Introduction: From a Single-Injury Model to a Systemic Regulatory Framework
The Dual Challenge of Spinal Cord Injury Epidemiology
Spinal cord injury (SCI) is a severe and debilitating condition that commonly results from traffic accidents, falls, and sports-related trauma. It is caused primarily by compression, traction, or contusion of the spinal cord, leading to partial or complete loss of motor, sensory, and autonomic functions.1 Globally, it is estimated that 2 to 3 million individuals are living with SCI, with 250,000 to 500,000 new cases reported annually.2 The lifetime cost of medical care and assistance for SCI patients can range from $500,000 to $2 million.3 While paralysis represents the most visible consequence of SCI, its associated systemic complications are equally significant, including cardiovascular disease, metabolic syndrome, urogenital and gastrointestinal dysfunction, chronic pain, immunosuppression, anxiety, and depression.4,5 However, most current therapies are limited in their ability to modulate systemic inflammatory responses or address long-term multisystem dysfunctions, highlighting the need for novel interventions that target upstream drivers of secondary injury.
The prognosis of SCI is closely linked to the severity of the primary injury, the timing of therapeutic interventions, and the progression of secondary pathological responses.6 Surgical decompression within 24–36 hours after injury is recognized as an effective and critical treatment for improving neurological outcomes in SCI patients.7 Adjunctive therapies such as hyperbaric oxygen, pulsed electrical stimulation, mild hypothermia, acupuncture, and laser puncture have also been employed to support functional recovery.8 Pharmacological treatments, neurotrophic factor administration, cell transplantation, gene therapy, and signaling pathway modulation hold therapeutic promise but still require extensive clinical validation.7,9
SCI poses dual therapeutic challenges: minimizing secondary damage in the acute phase and promoting functional restoration in the chronic phase. In the early stage, the focus was on neuroprotection and the preservation of spinal cord integrity. In contrast, the chronic stage requires sustained strategies that target neural regeneration, control of inflammation, and systemic recovery. This multifaceted progression suggests that SCI treatment cannot depend on a single modality but rather demands interdisciplinary, multimodal strategies tailored to different stages of recovery.
Evolution and Scientific Significance of the “Gut‒Spinal Cord Axis” Concept
The concept of the “gut‒spinal cord axis” is built upon extensive research into the interactions between the gut microbiota and the central nervous system (CNS). Early studies focused on the regulatory mechanisms of the neuroendocrine–immune network within the “gut–brain axis”, revealing that gut microbes can influence brain function via the vagus nerve, short-chain fatty acids (SCFAs), and various cytokines.10–12 However, accumulating evidence has indicated that the influence of the gut microbiota extends beyond the brain, playing a pivotal role in modulating the spinal cord microenvironment as well.13,14 Through metabolites, immune signaling molecules, and neuroactive compounds, the gut microbiota can directly or indirectly regulate spinal cord inflammation, oxidative stress, and axonal regeneration, thus giving rise to the theoretical framework of the gut‒spinal cord axis (Figure 1).15,16
Figure 1 Conceptual framework of the gut–spinal cord axis. A schematic illustration showing the bidirectional communication between the gut microbiota and the spinal cord. Red arrows indicate the influence of the spinal cord on the gut, while blue arrows indicate the influence of the gut on the spinal cord.
|
The bidirectional regulation of the gut‒spinal cord axis exhibits significant spatiotemporal dynamics. Temporally, gut dysbiosis triggered by acute SCI can aggravate secondary injury through the activation of proinflammatory signaling cascades. Disruption of the gut microbiota after SCI facilitates microbial translocation and systemic dissemination of pathogen-associated molecular patterns (PAMPs), such as lipopolysaccharide (LPS), which activate Toll-like receptor (TLR) and inflammasome pathways in the injured spinal cord.17 This immune activation contributes to sustained microglial activation, cytokine production, and blood–spinal cord barrier breakdown, thereby amplifying secondary neuroinflammatory damage.18 In contrast, the restoration of microbial metabolic homeostasis during the chronic phase contributes critically to neuroregeneration.19 Spatially, alterations in the spinal cord microenvironment—such as aberrant neuronal excitability and glial activation—can provide feedback to influence gut barrier integrity and microbiota composition via autonomic and humoral pathways. This bidirectional communication creates a cross-system cascade of “neurotrauma–gut dysbiosis–inflammatory amplification”.20 This spatiotemporal interplay suggests that the gut‒spinal cord axis is not a static or linear pathway but rather a dynamic regulatory network involving both local and systemic mechanisms. The emergence of this concept challenges the traditional paradigm that treats the neural, immune, and metabolic systems as independent entities. Instead, it highlights their systemic crosstalk, particularly in the complex pathophysiology following SCI. The gut microbiota and their metabolites not only contribute to neurorepair but also actively regulate immune responses during the recovery process. Immune and neural interactions occurring at distinct time points and spatial scales converge into a dynamic feedback loop, underscoring the interdependence of multisystem coordination in SCI pathology. This conceptual advancement offers new insights into therapeutic strategies, emphasizing the necessity of integrating neuroimmune and microbial modulation in SCI management.
