterial populations after stroke may uncover various mechanisms through which the microbiome influences inflammation post-stroke.

Table 5. Gut microbiota alterations in stroke.

7.2. Traumatic brain injury

Approximately 2 million people sustain a head injury annually in the United States.Citation176 Along with many other symptoms of traumatic brain injury (TBI), intestinal dysfunction has emerged as a chronic consequence of head injury. Studies in rat models of TBI show a loss of alpha diversity and alterations in bacterial taxa that reside in the gut.Citation177 These changes have also been observed in human fecal samples collected from athletes or trauma patients who sustain head injuries or concussions. One such study involving football players who had concussions demonstrated changes in specific bacterial species, such as Agathobacter and Ruminococcaceae, when compared to healthy, uninjured control subjects.Citation178 Similar results were found in other studiesCitation177–180 and are reviewed in . The effect of TBI on the gut microbiome is immediate. The intestinal microbiota becomes disrupted within hours of injury and can lead to chronic inflammatory processes.Citation181 The acuity of post-TBI gut microbiome changes provides evidence that microbiome-targeted therapies could be beneficial for TBI and related head injuries. Therapies targeting microbial alterations that occur with TBI could alleviate the chronic effects of the injury by limiting downstream consequences at the start of injury progression. A summary of the potential role of the gut microbiota in NDs is provided in .

Figure 5. Major symptoms and features of NDs are accompanied by multiple alterations in specific microbial species, changes which can be consistent or contradictory between the human microbiome and mouse models. Created in Biorender.com.

Figure 5. Major symptoms and features of NDs are accompanied by multiple alterations in specific microbial species, changes which can be consistent or contradictory between the human microbiome and mouse models. Created in Biorender.com.

Table 6. Gut microbiota alterations in traumatic brain injury (TBI).

Conclusions and future directions

Considerable progress has been made in our understanding of the role of gut microbiota and their metabolites in health and disease. In this review, we have summarized key findings demonstrating the regulatory role of microbiota in neurodevelopment, neuroinflammation, and behaviors of the host, specifically in aging and age-related NDs. Although some changes in the gut microbial composition vary depending on the context, and substantial limitations (e.g., discrepancy between preclinical animal studies, differences in the gut microbiota composition between animals and humans, and variations in microbiome sequencing and bioinformatic pipelines) still remain, it is accepted that gut microbiota and metabolites are targetable, suggesting that there are novel therapeutic options for NDs through manipulation of the MGBACitation155,Citation182–187 (). Of note, the microbiome has the potential to transform preventative care and reduce medical costs by enabling individual therapies in the field of precision medicine.Citation188,Citation189 Future studies will highlight bacterial strains, metabolites, and immune factors that might help identify new cellular and molecular targets for diagnostic tools and microbiome-targeting therapeutic and preventative approaches.

Table 7. Microbiome-targeted treatments for NDs.

Acknowledgments

This work was supported by National Institutes of Health grants R35NS132265, R01NS103592, and R01NS094543 (to LDM) and a Career Development Award (857947) from the American Heart Association (to JL).

Disclosure statement

No potential conflict of interest was reported by the author(s).

Additional information

Funding

The work was supported by the American Heart Association and National Institute of Neurological Disorders and Stroke.