The dynamic interplay between post-SCI inflammatory progression and gut microbiota homeostasis represents a critical regulatory nexus that determines the outcome of neural repair. This review systematically examines the spatiotemporal associations between neuroinflammation and gut dysbiosis following SCI, with a focus on the bidirectional modulation exerted by the gut–spinal cord axis on the cascading pathological events initiated by SCI. By integrating multidisciplinary evidence from neuroimmunology, metabolomics, and microbiome research, we aim to deconstruct the multidimensional network through which the gut microbiota influences spinal microenvironmental homeostasis via metabolic–immune–neural circuits. On this basis, we further evaluated the therapeutic potential of microbiota-targeted interventions—such as probiotics and fecal microbiota transplantation (FMT)—in disrupting the vicious cycle of inflammation–regeneration imbalance. This analytical framework not only offers new perspectives for overcoming the limitations of conventional monodimensional therapies but also advances SCI research toward a translational paradigm rooted in multisystem integration.
Inflammatory Responses Following SCI: Local Initiation and Systemic Amplification
Mechanisms Underlying the Local Initiation of Inflammation After SCI
The pathological mechanisms of SCI are complex and generally categorized into primary and secondary injuries.21,22 Primary injury results from mechanical compression or external forces that cause immediate disruption of spinal cord structures such as axons, blood vessels, and cells. The severity and extent of the primary injury directly influence the prognosis and overall outcome of SCI.1 As the damage caused by primary injury is often irreversible, current research efforts—both domestically and internationally—are focused primarily on targeting secondary injury.23 Among the various mechanisms of secondary injury, neuroinflammation plays a central role, and its spatiotemporal dynamics critically shape the microenvironment for neuronal survival.24
At the molecular level, neuroinflammation following SCI involves a complex hierarchical regulatory network.25 Damage-associated molecular patterns (DAMPs), which are released from necrotic cells at the injury site, have been identified as key initiators and accelerators of proinflammatory mediator release.26 These DAMPs activate TLR signaling pathways, thereby triggering rapid responses from microglia and astrocytes.27 Activated glial cells secrete proinflammatory cytokines such as IL-1β, TNF-α, and IL-6, along with chemokines such as CCL2 and CXCL1, which collectively recruit peripheral neutrophils and monocytes to the lesion site, initiating a neuroinflammatory cascade.14 Further studies have shown that a variety of mediators—such as matrix metalloproteinase-8 (MMP-8), cyclooxygenase-2 (COX-2), and interferon-gamma (IFN-γ)—are released in a sequential manner, contributing not only to neuronal apoptosis but also to the remodeling of blood‒spinal cord barrier (BSCB) permeability, thereby forming a self‒amplifying loop.28
Temporally, post-SCI neuroinflammation is typically divided into three phases: acute, subacute, and chronic.21,29 In humans, the transition from the acute to subacute phase generally occurs within several hours to 48 hours after injury, whereas progression to the chronic phase is expected to take place within six months.1 In the subacute and chronic stages, neuroinflammatory responses are key contributors to ongoing cell death and tissue degradation, thus constituting a fundamental component of secondary injury.30 Different types of immune cells are involved at distinct time points in the injury process. Neutrophils, as early responders, are rapidly recruited to the injury site within one hour via the release of the chemokines IL-1β and CXCL1/CXCL2 by activated microglia and astrocytes31 The infiltration of these cells becomes detectable within three hours, peaks at 1–3 days, and resolves by approximately day seven.32 These cells exacerbate tissue damage by releasing reactive oxygen species (ROS) and matrix metalloproteinases (MMPs) and further propagate the inflammatory cascade by recruiting peripheral monocytes through proinflammatory signals.33 By day three postinjury, monocytes infiltrate the lesion and differentiate into macrophages.34 These monocyte-derived macrophages (MDMs) predominantly exhibit a proinflammatory M1 phenotype and are typically restricted to the first week after SCI. In contrast, microglia serve as the primary source of anti-inflammatory M2 cells.34 As intrinsic immune cells of the central nervous system, microglia are rapidly activated following injury, reach peak activity between days 3–7, and remain persistently activated for more than 180 days, spanning the entire course from acute to chronic inflammation.32,35 During the chronic phase of SCI, adaptive immunity becomes increasingly dominant. T-cell subsets, such as Th1 and Th17 cells, participate in the regulation of neuroinflammation. Hyperactivation of Th1 cells has been implicated in the disruption of the BSCB, thereby aggravating demyelinating injury.36 Concurrently, B cells can initiate systemic autoimmune responses, ultimately leading to the amplification of local inflammation and systemic immune dysregulation.37 Collectively, the spatiotemporal dynamics of local neuroinflammation, orchestrated by a coordinated interplay of diverse cellular and molecular mediators, drive the progression of secondary injury. This not only shapes the pathological microenvironment at the lesion site but also lays the groundwork for the subsequent propagation of systemic inflammation and involvement of distal organs.
Spatiotemporal Expansion of Systemic Inflammation and Effects on Distant Organs After SCI
SCI is not confined to local neuroinflammation but also induces systemic immune dysregulation and multiorgan dysfunction. The resulting inflammatory cascade exhibits both time-dependent and spatially expansive characteristics.30 The intensity, extent, and duration of the systemic inflammatory response are closely associated with the severity of the injury. During the acute phase, the levels of proinflammatory cytokines such as IL-1β and TNF-α are rapidly elevated, accompanied by massive infiltration of immune cells, including monocytes/macrophages and microglia.38 As the injury progresses into the chronic phase (>40 days), persistent activation of inflammatory cells, such as Ly6C+ macrophages, can still be detected in cases of severe injury, whereas inflammatory responses tend to subside in mild injuries.38 Furthermore, spinal cord injury-induced immune depression syndrome (SCI-IDS) is characterized by a profound decline in peripheral immune function in patients with severe SCI. Although this may reduce the risk of autoimmune responses, it significantly increases susceptibility to infections.39 The sustained state of chronic inflammation not only hampers neural repair but also contributes to distal organ damage, such as neuropathic pain and gastrointestinal dysfunction, thereby forming a vicious cycle of “inflammation–neuroimbalance”.40
One of the key mechanisms underlying the systemic dissemination of inflammation is the hemodynamic disturbance caused by SCI. The descending sympathetic vasomotor fibers originating from the spinal cord are disrupted, resulting in diminished sympathetic tone, systemic arterial hypotension, and redistribution of peripheral blood flow.41 It has been reported that up to 22% of patients with acute SCI develop gastrointestinal bleeding (GIB), which poses a significant risk to both morbidity and mortality.42 Margo et al43 described a 21-year-old female with traumatic SCI who died from asymptomatic yet fatal GIB due to autonomic dysfunction, highlighting the high-risk nature of SCI-associated gastrointestinal pathology. In addition to reducing gastrointestinal perfusion and inducing ischemia‒reperfusion injury, hemodynamic instability may impair the autonomic regulation of gastrointestinal function.41 Under normal conditions, gastrointestinal motility and secretion are coregulated by the enteric nervous system (ENS) and the autonomic nervous system (ANS), with the ANS primarily modulating rather than directly controlling ENS activity.44 Following SCI, disruption of spinal sympathetic axons impairs central signaling, resulting in colonic dysrhythmia, disrupted sensory transmission, and sphincter dyssynergia.44,45 Moreover, autonomic imbalance exacerbates gastrointestinal dysmotility, manifesting as prolonged intestinal transit time, decreased peristaltic frequency, and vascular tone dysregulation.46 Consequently, more than 60% of SCI patients experience constipation or fecal incontinence.47,48 These pathological manifestations are defined as neurogenic bowel dysfunction (NBD), which is clinically classified into upper motor neuron (UMN) and lower motor neuron (LMN) types on the basis of the degree of injury.49 Both forms involve ENS impairment and persistent intestinal inflammation. Although the ENS possesses intrinsic neuroplasticity in response to physiological stimuli,50 experimental models have shown that SCI leads to structural atrophy (ie, decreased neuronal density) and functional impairment (ie, reduced intestinal myoelectric activity) of the ENS.51 These pathological changes result in minimal recovery over long-term follow-up52 It is hypothesized that SCI-induced systemic imbalances, such as the activation of inflammatory cascades and gut microbial dysbiosis, may alter the local intestinal microenvironment and hinder the regenerative capacity of the ENS.53,54 This pathological cascade may result in persistent NBD, contributing significantly to increased hospitalization rates and long-term mortality.55–57
The traumatic nature of SCI provides a unique therapeutic window for early intervention in NBD. However, current first-line therapies, including laxatives and stool softeners, are poorly effective in the context of SCI and may even worsen fecal incontinence,58 indicating that symptom-based management fails to address the core pathophysiological mechanisms of the gut‒spinal axis. Numerous studies have revealed a strong association between gut microbiota dysbiosis and NBD symptoms in SCI patients. Zhang et al59 emphasized the disrupted gut microbial ecology in NBD patients and its correlation with prolonged defecation time. Moreover, patients with quadriplegia had significantly longer defecation times than paraplegic patients and healthy individuals did. Yu et al60 further demonstrated that gut microbiota diversity was significantly lower in patients with complete SCI than in those with incomplete SCI and that reduced microbial diversity was correlated with higher NBD scores. These findings suggest that SCI may aggravate NBD not only by affecting autonomic and ENS function but also by altering the composition of the gut microbiota.
Gut Microbiota Dysbiosis: A Systemic Inflammation Amplifier in SCI
Onset and Dynamic Characteristics of Gut Microbiota Dysbiosis Following SCI
Significant Alterations in Microbiota Composition
The gut microbiota plays a crucial role in maintaining bidirectional communication between the gastrointestinal tract and the central nervous system, as well as in regulating immune responses and metabolic homeostasis.14 Most current studies investigating gut microbiota alterations in SCI patients utilize fecal samples because of their noninvasive nature and lower risk of cross-contamination than do tissue biopsies.61 However, even for the same condition, the results across different studies often vary significantly. These discrepancies may stem from factors such as individual variability, sampling techniques, and sequencing depth but also reflect the methodological limitations of the current research. For example, the majority of studies rely on 16S rRNA sequencing, which offers limited taxonomic resolution—typically at the genus level—thus constraining our understanding of the functional changes in the microbiota associated with SCI.62
Despite these limitations, numerous clinical and animal studies have consistently demonstrated characteristic alterations in the gut microbiota composition following SCI, with alpha diversity changes receiving particular attention. Some clinical studies have reported significantly reduced Shannon, Chao1, and phylogenetic diversity (PD) indices in patients with complete or incomplete SCI, indicating decreased richness and evenness.63 However, other studies have reported increased Chao1 and decreased Simpson indices in patients with chronic thoracolumbar or cervical injuries, suggesting increased richness but reduced evenness.59,64 Some even report higher overall alpha diversity (Chao1, PD, Shannon, and Simpson indices) in SCI patients than in healthy controls.65 These contradictions may result not only from differences in species abundance estimation methods (eg, OTU clustering bias) but also from variations in the injury stage—acute disruptions may be followed by compensatory microbiota remodeling during the chronic phase.66 Notably, discrepancies between animal and human studies are further influenced by research design. Rodent models (eg, homogenized female samples under controlled interventions) often show no significant differences in alpha diversity,49,67–70 whereas human studies (typically male-dominated and confounded by disease duration, medications, and diet) are more likely to reveal heterogeneous patterns, although causality remains unclear owing to the observational nature of these data.71,72 Although alpha diversity is often regarded as an indicator of gut health,73 microbiota reconstruction following SCI frequently involves the expansion of proinflammatory bacteria, highlighting that shifts in diversity metrics alone cannot fully reflect ecosystem functional stability.66