References

  • Martin CR, Osadchiy V, Kalani A, Mayer EA. The brain-gut-microbiome axis. Cell Mol Gastroenterol Hepatol. 2018;6:133–27.
  • Walrath T, Dyamenahalli KU, Hulsebus HJ, McCullough RL, Idrovo JP, Boe DM, McMahan RH, Kovacs EJ. Age-related changes in intestinal immunity and the microbiome. J Leukoc Biol. 2021;109(6):1045–1061. doi:10.1002/JLB.3RI0620-405RR.
  • Franceschi C, Bonafè M, Valensin S, Olivieri F, De Luca M, Ottaviani E, DE BENEDICTIS G. Inflamm-aging: An Evolutionary Perspective on Immunosenescence. Ann N Y Acad Sci. 2000;908(1):244–254. doi:10.1111/j.1749-6632.2000.tb06651.x.
  • Takiishi T, Fenero CIM, Câmara NOS. Intestinal barrier and gut microbiota: shaping our immune responses throughout life. Tissue Barriers. 2017;5(4):e1373208. doi:10.1080/21688370.2017.1373208.
  • Li X, Li C, Zhang W, Wang Y, Qian P, Huang H. Inflammation and aging: signaling pathways and intervention therapies. Signal Transduct Target Ther. 2023;8(1):239. doi:10.1038/s41392-023-01502-8.
  • Ghosh TS, Shanahan F, O’Toole PW. Toward an improved definition of a healthy microbiome for healthy aging. Nat Aging. 2022;2(11):1054–69. doi:10.1038/s43587-022-00306-9.
  • Thursby E, Juge N. Introduction to the human gut microbiota. Biochem J. 2017;474(11):1823–36. doi:10.1042/BCJ20160510.
  • Human Microbiome Project C. Structure, function and diversity of the healthy human microbiome. Nature. 2012;486(7402):207–14. doi:10.1038/nature11234.
  • Fan Y, Pedersen O. Gut microbiota in human metabolic health and disease. Nat Rev Microbiol. 2021;19(1):55–71. doi:10.1038/s41579-020-0433-9.
  • Carding S, Verbeke K, Vipond DT, Corfe BM, Owen LJ. Dysbiosis of the gut microbiota in disease. Microb Ecol Health Dis. 2015;26:26191. doi:10.3402/mehd.v26.26191.
  • Bravo JA, Forsythe P, Chew MV, Escaravage E, Savignac HM, Dinan TG, Bienenstock J, Cryan JF. Ingestion of lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci U S A. 2011;108(38):16050–16055. doi:10.1073/pnas.1102999108.
  • Cai Y, Zhou H, Zhu Y, Sun Q, Ji Y, Xue A, Wang Y, Chen W, Yu X, Wang L. et al. Elimination of senescent cells by β-galactosidase-targeted prodrug attenuates inflammation and restores physical function in aged mice. Cell Res. 2020;30(7):574–589. doi:10.1038/s41422-020-0314-9.
  • Kawamoto S, Uemura K, Hori N, Takayasu L, Konishi Y, Katoh K, Matsumoto T, Suzuki M, Sakai Y, Matsudaira T. et al. Bacterial induction of B cell senescence promotes age-related changes in the gut microbiota. Nat Cell Biol. 2023;25(6):865–76. doi:10.1038/s41556-023-01145-5.
  • Cook TM, Mansuy-Aubert V. Communication between the gut microbiota and peripheral nervous system in health and chronic disease. Gut Microbes. 2022;14(1):2068365. doi:10.1080/19490976.2022.2068365.
  • Gibbons CH. Basics of autonomic nervous system function. Handb Clin Neurol. 2019;160:407–418.
  • Durgan DJ, Lee J, McCullough LD, Bryan RM. Examining the role of the microbiota-gut-brain axis in stroke. Stroke. 2019;50(8):2270–7. doi:10.1161/STROKEAHA.119.025140.
  • Bonaz B, Bazin T, Pellissier S. The vagus nerve at the interface of the microbiota-gut-brain axis. Front Neurosci. 2018;12:49. doi:10.3389/fnins.2018.00049.
  • Kaelberer MM, Buchanan KL, Klein ME, Barth BB, Montoya MM, Shen X, Bohórquez DV. A gut-brain neural circuit for nutrient sensory transduction. Sci. 2018;361(6408):361. doi:10.1126/science.aat5236.
  • Fulling C, Dinan TG, Cryan JF. Gut microbe to brain signaling: what happens in vagus…. Neuron. 2019;101(6):998–1002. doi:10.1016/j.neuron.2019.02.008.
  • Arora T, Vanslette AM, Hjorth SA, Backhed F. Microbial regulation of enteroendocrine cells. Med. 2021;2(5):553–70. doi:10.1016/j.medj.2021.03.018.
  • Breit S, Kupferberg A, Rogler G, Hasler G. Vagus nerve as modulator of the brain-gut axis in psychiatric and inflammatory disorders. Front Psychiatry. 2018;9:44. doi:10.3389/fpsyt.2018.00044.
  • Klarer M, Arnold M, Gunther L, Winter C, Langhans W, Meyer U. Gut vagal afferents differentially modulate innate anxiety and learned fear. J Neurosci. 2014;34(21):7067–76. doi:10.1523/JNEUROSCI.0252-14.2014.
  • Klarer M, Weber-Stadlbauer U, Arnold M, Langhans W, Meyer U. Cognitive effects of subdiaphragmatic vagal deafferentation in rats. Neurobiol Learn Mem. 2017;142:190–9. doi:10.1016/j.nlm.2017.05.006.
  • Itoh S, Katsuura G, Hirota R. Diminished circadian rhythm of locomotor activity after vagotomy in rats. Jpn J Physiol. 1981;31(6):957–61. doi:10.2170/jjphysiol.31.957.
  • Klarer M, Krieger JP, Richetto J, Weber-Stadlbauer U, Gunther L, Winter C, Arnold M, Langhans W, Meyer U. Abdominal Vagal Afferents Modulate the Brain Transcriptome and Behaviors Relevant to Schizophrenia. J Neurosci. 2018;38(7):1634–1647. doi:10.1523/JNEUROSCI.0813-17.2017.
  • Shin HC, Jo BG, Lee CY, Lee KW, Namgung U. Hippocampal activation of 5-HT1B receptors and BDNF production by vagus nerve stimulation in rats under chronic restraint stress. Eur J Neurosci. 2019;50(1):1820–30. doi:10.1111/ejn.14368.
  • George MS, Ward HE Jr., Ninan PT, Pollack M, Nahas Z, Anderson B, Kose S, Howland RH, Goodman WK, Ballenger JC. et al. A pilot study of vagus nerve stimulation (VNS) for treatment-resistant anxiety disorders. Brain Stimul. 2008;1(2):112–21. doi:10.1016/j.brs.2008.02.001.
  • Pena DF, Engineer ND, McIntyre CK. Rapid remission of conditioned fear expression with extinction training paired with vagus nerve stimulation. Biol Psychiatry. 2013;73(11):1071–7. doi:10.1016/j.biopsych.2012.10.021.
  • Han W, Tellez LA, Perkins MH, Perez IO, Qu T, Ferreira J, Ferreira TL, Quinn D, Liu Z-W, Gao X-B. et al. A Neural Circuit for Gut-Induced Reward. Cell. 2018;175(3):665–78 e23. doi:10.1016/j.cell.2018.08.049.
  • Goehler LE, Gaykema RP, Opitz N, Reddaway R, Badr N, Lyte M. Activation in vagal afferents and central autonomic pathways: early responses to intestinal infection with Campylobacter jejuni. Brain Behav Immun. 2005;19(4):334–44. doi:10.1016/j.bbi.2004.09.002.
  • Lyte M, Varcoe JJ, Bailey MT. Anxiogenic effect of subclinical bacterial infection in mice in the absence of overt immune activation. Physiology & Behavior. 1998;65(1):63–8. doi:10.1016/S0031-9384(98)00145-0.
  • Nohr MK, Egerod KL, Christiansen SH, Gille A, Offermanns S, Schwartz TW, Møller M. Expression of the short chain fatty acid receptor GPR41/FFAR3 in autonomic and somatic sensory ganglia. Neuroscience. 2015;290:126–137. doi:10.1016/j.neuroscience.2015.01.040.
  • Anitha M, Vijay-Kumar M, Sitaraman SV, Gewirtz AT, Srinivasan S. Gut microbial products regulate murine gastrointestinal motility via Toll-like receptor 4 signaling. Gastroenterology. 2012;143(4):1006–16 e4. doi:10.1053/j.gastro.2012.06.034.
  • McVey Neufeld KA, Mao YK, Bienenstock J, Foster JA, Kunze WA. The microbiome is essential for normal gut intrinsic primary afferent neuron excitability in the mouse. Neurogastroenterol Motil. 2013;25:183–e88. doi:10.1111/nmo.12049.
  • De Vadder F, Grasset E, Manneras Holm L, Karsenty G, Macpherson AJ, Olofsson LE, Bäckhed F. Gut microbiota regulates maturation of the adult enteric nervous system via enteric serotonin networks. Proc Natl Acad Sci U S A. 2018;115(25):6458–6463. doi:10.1073/pnas.1720017115.
  • Caputi V, Marsilio I, Filpa V, Cerantola S, Orso G, Bistoletti M, Paccagnella N, De Martin S, Montopoli M, Dall’Acqua S. et al. Antibiotic-induced dysbiosis of the microbiota impairs gut neuromuscular function in juvenile mice. Br J Pharmacol. 2017;174(20):3623–39. doi:10.1111/bph.13965.
  • Mao YK, Kasper DL, Wang B, Forsythe P, Bienenstock J, Kunze WA. Bacteroides fragilis polysaccharide a is necessary and sufficient for acute activation of intestinal sensory neurons. Nat Commun. 2013;4(1):1465. doi:10.1038/ncomms2478.
  • Hamilton MK, Wall ES, Robinson CD, Guillemin K, Eisen JS, Baumler AJ. Enteric nervous system modulation of luminal pH modifies the microbial environment to promote intestinal health. PloS Pathog. 2022;18(2):e1009989. doi:10.1371/journal.ppat.1009989.
  • Backhed F, Roswall J, Peng Y, Feng Q, Jia H, Kovatcheva-Datchary P, Li Y, Xia Y, Xie H, Zhong H. et al. Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host & Microbe. 2015;17(6):852. doi:10.1016/j.chom.2015.05.012.
  • Mowat AM. To respond or not to respond — a personal perspective of intestinal tolerance. Nat Rev Immunol. 2018;18(6):405–415. doi:10.1038/s41577-018-0002-x.
  • Belkaid Y, Naik S. Compartmentalized and systemic control of tissue immunity by commensals. Nat Immunol. 2013;14(7):646–53. doi:10.1038/ni.2604.
  • Cryan JF, Dinan TG. Microbiota and neuroimmune signalling—Metchnikoff to microglia. Nat Rev Gastroenterol Hepatol. 2015;12(9):494–496. doi:10.1038/nrgastro.2015.127.
  • Erny D, Hrabe de Angelis AL, Jaitin D, Wieghofer P, Staszewski O, David E, Keren-Shaul H, Mahlakoiv T, Jakobshagen K, Buch T. et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci. 2015;18(7):965–77. doi:10.1038/nn.4030.
  • Thion MS, Low D, Silvin A, Chen J, Grisel P, Schulte-Schrepping J, Blecher R, Ulas T, Squarzoni P, Hoeffel G. et al. Microbiome Influences Prenatal and Adult Microglia in a Sex-Specific Manner. Cell. 2018;172(3):500–16 e16. doi:10.1016/j.cell.2017.11.042.
  • Menassa DA, Gomez-Nicola D. Microglial dynamics during human brain development. Front Immunol. 2018;9:1014. doi:10.3389/fimmu.2018.01014.
  • Erny D, Hrabě de Angelis AL, Jaitin D, Wieghofer P, Staszewski O, David E, Keren-Shaul H, Mahlakoiv T, Jakobshagen K, Buch T. et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci. 2015;18(7):965–77. doi:10.1038/nn.4030.
  • Pasciuto E, Burton OT, Roca CP, Lagou V, Rajan WD, Theys T, Mancuso R, Tito RY, Kouser L, Callaerts-Vegh Z. et al. Microglia Require CD4 T Cells to Complete the Fetal-to-Adult Transition. Cell. 2020;182(3):625–40.e24. doi:10.1016/j.cell.2020.06.026.
  • Colonna M, Butovsky O. Microglia function in the central nervous system during health and neurodegeneration. Annu Rev Immunol. 2017;35(1):441–68. doi:10.1146/annurev-immunol-051116-052358.
  • Huang Y, Wu J, Zhang H, Li Y, Wen L, Tan X, Cheng K, Liu Y, Pu J, Liu L. et al. The gut microbiome modulates the transformation of microglial subtypes. Mol Psychiatry. 2023;28(4):1611–1621. doi:10.1038/s41380-023-02017-y.
  • Yamawaki Y, Yoshioka N, Nozaki K, Ito H, Oda K, Harada K, Shirawachi S, Asano S, Aizawa H, Yamawaki S. et al. Sodium butyrate abolishes lipopolysaccharide-induced depression-like behaviors and hippocampal microglial activation in mice. Brain Res. 2018;1680:13–38. doi:10.1016/j.brainres.2017.12.004.
  • Boehme M, Van de Wouw M, Van Sandhu K, Lyons K, Fouhy F, Olavarria Ramirez L, Van Leuven L, Golubeva A, Scott KA, Stanton C. et al. P.1.020 - Targeting the gut microbiome to reverse microglia activation and stress-induced immune priming in ageing. Eur Neuropsychopharm. 2018;28:SS18–SS19. doi:10.1016/j.euroneuro.2017.12.038.
  • Zhou R, Qian S, Cho WCS, Zhou J, Jin C, Zhong Y, Wang J, Zhang X, Xu Z, Tian M. et al. Microbiota-microglia connections in age-related cognition decline. Aging Cell. 2022;21(5):e13599. doi:10.1111/acel.13599.
  • Park JH, Kang I, Lee HK. gammadelta T Cells in Brain Homeostasis and Diseases. Front Immunol. 2022;13:886397.
  • Matcovitch-Natan O, Winter DR, Giladi A, Vargas Aguilar S, Spinrad A, Sarrazin S, Ben-Yehuda H, David E, Zelada González F, Perrin P. et al. Microglia development follows a stepwise program to regulate brain homeostasis. Sci. 2016;353(6301):aad8670. doi:10.1126/science.aad8670.
  • Erny D, Dokalis N, Mezo C, Castoldi A, Mossad O, Staszewski O, Frosch M, Villa M, Fuchs V, Mayer A. et al. Microbiota-derived acetate enables the metabolic fitness of the brain innate immune system during health and disease. Cell Metab. 2021;33(11):2260–76 e7. doi:10.1016/j.cmet.2021.10.010.
  • Dupraz L, Magniez A, Rolhion N, Richard ML, Da Costa G, Touch S, Mayeur C, Planchais J, Agus A, Danne C. et al. Gut microbiota-derived short-chain fatty acids regulate IL-17 production by mouse and human intestinal γδ T cells. Cell Rep. 2021;36(1):109332. doi:10.1016/j.celrep.2021.109332.