The gut microbiota composition after SCI is typically evaluated at both the phylum and genus levels.74 At the phylum level, the dominant taxa include Firmicutes, Bacteroidetes, Actinobacteria, Proteobacteria, and Verrucomicrobia,75 with Firmicutes and Bacteroidetes together accounting for approximately 90% of total gut bacteria and playing essential roles in intestinal homeostasis.76 A reduced Firmicutes/Bacteroidetes (F/B) ratio is widely considered a hallmark of dysbiosis and is frequently observed in patients with inflammatory bowel disease (IBD)77 and similarly reported in patients with SCI.78 In a cohort study involving 54 Turkish SCI patients, Gungor et al49 reported significantly reduced Firmicutes abundance compared with that in healthy controls. Kong et al71 confirmed this trend and reported an increase in Synergistota. Conversely, a Chinese cohort study revealed elevated levels of Proteobacteria and Verrucomicrobia alongside reduced Bacteroidetes in patients with chronic complete SCI.59 Findings from animal models partly mirror those of human studies. For example, Jing et al79 reported reduced Firmicutes and increased Bacteroidetes in SCI mice, with Firmicutes abundance inversely correlated with motor recovery. Myers et al80 reported increased Proteobacteria and elevated systemic inflammation marker CD14 in a C57BL/6 mouse SCI model. At the genus level, although species-specific changes vary among studies, a consistent core pattern emerges: expansion of proinflammatory taxa and loss of anti-inflammatory genera. Multiple studies have reported significant reductions in Lactobacillus, Allobaculum, Sutterella, Prevotella, Faecalibacterium, and Dialister and enrichment of proinflammatory genera such as Bacteroides, Blautia, Escherichia-Shigella, Alistipes, Rikenella, Staphylococcus, Anaerotruncus, and Mucispirillum.49,63,78,81 Notably, host specificity strongly influences the trajectory of certain genera. For example, Blautia abundance is elevated in patients with SCI, particularly during the chronic phase,49,59,64 whereas it tends to decrease in animal models.79 Similarly, Bacteroides is not emphasized in animal studies but is consistently enriched in human SCI cohorts,59,64 likely reflecting host-specific regulatory effects on the gut microbiome. Differences in microbial shifts may also be shaped by SCI severity, diet, environmental factors, and antibiotic usage.71 However, most human studies to date are cross-sectional and observational in design, which limits their ability to establish causal relationships between microbiota changes and SCI pathophysiology. Although specific microbial signatures have been associated with injury severity, disease stage, and systemic inflammation, it remains unclear whether these alterations are causative or merely reflective of host responses and confounding variables. To address these challenges, future research should prioritize longitudinal cohort studies and multi-omics integration—including metagenomics, metabolomics, and host immune profiling—to unravel the temporal dynamics and functional relevance of microbiota shifts in SCI.
While current SCI microbiome studies have predominantly focused on bacterial populations, recent work in murine models has demonstrated that the virome also undergoes significant shifts after SCI. Specifically, Du et al81 applied genome-resolved metagenomic and viromic profiling to SCI mice and observed decreases in phages associated with beneficial commensals and increases in phages linked to potentially pathogenic bacteria. Although direct evidence in human SCI is still lacking, insights from gastrointestinal diseases such as inflammatory bowel syndrome suggest that mycobiome and virome alterations may influence intestinal barrier integrity, immune activation, and microbial composition.82,83 Future multiomics studies in SCI should therefore aim to include viromic and mycobiomic analyses to better understand gut–CNS interactions across microbial kingdoms.
Time-Dependent Shifts in the Gut Microbiota Post-Injury
The temporal dynamics of gut microbiota dysbiosis following SCI are markedly time dependent, and inconsistencies across studies may stem from variability in sampling time points. Initial alterations appear as early as 3 days postinjury, becoming more pronounced between days 7 and 40.38,80,84 However, some studies report partial microbiota recovery during the subacute phase, with certain taxa returning to baseline levels.13,68,69,85 For example, O’Connor et al67 reported decreased Clostridium abundance and increased IL-1β levels at 8 weeks post-SCI, whereas Jing et al86 reported increased Clostridium abundance at 4 weeks, underscoring the temporal sensitivity of microbiota changes. Doelman et al,85 using a porcine model, delineated distinct shifts between the acute (0–14 days) and subacute (>14 days) phases, with phyla such as Firmicutes and Spirochaetes exhibiting dynamic changes, indicating that dysbiosis represents an ongoing, adaptive process. Acute-phase changes (3–40 days) tend to stabilize progressively, with an increase in Sutterella potentially linked to heightened inflammation, whereas a decline in Burkholderiaceae in the chronic phase (>41 days) correlates with persistent motor deficits.85 Although some studies report partial restoration of specific bacterial taxa, the gut microbial ecosystem rarely returns to a preinjury homeostatic state, emphasizing the critical importance of early therapeutic interventions.87
Injury Severity–Associated Patterns of Microbial Dysbiosis
The pattern of gut microbiota dysregulation after SCI is influenced by both the anatomical location of the injury (cervical vs thoracolumbar) and the extent of neurological impairment (complete vs incomplete injury), with both factors contributing to microbial community heterogeneity.88 Among patients categorized as AIS grade A or B by the American Spinal Injury Association, higher abundances of Lactobacillus have been observed, whereas Bacteroides, Faecalibacterium, and Helicobacteraceae were more prevalent in AIS grade C or D patients.89 A comparative study of complete (CTSCI) and incomplete (ITSCI) injuries revealed nine significantly different genera: Coriobacteriaceae, Synergistetes, Eubacterium, and Cloacibacillus were enriched in CTSCI, whereas Lactobacillaceae, Lachnospiraceae, Eubacterium, Clostridium, and Sutterella were more abundant in ITSCI60. Different severities of injury also result in distinct microbial profiles. Zhang et al59 reported increased Bacteroides abundance in tetraplegia patients and increased Blautia, Porphyromonadaceae, and Lactobacillus abundance in paraplegia patients. Another study revealed reduced Firmicutes at the T10 level and increased Actinobacteria at both T4 and T10.81 Together, injury severity and progression time jointly shape microbiota dynamics, accounting for interstudy variability. For example, during the early postinjury stages (day 3), the abundance of the orders Bifidobacteriales and Lactobacillales increased, whereas that of Bifidobacterium decreased significantly; by day 13, Akkermansia had nearly disappeared. By day 41, more severe injuries were associated with more pronounced microbiota disruptions.13
Mechanisms of Gut Microbiota Dysbiosis Following SCI
The mechanisms underlying gut microbiota dysbiosis following SCI are multifaceted and involve a range of interrelated factors, including disrupted neural regulation, intestinal immune imbalance, compromised epithelial barrier integrity, and prolonged antibiotic use (Figure 2). Collectively, these factors disrupt intestinal homeostasis and ultimately lead to microbial dysbiosis.
![]() |
Figure 2 Mechanisms underlying gut microbiota dysbiosis following SCI.
|
Disruption of Central and Enteric Neural Control of Gastrointestinal Function
SCI disrupts supraspinal regulation of sympathetic preganglionic neurons, impairing postganglionic control of gastrointestinal (GI) function and systemic homeostasis.20 The loss of autonomic neural input compromises gastrointestinal motility, mucus secretion, immune activation, and epithelial barrier integrity, facilitating bacterial translocation and contributing to gut microbial dysbiosis.49 This may be attributed to damaged sympathetic regulation of the stomach, small intestine, and colon, subsequently leading to imbalances in the ENS.14 Lefèvre et al90 reported that in mice with T8 SCI, nitrergic neurons were diminished, and acetylcholine levels decreased, although the proportion of cholinergic neurons remained unchanged. Conversely, White et al91 reported reductions in both nitrergic and cholinergic neurons in T3 SCI mice. These findings suggest that SCI perturbs intestinal motility and secretion by altering ENS neuronal subtypes. Moreover, prolonged bed rest in SCI patients exacerbates GI dysmotility, impairs mucosal secretion, and compromises immune function, thereby further disrupting the gut microenvironment.39 This chronic dysfunction not only undermines gastrointestinal physiology but also interacts with microbial dysbiosis to increase infection risk and the immune burden, ultimately impeding patient recovery and quality of life.