  • Cussotto S, Sandhu KV, Dinan TG, Cryan JF. The neuroendocrinology of the microbiota-gut-brain axis: a behavioural perspective. Front Neuroendocrinol. 2018;51:80–101. doi:10.1016/j.yfrne.2018.04.002.
  • Farzi A, Frohlich EE, Holzer P. Gut Microbiota and the Neuroendocrine System. Neurotherapeutics. 2018;15(1):5–22. doi:10.1007/s13311-017-0600-5.
  • Rastelli M, Cani PD, Knauf C. The gut microbiome influences host endocrine functions. Endocr Rev. 2019;40(5):1271–84. doi:10.1210/er.2018-00280.
  • Dinan TG, Cryan JF. Regulation of the stress response by the gut microbiota: implications for psychoneuroen-docrinology. Psychoneuroendocrinology. 2012;37(9):1369–78. doi:10.1016/j.psyneuen.2012.03.007.
  • Misiak B, Loniewski I, Marlicz W, Frydecka D, Szulc A, Rudzki L, Samochowiec J. The HPA axis dysregulation in severe mental illness: can we shift the blame to gut microbiota? Prog Neuropsychopharmacol Biol Psychiatry. 2020;102:109951. doi:10.1016/j.pnpbp.2020.109951.
  • Camilleri M, Lasch K, Zhou W. Irritable bowel syndrome: methods, mechanisms, and pathophysiology. The confluence of increased permeability, inflammation, and pain in irritable bowel syndrome. Am J Physiol Gastrointest Liver Physiol. 2012;303(7):G775–85. doi:10.1152/ajpgi.00155.2012.
  • Rajilic-Stojanovic M, Jonkers DM, Salonen A, Hanevik K, Raes J, Jalanka J, de Vos WM, Manichanh C, Golic N, Enck P. et al. Intestinal microbiota and diet in IBS: causes, consequences, or epiphenomena? Am J Gastroenterol. 2015;110(2):278–87. doi:10.1038/ajg.2014.427.
  • Slyepchenko A, Maes M, Jacka FN, Kohler CA, Barichello T, McIntyre RS, Berk M, Grande I, Foster JA, Vieta E. et al. Gut microbiota, bacterial translocation, and interactions with diet: pathophysiological links between major depressive disorder and non-communicable medical comorbidities. Psychother Psychosom. 2017;86(1):31–46. doi:10.1159/000448957.
  • Jiang H, Ling Z, Zhang Y, Mao H, Ma Z, Yin Y, Wang W, Tang W, Tan Z, Shi J. et al. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav Immun. 2015;48:186–94. doi:10.1016/j.bbi.2015.03.016.
  • Shirtcliff EA, Coe CL, Pollak SD. Early childhood stress is associated with elevated antibody levels to herpes simplex virus type 1. Proc Natl Acad Sci U S A. 2009;106(8):2963–7. doi:10.1073/pnas.0806660106.
  • Serrats J, Schiltz JC, Garcia-Bueno B, van Rooijen N, Reyes TM, Sawchenko PE. Dual roles for perivascular macrophages in immune-to-brain signaling. Neuron. 2010;65(1):94–106. doi:10.1016/j.neuron.2009.11.032.
  • El Aidy S, Dinan TG, Cryan JF. Gut microbiota: the conductor in the orchestra of immune–neuroendocrine communication. Clin Ther. 2015;37(5):954–967. doi:10.1016/j.clinthera.2015.03.002.
  • de Punder K, Pruimboom L. Stress induces endotoxemia and low-grade inflammation by increasing barrier permeability. Front Immunol. 2015;6:223. doi:10.3389/fimmu.2015.00223.
  • Muller PA, Koscsó B, Rajani GM, Stevanovic K, Berres ML, Hashimoto D, Mortha A, Leboeuf M, Li X-M, Mucida D. et al. Crosstalk between muscularis macrophages and enteric neurons regulates gastrointestinal motility. Cell. 2014;158(2):300–13. doi:10.1016/j.cell.2014.04.050.
  • Arentsen T, Qian Y, Gkotzis S, Femenia T, Wang T, Udekwu K, Forssberg H, Diaz Heijtz R. The bacterial peptidoglycan-sensing molecule Pglyrp2 modulates brain development and behavior. Mol Psychiatry. 2017;22(2):257–266. doi:10.1038/mp.2016.182.
  • Roceri M, Hendriks W, Racagni G, Ellenbroek BA, Riva MA. Early maternal deprivation reduces the expression of BDNF and NMDA receptor subunits in rat hippocampus. Mol Psychiatry. 2002;7(6):609–16. doi:10.1038/sj.mp.4001036.
  • Sudo N, Chida Y, Aiba Y, Sonoda J, Oyama N, Yu XN, Kubo C, Koga Y. Postnatal microbial colonization programs the hypothalamic–pituitary–adrenal system for stress response in mice. J Physiol. 2004;558(1):263–275. doi:10.1113/jphysiol.2004.063388.
  • Holzer P. Neuropeptides, Microbiota, and Behavior. Int Rev Neurobiol. 2016;131:67–89.
  • Strandwitz P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018;1693:128–33. doi:10.1016/j.brainres.2018.03.015.
  • Chu H, Mazmanian SK. Innate immune recognition of the microbiota promotes host-microbial symbiosis. Nat Immunol. 2013;14(7):668–75. doi:10.1038/ni.2635.
  • Garcia-Gomez E, Gonzalez-Pedrajo B, Camacho-Arroyo I. Role of sex steroid hormones in bacterial-host interactions. Biomed Res Int. 2013;2013:928290. doi:10.1155/2013/928290.
  • Dodd D, Spitzer MH, Van Treuren W, Merrill BD, Hryckowian AJ, Higginbottom SK, Le A, Cowan TM, Nolan GP, Fischbach MA. et al. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature. 2017;551(7682):648–52. doi:10.1038/nature24661.
  • Schwarcz R, Bruno JP, Muchowski PJ, Wu HQ. Kynurenines in the mammalian brain: when physiology meets pathology. Nat Rev Neurosci. 2012;13(7):465–77. doi:10.1038/nrn3257.
  • Lukic I, Getselter D, Koren O, Elliott E. Role of tryptophan in microbiota-induced depressive-like behavior: evidence from tryptophan depletion study. Front Behav Neurosci. 2019;13:123. doi:10.3389/fnbeh.2019.00123.
  • Monteagudo-Mera A, Fanti V, Rodriguez-Sobstel C, Gibson G, Wijeyesekera A, Karatzas KA, Chakrabarti B. Gamma aminobutyric acid production by commercially available probiotic strains. J Appl Microbiol. 2023;134(2):134. doi:10.1093/jambio/lxac066.
  • Dalile B, Van Oudenhove L, Vervliet B, Verbeke K. The role of short-chain fatty acids in microbiota–gut–brain communication. Nat Rev Gastroenterol Hepatol. 2019;16(8):461–478. doi:10.1038/s41575-019-0157-3.
  • Koh A, De Vadder F, Kovatcheva-Datchary P, Backhed F. From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell. 2016;165(6):1332–45. doi:10.1016/j.cell.2016.05.041.
  • O’Riordan KJ, Collins MK, Moloney GM, Knox EG, Aburto MR, Fülling C, Morley SJ, Clarke G, Schellekens H, Cryan JF. et al. Short chain fatty acids: microbial metabolites for gut-brain axis signalling. Mol Cell Endocrinol. 2022;546:111572. doi:10.1016/j.mce.2022.111572.
  • Needham BD, Kaddurah-Daouk R, Mazmanian SK. Gut microbial molecules in behavioural and neurodegenerative conditions. Nat Rev Neurosci. 2020;21(12):717–31. doi:10.1038/s41583-020-00381-0.
  • van der Hee B, Wells JM. Microbial regulation of host physiology by short-chain fatty acids. Trends Microbiol. 2021;29(8):700–12. doi:10.1016/j.tim.2021.02.001.
  • Roager HM, Licht TR. Microbial tryptophan catabolites in health and disease. Nat Commun. 2018;9(1):3294. doi:10.1038/s41467-018-05470-4.
  • Gao J, Xu K, Liu H, Liu G, Bai M, Peng C, Li T, Yin Y. Impact of the Gut Microbiota on Intestinal Immunity Mediated by Tryptophan Metabolism. Front Cell Infect Microbiol. 2018;8:13. doi:10.3389/fcimb.2018.00013.
  • Ramírez-Pérez O, Cruz-Ramón V, Chinchilla-López P, Méndez-Sánchez N. The role of the gut microbiota in bile acid metabolism. Ann Hepatol. 2017;16:s15–s20. doi:10.5604/01.3001.0010.5672.
  • McMillin M, DeMorrow S. Effects of bile acids on neurological function and disease. FASEB J. 2016;30(11):3658–68. doi:10.1096/fj.201600275R.
  • Godlewska U, Bulanda E, Wypych TP. Bile acids in immunity: bidirectional mediators between the host and the microbiota. Front Immunol. 2022;13:949033. doi:10.3389/fimmu.2022.949033.
  • O’Mahony SM, Clarke G, Borre YE, Dinan TG, Cryan JF. Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav Brain Res. 2015;277:32–48. doi:10.1016/j.bbr.2014.07.027.
  • Banskota S, Khan WI. Gut-derived serotonin and its emerging roles in immune function, inflammation, metabolism and the gut–brain axis. Curr Opin Endocrinol Diabetes Obes. 2022;29(2):177–182. doi:10.1097/MED.0000000000000713.
  • Romano-Keeler J, Sun J. The first 1000 days: assembly of the neonatal microbiome and its impact on health outcomes. Newborn (Clarksville). 2022;1(2):219–26. doi:10.5005/jp-journals-11002-0028.
  • Agosti M, Tandoi F, Morlacchi L, Bossi A. Nutritional and metabolic programming during the first thousand days of life. Pediatr Med Chir. 2017;39(2):157. doi:10.4081/pmc.2017.157.
  • Ville A, Levine E, Zhi D, Lararia B, Wojcicki JM. Alterations in the gut microbiome at 6 months of age in obese latino infants. J Am Coll Nutr. 2020;39(1):47–53. doi:10.1080/07315724.2019.1606744.
  • Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013;153(6):1194–217. doi:10.1016/j.cell.2013.05.039.
  • Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. Hallmarks of aging: an expanding universe. Cell. 2023;186(2):243–78. doi:10.1016/j.cell.2022.11.001.
  • Aiello A, Farzaneh F, Candore G, Caruso C, Davinelli S, Gambino CM, Ligotti ME, Zareian N, Accardi G. Immunosenescence and its hallmarks: how to oppose aging strategically? a review of potential options for therapeutic intervention. Front Immunol. 2019;10:2247. doi:10.3389/fimmu.2019.02247.
  • Alsegiani AS, Shah ZA. The influence of gut microbiota alteration on age-related neuroinflammation and cognitive decline. Neural Regen Res. 2022;17(11):2407–12. doi:10.4103/1673-5374.335837.
  • Shin J, Noh JR, Choe D, Lee N, Song Y, Cho S, Kang E-J, Go M-J, Ha SK, Chang D-H. et al. Ageing and rejuvenation models reveal changes in key microbial communities associated with healthy ageing. Microbiome. 2021;9(1):240. doi:10.1186/s40168-021-01189-5.
  • Rodrigues VF, Elias-Oliveira J, Pereira Í, Pereira JA, Barbosa SC, Machado MSG, Carlos D. Akkermansia muciniphila and gut immune system: a good friendship that attenuates inflammatory bowel disease, obesity, and diabetes. Front Immunol. 2022;13:934695. doi:10.3389/fimmu.2022.934695.
  • Odamaki T, Kato K, Sugahara H, Hashikura N, Takahashi S, Xiao JZ, Abe F, Osawa R. Age-related changes in gut microbiota composition from newborn to centenarian: a cross-sectional study. BMC Microbiol. 2016;16(1):90. doi:10.1186/s12866-016-0708-5.
  • Maffei VJ, Kim S, Blanchard E, Luo M, Jazwinski SM, Taylor CM, Welsh DA. Biological aging and the human gut microbiota. J Gerontol A Biol Sci Med Sci. 2017;72(11):1474–1482. doi:10.1093/gerona/glx042.
  • Xu C, Zhu H, Qiu P. Aging progression of human gut microbiota. BMC Microbiol. 2019;19(1):236. doi:10.1186/s12866-019-1616-2.
  • Scott KA, Ida M, Peterson VL, Prenderville JA, Moloney GM, Izumo T, Murphy K, Murphy A, Ross RP, Stanton C. et al. Revisiting metchnikoff: age-related alterations in microbiota-gut-brain axis in the mouse. Brain Behav Immun. 2017;65:20–32. doi:10.1016/j.bbi.2017.02.004.
  • Robinson O, Vrijheid M. The pregnancy exposome. Curr Environ Health Rep. 2015;2(2):204–213. doi:10.1007/s40572-015-0043-2.
  • Agier L, Basagana X, Hernandez-Ferrer C, Maitre L, Tamayo Uria I, Urquiza J, Andrusaityte S, Casas M, de Castro M, Cequier E. et al. Association between the pregnancy exposome and fetal growth. Int J Epidemiol. 2020;49(2):572–86. doi:10.1093/ije/dyaa017.
  • Barker DJ, Osmond C, Simmonds SJ, Wield GA. The relation of small head circumference and thinness at birth to death from cardiovascular disease in adult life. BMJ. 1993;306(6875):422–6. doi:10.1136/bmj.306.6875.422.
  • Buffington SA, Di Prisco GV, Auchtung TA, Ajami NJ, Petrosino JF, Costa-Mattioli M. Microbial reconstitution reverses maternal diet-induced social and synaptic deficits in offspring. Cell. 2016;165(7):1762–75. doi:10.1016/j.cell.2016.06.001.
  • Hsiao EY, McBride SW, Hsien S, Sharon G, Hyde ER, McCue T, Codelli J, Chow J, Reisman S, Petrosino J. et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell. 2013;155(7):1451–63. doi:10.1016/j.cell.2013.11.024.
  • Di Gesu CM, Matz LM, Bolding IJ, Fultz R, Hoffman KL, Gammazza AM, Petrosino JF, Buffington SA. Maternal gut microbiota mediate intergenerational effects of high-fat diet on descendant social behavior. Cell Rep. 2022;41(2):111461. doi:10.1016/j.celrep.2022.111461.
  • Badal VD, Vaccariello ED, Murray ER, Yu KE, Knight R, Jeste DV, Nguyen TT. The gut microbiome, aging, and longevity: a systematic review. Nutrients. 2020;12(12):12. doi:10.3390/nu12123759.
  • Glick I, Kadish E, Rottenstreich M. Management of pregnancy in women of advanced maternal age: improving outcomes for mother and baby. Int J Women's Health. 2021;13:751–9. doi:10.2147/IJWH.S283216.