Immune Dysregulation in Gut-Associated Lymphoid Tissue (GALT)
Approximately 70–80% of the body’s immune cells reside in GALT, which is under sympathetic regulation.92 SCI disrupts this neural control, destabilizing immune homeostasis within the GALT.92 Kigerl et al93 demonstrated increased expression of TNF, IL-10, IL-1β, and TGF-β in mesenteric lymph nodes by day 3 postinjury. Temporal analysis revealed that acute-phase alterations in microbial composition were associated with IL-12-driven Th1 immune responses, whereas chronic-phase changes were linked to IL-1β-associated OTUs (n = 23),67 indicating the dynamic participation of inflammatory mediators in microbiota remodeling. These findings imply that sympathetic dysregulation following SCI alters GALT reactivity, thereby impacting the gut microbial balance.
Compromised Intestinal Epithelial Barrier Integrity
The ENS maintains the intestinal barrier by segregating the sterile lamina propria from antigen-rich luminal contents.94 Tight junctions, comprising claudins, occludins, and members of the TAMP (tight junction-associated MARVEL protein) family, are essential for barrier integrity.95 Occludin, a key tight junction protein, is downregulated by proinflammatory cytokines such as TNF-α via transcriptional suppression.96 Barrier dysfunction following SCI has been well reported.97,98 Jing et al86 reported reduced expression of zonula occludens-1 (ZO-1), occludin (OCLN), claudin-3 (CLDN3), and claudin-5 (CLDN5) in the colonic tissues of SCI mice, accompanied by increased intestinal permeability. This disruption facilitates the translocation of bacteria and their metabolites into systemic circulation, increasing susceptibility to enterogenic infections and intestinal diseases. SCI-induced barrier impairment also delays locomotor recovery in mice.93 Notably, FMT was shown to restore occludin expression and reduce TNF-α, NF-κB, and IL-1β levels in colon tissue, suggesting a regulatory role for inflammatory cytokines in barrier function.99 Additionally, hyperactivation of the hypothalamic–pituitary–adrenal (HPA) axis post-SCI increases glucocorticoid release, which further increases intestinal permeability and exacerbates dysbiosis.100
Long-Term Use of Antibiotics
Antibiotics are frequently administered after SCI because of their anti-inflammatory and neuroprotective properties.101 However, accumulating evidence indicates that prolonged antibiotic use exacerbates microbial dysbiosis, which is characterized by increased relative abundances of Firmicutes, Proteus, and Actinobacillus and decreased Bacteroides, ultimately impairing functional recovery.86,89,102 Furthermore, SCI itself induces autonomic dysfunction, fostering microbial imbalance and immunosuppression, which increases susceptibility to infection.103 This vulnerability often necessitates repeated antibiotic interventions, establishing a vicious cycle that further disrupts microbial homeostasis.20 Notably, recent findings suggest a synergistic effect between SCI-induced dysbiosis and antibiotic-mediated microbial depletion, leading to increased variability in microbiota composition.67
Gut Microbiota-Mediated Mechanisms Regulating Inflammatory Responses After SCI
Microbiota-Derived Metabolites and Their Signaling Roles in Neuroinflammation After SCI
The gut microbiota exerts profound immunomodulatory effects through the production of various metabolites, which serve as critical signaling mediators in the pathogenesis of neuroinflammation following SCI. Among these, representative classes such as LPS, SCFAs, tryptophan-derived compounds and bile acid have been the most extensively studied. Although these metabolites differ in origin and mode of action, they converge upon key immunological and neuroinflammatory pathways. In this section, we comprehensively review each major class of microbiota-derived metabolites, their signaling mechanisms, and their roles in mediating peripheral and central inflammation after SCI.