  • Cavazos-Rehg PA, Krauss MJ, Spitznagel EL, Bommarito K, Madden T, Olsen MA, Subramaniam H, Peipert JF, Bierut LJ. Maternal age and risk of labor and delivery complications. Matern Child Health J. 2015;19(6):1202–1211. doi:10.1007/s10995-014-1624-7.
  • Cooke CLM, Davidge ST. Advanced maternal age and the impact on maternal and offspring cardiovascular health. Am J Physiol Heart Circ Physiol. 2019;317(2):H387–H394. doi:10.1152/ajpheart.00045.2019.
  • Lopez-Castroman J, Gomez DD, Belloso JJ, Fernandez-Navarro P, Perez-Rodriguez MM, Villamor IB, Navarrete FF, Ginestar CM, Currier D, Torres MR. et al. Differences in maternal and paternal age between schizophrenia and other psychiatric disorders. Schizophr Res. 2010;116(2–3):184–90. doi:10.1016/j.schres.2009.11.006.
  • Sandin S, Hultman CM, Kolevzon A, Gross R, MacCabe JH, Reichenberg A. Advancing maternal age is associated with increasing risk for autism: a review and meta-analysis. J Am Acad Child Adolesc Psychiatry. 2012;51(5):477–486.e1. doi:10.1016/j.jaac.2012.02.018.
  • Polga N, Macul Ferreira de Barros P, Farhat LC, Bloch MH, Lafer B, de Almeida KM. Parental age and the risk of bipolar disorder in the offspring: A systematic review and meta-analysis. Acta Psychiatr Scand. 2022;145(6):568–577. doi:10.1111/acps.13418.
  • Ge ZJ, Schatten H, Zhang CL, Sun QY. Oocyte ageing and epigenetics. Reproduction. 2015;149(3):R103–14. doi:10.1530/REP-14-0242.
  • Hamilton B, Martin J, Osterman M, Driscoll A, Rossen L. Births: provisional data for 2021 vital statistics rapid release. National Center for Health Statistics. 2020. Available online: https://wwwcdc.gov/nchs/data/vsrr/vsrr020.pdf accessed on 31 August 2022.
  • Osterman MJ, Hamilton BE, Martin JA, Driscoll AK, Valenzuela CP. Births: final data for 2021. Natl Vital Stat Rep. 2023 Jan;72(1):1–53.
  • Jin J, Gao L, Zou X, Zhang Y, Zheng Z, Zhang X, Li J, Tian Z, Wang X, Gu J. et al. Gut dysbiosis promotes preeclampsia by regulating macrophages and trophoblasts. Circ Res. 2022;131(6):492–506. doi:10.1161/CIRCRESAHA.122.320771.
  • Liberale L, Montecucco F, Tardif JC, Libby P, Camici GG. Inflamm-ageing: the role of inflammation in age-dependent cardiovascular disease. Eur Heart J. 2020;41(31):2974–82. doi:10.1093/eurheartj/ehz961.
  • Huang Y, Li D, Cai W, Zhu H, Shane MI, Liao C, Pan S. Distribution of vaginal and gut microbiome in advanced maternal age. Front Cell Infect Microbiol. 2022;12:819802. doi:10.3389/fcimb.2022.819802.
  • Lee J, d’Aigle J, Atadja L, Quaicoe V, Honarpisheh P, Ganesh BP, Hassan A, Graf J, Petrosino J, Putluri N. et al. Gut microbiota–derived short-chain fatty acids promote poststroke recovery in aged mice. Circ Res. 2020;127(4):453–465. doi:10.1161/CIRCRESAHA.119.316448.
  • Wang S, Liu Y, Qin S, Yang H. Composition of maternal circulating short-chain fatty acids in gestational diabetes mellitus and their associations with placental metabolism. Nutrients. 2022;14(18):14. doi:10.3390/nu14183727.
  • Chun J, Toldi G. The impact of short-chain fatty acids on neonatal regulatory T cells. Nutrients. 2022;14(18):14. doi:10.3390/nu14183670.
  • Lim AI, McFadden T, Link VM, Han SJ, Karlsson RM, Stacy A, Farley TK, Lima-Junior DS, Harrison OJ, Desai JV. et al. Prenatal maternal infection promotes tissue-specific immunity and inflammation in offspring. Sci. 2021;373(6558). doi:10.1126/science.abf3002.
  • Meade RM, Fairlie DP, Mason JM. Alpha-synuclein structure and Parkinson’s disease – lessons and emerging principles. Mol Neurodegener. 2019;14(1):29. doi:10.1186/s13024-019-0329-1.
  • Fasano A, Visanji NP, Liu LW, Lang AE, Pfeiffer RF. Gastrointestinal dysfunction in Parkinson’s disease. Lancet Neurol. 2015;14(6):625–39. doi:10.1016/S1474-4422(15)00007-1.
  • Aktas B. Gut Microbial Alteration in MPTP Mouse Model of Parkinson Disease is Administration Regimen Dependent. Cell Mol Neurobiol. 2023;43(6):2815–29. doi:10.1007/s10571-023-01319-7.
  • Gerhardt S, Mohajeri MH. Changes of colonic bacterial composition in Parkinson’s disease and other neurodegenerative diseases. Nutrients. 2018 Jun 1;10(6):708. doi:10.3390/nu10060708.
  • Shen T, Yue Y, He T, Huang C, Qu B, Lv W, Lai H-Y. The Association Between the Gut Microbiota and Parkinson’s Disease, a Meta-Analysis. Front Aging Neurosci. 2021;13:636545. doi:10.3389/fnagi.2021.636545.
  • Cirstea MS, Yu AC, Golz E, Sundvick K, Kliger D, Radisavljevic N, Foulger LH, Mackenzie M, Huan T, Finlay BB. et al. Microbiota composition and metabolism are associated with gut function in Parkinson’s disease. Mov Disord. 2020;35(7):1208–17. doi:10.1002/mds.28052.
  • Boertien JM, Murtomäki K, Pereira PAB, van der Zee S, Mertsalmi TH, Levo R, Nojonen T, Mäkinen E, Jaakkola E, Laine P. et al. Fecal microbiome alterations in treatment-naive de novo Parkinson’s disease. NPJ Parkinsons Dis. 2022;8(1):129. doi:10.1038/s41531-022-00395-8.
  • Lai F, Jiang R, Xie W, Liu X, Tang Y, Xiao H, Gao J, Jia Y, Bai Q. Intestinal pathology and gut microbiota alterations in a Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson’s disease. Neurochem Res. 2018;43(43):1986–1999. doi:10.1007/s11064-018-2620-x.
  • Sampson TR, Debelius JW, Thron T, Janssen S, Shastri GG, Ilhan ZE, Challis C, Schretter CE, Rocha S, Gradinaru V. et al. Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson’s Disease. Cell. 2016;167(6):1469–80.e12. doi:10.1016/j.cell.2016.11.018.
  • Choi JG, Kim N, Ju IG, Eo H, Lim SM, Jang SE, Kim D-H, Oh MS. Oral administration of Proteus mirabilis damages dopaminergic neurons and motor functions in mice. Sci Rep. 2018;8(1):1275. doi:10.1038/s41598-018-19646-x.
  • Xicoy H, Wieringa B, Martens GJM. The role of lipids in Parkinson’s disease. Cells. 2019;8(1):27. doi:10.3390/cells8010027.
  • Hamamah S, Hajnal A, Covasa M. Impact of Nutrition, Microbiota Transplant and Weight Loss Surgery on Dopaminergic Alterations in Parkinson’s Disease and Obesity. Int J Mol Sci. 2022;23(14):23. doi:10.3390/ijms23147503.
  • Guerreiro R, Bras J. The age factor in Alzheimer’s disease. Genome Med. 2015;7(1):106. doi:10.1186/s13073-015-0232-5.
  • Plascencia-Villa G, Perry G. Lessons from antiamyloid-β immunotherapies in Alzheimer’s disease. Handb Clin Neurol. 2023;193:267–292.
  • Honarpisheh P, Reynolds CR, Blasco Conesa MP, Moruno Manchon JF, Putluri N, Bhattacharjee MB, Urayama A, McCullough LD, Ganesh BP. Dysregulated Gut Homeostasis Observed Prior to the Accumulation of the Brain Amyloid-β in Tg2576 Mice. Int J Mol Sci. 2020;21(5):21. doi:10.3390/ijms21051711.
  • Vogt NM, Kerby RL, Dill-McFarland KA, Harding SJ, Merluzzi AP, Johnson SC, Carlsson CM, Asthana S, Zetterberg H, Blennow K. et al. Gut microbiome alterations in Alzheimer’s disease. Sci Rep. 2017;7(1):13537. doi:10.1038/s41598-017-13601-y.
  • Liu P, Wu L, Peng G, Han Y, Tang R, Ge J, Zhang L, Jia L, Yue S, Zhou K. et al. Altered microbiomes distinguish Alzheimer’s disease from amnestic mild cognitive impairment and health in a Chinese cohort. Brain Behav Immun. 2019;80:633–43. doi:10.1016/j.bbi.2019.05.008.
  • Cattaneo A, Cattane N, Galluzzi S, Provasi S, Lopizzo N, Festari C, Ferrari C, Guerra UP, Paghera B, Muscio C. et al. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol Aging. 2017;49:60–8. doi:10.1016/j.neurobiolaging.2016.08.019.
  • He Y, Li B, Sun D, Chen S. Gut Microbiota: Implications in Alzheimer’s Disease. JCM. 2020;9(7):2042. doi:10.3390/jcm9072042.
  • Fan KC, Lin CC, Liu YC, Chao YP, Lai YJ, Chiu YL, Chuang Y-F. Altered gut microbiota in older adults with mild cognitive impairment: a case-control study. Front Aging Neurosci. 2023;15:1162057. doi:10.3389/fnagi.2023.1162057.
  • Shukla PK, Delotterie DF, Xiao J, Pierre JF, Rao R, McDonald MP, Khan MM. Alterations in the gut-microbial-inflammasome-brain axis in a mouse model of alzheimer’s disease. Cells. 2021;10(4):10. doi:10.3390/cells10040779.
  • Kim MS, Kim Y, Choi H, Kim W, Park S, Lee D, Kim DK, Kim HJ, Choi H, Hyun D-W. et al. Transfer of a healthy microbiota reduces amyloid and tau pathology in an Alzheimer’s disease animal model. Gut. 2020;69(2):283–94. doi:10.1136/gutjnl-2018-317431.
  • Wei Z, Li D, Shi J. Alterations of spatial memory and gut microbiota composition in alzheimer’s disease triple-transgenic mice at 3, 6, and 9 months of age. Am J Alzheimers Dis Other Demen. 2023;38:15333175231174193. doi:10.1177/15333175231174193.
  • Jankowsky JL, Fadale DJ, Anderson J, Xu GM, Gonzales V, Jenkins NA, Copeland NG, Lee MK, Younkin LH, Wagner SL. et al. Mutant presenilins specifically elevate the levels of the 42 residue β-amyloid peptide in vivo: evidence for augmentation of a 42-specific γ secretase. Hum Mol Genet. 2004;13(2):159–170. doi:10.1093/hmg/ddh019.
  • Volianskis A, Køstner R, Mølgaard M, Hass S, Jensen MS. Episodic memory deficits are not related to altered glutamatergic synaptic transmission and plasticity in the CA1 hippocampus of the APPswe/PS1ΔE9-deleted transgenic mice model of β-amyloidosis. Neurobiol Aging. 2010;31(7):1173–1187. doi:10.1016/j.neurobiolaging.2008.08.005.
  • Sun J, Xu J, Ling Y, Wang F, Gong T, Yang C, Ye S, Ye K, Wei D, Song Z. et al. Fecal microbiota transplantation alleviated Alzheimer’s disease-like pathogenesis in APP/PS1 transgenic mice. Transl Psychiatry. 2019;9(1):189. doi:10.1038/s41398-019-0525-3.
  • Kim N, Jeon SH, Ju IG, Gee MS, Do J, Oh MS, Lee JK. Transplantation of gut microbiota derived from Alzheimer’s disease mouse model impairs memory function and neurogenesis in C57BL/6 mice. Brain Behav Immun. 2021;98:357–365. doi:10.1016/j.bbi.2021.09.002.
  • Richard BC, Kurdakova A, Baches S, Bayer TA, Weggen S, Wirths O. Gene dosage dependent aggravation of the neurological phenotype in the 5XFAD mouse model of Alzheimer’s disease. J Alzheimers Dis. 2015;45(4):1223–36. doi:10.3233/JAD-143120.
  • Jawhar S, Trawicka A, Jenneckens C, Bayer TA, Wirths O. Motor deficits, neuron loss, and reduced anxiety coinciding with axonal degeneration and intraneuronal Aβ aggregation in the 5XFAD mouse model of Alzheimer’s disease. Neurobiology Of Aging. 2012;33(1):196.e29–40. doi:10.1016/j.neurobiolaging.2010.05.027.
  • Thal DR, Grinberg LT, Attems J. Vascular dementia: different forms of vessel disorders contribute to the development of dementia in the elderly brain. Exp Gerontol. 2012;47(11):816–24. doi:10.1016/j.exger.2012.05.023.
  • Gireud-Goss M, Mack AF, McCullough LD, Urayama A. Cerebral amyloid angiopathy and blood-brain barrier dysfunction. Neuroscientist. 2021;27(6):668–84. doi:10.1177/1073858420954811.
  • Chen Y, Fang L, Chen S, Zhou H, Fan Y, Lin L, Li J, Xu J, Chen Y, Ma Y. et al. Gut microbiome alterations precede cerebral amyloidosis and microglial pathology in a mouse model of Alzheimer’s disease. Biomed Res Int. 2020;2020:1–15. doi:10.1155/2020/8456596.
  • Wasén C, Simonsen E, Ekwudo MN, Profant MR, Cox LM. The emerging role of the microbiome in Alzheimer’s disease. Int Rev Neurobiol. 2022;167:101–139.
  • Powers WJ, Rabinstein AA, Ackerson T, Adeoye OM, Bambakidis NC, Becker K, Biller J, Brown M, Demaerschalk BM, Hoh B. et al. Guidelines for the early management of patients with acute ischemic stroke: 2019 update to the 2018 guidelines for the early management of acute ischemic stroke: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2019;50(12):e344–e418. doi:10.1161/STR.0000000000000211.
  • Teasell R, Mehta S, Pereira S, McIntyre A, Janzen S, Allen L, Lobo L, Viana R. Time to rethink long-term rehabilitation management of stroke patients. Top Stroke Rehabil. 2012;19(6):457–462. doi:10.1310/tsr1906-457.
  • Sommer CJ. Ischemic stroke: experimental models and reality. Acta Neuropathol. 2017;133(2):245–61. doi:10.1007/s00401-017-1667-0.
  • Singh V, Roth S, Llovera G, Sadler R, Garzetti D, Stecher B, Dichgans M, Liesz A. Microbiota dysbiosis controls the neuroinflammatory response after stroke. J Neurosci. 2016;36(28):7428–7440. doi:10.1523/JNEUROSCI.1114-16.2016.
  • Singh V, Sadler R, Heindl S, Llovera G, Roth S, Benakis C, Liesz A. The gut microbiome primes a cerebroprotective immune response after stroke. J Cereb Blood Flow Metab. 2018;38(8):1293–1298. doi:10.1177/0271678X18780130.
  • Benakis C, Brea D, Caballero S, Faraco G, Moore J, Murphy M, Sita G, Racchumi G, Ling L, Pamer EG. et al. Commensal microbiota affects ischemic stroke outcome by regulating intestinal γδ T cells. Nat Med. 2016;22(5):516–23. doi:10.1038/nm.4068.
  • Spychala MS, Venna VR, Jandzinski M, Doran SJ, Durgan DJ, Ganesh BP, Ajami NJ, Putluri N, Graf J, Bryan RM. et al. Age-related changes in the gut microbiota influence systemic inflammation and stroke outcome. Ann Neurol. 2018;84(1):23–36. doi:10.1002/ana.25250.
  • Stanley D, Mason LJ, Mackin KE, Srikhanta YN, Lyras D, Prakash MD, Nurgali K, Venegas A, Hill MD, Moore RJ. et al. Translocation and dissemination of commensal bacteria in post-stroke infection. Nat Med. 2016;22(11):1277–84. doi:10.1038/nm.4194.
  • Crapser J, Ritzel R, Verna R, Venna VR, Liu F, Chauhan A, Koellhoffer E, Patel A, Ricker A, Maas K. et al. Ischemic stroke induces gut permeability and enhances bacterial translocation leading to sepsis in aged mice. Aging (Albany NY). 2016;8(5):1049–1063. doi:10.18632/aging.100952.
  • Zeng X, Gao X, Peng Y, Wu Q, Zhu J, Tan C, Xia G, You C, Xu R, Pan S. et al. Higher risk of stroke is correlated with increased opportunistic pathogen load and reduced levels of butyrate-producing bacteria in the gut. Front Cell Infect Microbiol. 2019;9:4. doi:10.3389/fcimb.2019.00004.
  • Zhang X, Wang X, Zhao H, Cao R, Dang Y, Yu B, Jia X-Z. Imbalance of microbacterial diversity is associated with functional prognosis of stroke. Neural Plast. 2023;2023:1–13. doi:10.1155/2023/6297653.
  • Meng C, Deng P, Miao R, Tang H, Li Y, Wang J, Wu J, Wang W, Liu S, Xia J. et al. Gut microbiome and risk of ischaemic stroke: a comprehensive Mendelian randomization study. Eur J Prev Cardiol. 2023;30(7):613–20. doi:10.1093/eurjpc/zwad052.
  • Wang H, Zhang M, Li J, Liang J, Yang M, Xia G, Ren Y, Zhou H, Wu Q, He Y. et al. Gut microbiota is causally associated with poststroke cognitive impairment through lipopolysaccharide and butyrate. J Neuroinflammation. 2022;19(1):76. doi:10.1186/s12974-022-02435-9.
  • Taylor CA, Bell JM, Breiding MJ, Xu L. Traumatic brain injury–related emergency department visits, hospitalizations, and deaths — United States, 2007 and 2013. MMWR Surveill Summ. 2017;66(9):1–16. doi:10.15585/mmwr.ss6609a1.
  • Taraskina A, Ignatyeva O, Lisovaya D, Ivanov M, Ivanova L, Golovicheva V, Baydakova G, Silachev D, Popkov V, Ivanets T. et al. Effects of Traumatic Brain Injury on the Gut Microbiota Composition and Serum Amino Acid Profile in Rats. Cells. 2022;11(9):11. doi:10.3390/cells11091409.
  • Soriano S, Curry K, Sadrameli SS, Wang Q, Nute M, Reeves E, Kabir R, Wiese J, Criswell A, Schodrof S. et al. Alterations to the gut microbiome after sport-related concussion in a collegiate football players cohort: A pilot study. Brain Behav Immun Health. 2022;21:100438. doi:10.1016/j.bbih.2022.100438.
  • Urban RJ, Pyles RB, Stewart CJ, Ajami N, Randolph KM, Durham WJ, Danesi CP, Dillon EL, Summons JR, Singh CK. et al. Altered fecal microbiome years after traumatic brain injury. J Neurotrauma. 2020;37(8):1037–51. doi:10.1089/neu.2019.6688.
  • Celorrio M, Shumilov K, Rodgers R, Schriefer L, Li Y, Baldridge MT, Friess SH. Innate and Peripheral Immune Alterations after Traumatic Brain Injury Are Regulated in a Gut Microbiota-Dependent Manner in Mice. J Neurotrauma. 2023;40(7–8):772–787. doi:10.1089/neu.2022.0356.
  • Hayakawa M, Asahara T, Henzan N, Murakami H, Yamamoto H, Mukai N, Minami Y, Sugano M, Kubota N, Uegaki S. et al. Dramatic changes of the gut flora immediately after severe and sudden insults. Dig Dis Sci. 2011;56(8):2361–5. doi:10.1007/s10620-011-1649-3.
  • Aljumaah MR, Bhatia U, Roach J, Gunstad J, Azcarate Peril MA. The gut microbiome, mild cognitive impairment, and probiotics: A randomized clinical trial in middle-aged and older adults. Clin Nutr. 2022;41(11):2565–2576. doi:10.1016/j.clnu.2022.09.012.
  • Tian H, Wang J, Feng R, Zhang R, Liu H, Qin C. et al. Efficacy of faecal microbiota transplantation in patients with progressive supranuclear palsy-Richardson’s syndrome: a phase 2, single centre, randomised clinical trial. EClinicalMedicine. 2023;58:101888. doi:10.1016/j.eclinm.2023.101888.
  • Marseglia A, Xu W, Fratiglioni L, Fabbri C, Berendsen AAM, Bialecka-Debek A, Jennings A, Gillings R, Meunier N, Caumon E. et al. Effect of the NU-AGE diet on cognitive functioning in older adults: a randomized controlled trial. Front Physiol. 2018;9:349. doi:10.3389/fphys.2018.00349.
  • Cuervo-Zanatta D, Syeda T, Sánchez-Valle V, Irene-Fierro M, Torres-Aguilar P, Torres-Ramos MA, Shibayama-Salas M, Silva-Olivares A, Noriega LG, Torres N. et al. Dietary fiber modulates the release of gut bacterial products preventing cognitive decline in an alzheimer’s mouse model. Cell Mol Neurobiol. 2023;43(4):1595–618. doi:10.1007/s10571-022-01268-7.
  • Abdelhamid M, Zhou C, Jung CG, Michikawa M. Probiotic Bifidobacterium breve MCC1274 mitigates Alzheimer’s disease-related pathologies in wild-type mice. Nutrients. 2022;14(12):14. doi:10.3390/nu14122543.
  • Zhao Z, Ning J, Bao XQ, Shang M, Ma J, Li G, Zhang D. Fecal microbiota transplantation protects rotenone-induced Parkinson’s disease mice via suppressing inflammation mediated by the lipopolysaccharide-TLR4 signaling pathway through the microbiota-gut-brain axis. Microbiome. 2021;9(1):226. doi:10.1186/s40168-021-01107-9.
  • Scarpellini E, Rinninella E, Basilico M, Colomier E, Rasetti C, Larussa T, Santori P, Abenavoli L. From pre- and probiotics to post-biotics: a narrative review. Int J Environ Res Public Health. 2021;19(1):37. doi:10.3390/ijerph19010037.
  • Behrouzi A, Nafari AH, Siadat SD. The significance of microbiome in personalized medicine. Clin Transl Med. 2019;8(1):16. doi:10.1186/s40169-019-0232-y.