LPS–TLR Signaling: A Proinflammatory Axis Amplified by Gram-Negative Bacteria
LPS, a major component of gram-negative bacterial cell walls, is released upon bacterial lysis in the gut.104 In the context of gastrointestinal dysfunction and compromised intestinal barrier integrity, LPS may translocate across the intestinal epithelium into the systemic circulation, thereby eliciting systemic inflammatory responses.105 This translocation occurs primarily via paracellular leakage due to tight junction disruption, as well as increased epithelial permeability following microbial dysbiosis, inflammation, and enterocyte apoptosis.106 Circulating LPS activates innate immune receptors in peripheral tissues, sustaining a systemic proinflammatory state characterized by elevated cytokine production and immune cell infiltration.107 LPS activates pattern recognition receptors such as TLR4,108 promoting the systemic translocation of bacterial products and inducing both local and systemic inflammation.109 While LPS cannot cross the intact blood–brain barrier (BBB), traumatic insults such as SCI increase BBB permeability, allowing LPS infiltration into the CNS, where it activates resident immune cells such as microglia, initiating neuroinflammatory responses.110,111 MYERS et al80 demonstrated that SCI leads to expansion of the phylum Proteobacteria—rich in gram-negative species—concomitant with systemic endotoxemia and endoplasmic reticulum stress within the injured spinal cord. These findings highlight that microbial-derived endotoxins influence both peripheral and central inflammation following SCI. In the spinal cord, LPS binds to TLR4 on microglia, activating the MyD88/NF-κB signaling cascade and increasing the levels of proinflammatory cytokines such as TNF-α and IL-1β, thereby exacerbating neuroinflammation and neuronal damage.112–115 In addition to TLR4, microglia express a wide array of TLRs (1–9), whereas astrocytes express TLR3 under basal conditions and upregulate TLR2 and TLR4 upon activation.27 LPS-induced TLR4 activation leads not only to NF-κB signaling but also to NLRP3 inflammasome activation and pyroptosis.116 Dong et al117 reported that oral administration of curcumin in a TBI model modulated gut microbial diversity, reduced serum LPS levels, and suppressed the TLR4/MyD88/NF-κB pathway, leading to decreased inflammatory cytokine production. These findings support the existence of a pathological gut–LPS–neuroinflammation axis following SCI, wherein microbiota-derived LPS breaches the compromised intestine and BBB to activate neuroimmune responses, thus contributing to the secondary progression of spinal cord pathology.
SCFAs and Multitarget Pathways: Multifaceted Modulators of Neuroinflammation
SCFAs, including acetate, propionate, and butyrate, are microbial fermentation products of dietary fiber and complex carbohydrates.118 SCFAs not only serve as an energy source for colonic epithelial cells but also act as signaling molecules that confer numerous health benefits.119 Physiological concentrations of SCFAs are essential for maintaining intestinal and blood–brain barrier integrity, immune homeostasis, and CNS health.120,121 In SCI patients, a marked reduction in Firmicutes and altered Firmicutes-to-Bacteroidetes ratios are correlated with diminished SCFA production.89 Acetate, propionate, and butyrate levels are strongly positively correlated with beneficial taxa such as Faecalibacterium, Agathobacter, and Megamonas.122 SCFAs may cross the intestinal barrier into the circulation and influence CNS cells either directly or indirectly via immune and endocrine signaling.20,93,123,124
In germ-free mice colonized with SCFA-producing Clostridium butyricum and Bacteroides thetaiotaomicron, BBB permeability was reduced, and the expression of tight junction proteins in the cortex and hypothalamus was elevated.125 Similar protective effects extend to the BSCB, where SCFAs—particularly valproic acid—have been shown to increase the expression of tight junction proteins and reduce paracellular leakage following SCI, which helps preserve BSCB integrity and prevents the infiltration of peripheral immune cells into the injured spinal cord.126,127 These effects are attributable to the direct actions of SCFAs on epithelial and endothelial barriers and neuronal structures, as well as their ability to regulate immune responses.128 Jing et al79 demonstrated that FMT from healthy mice into SCI models increased microbial diversity, restored SCFA levels, suppressed NF-κB activation, and improved motor function. Mechanistically, SCFAs act as endogenous inhibitors of histone deacetylases (HDACs), thereby suppressing NF-κB signaling in glial cells and downregulating proinflammatory cytokine expression.119,129 Among them, sodium butyrate is a potent HDAC inhibitor that directly modulates glial cell states. It reduces microglial activation, promotes anti-inflammatory M2-like polarization, and suppresses astrocytic reactivity, thereby mitigating glial-driven neuroinflammation.130 Other HDAC inhibitors such as valproic acid have shown similar anti-inflammatory effects, acting through the STAT3/NF-κB axis to suppress microglia-mediated neuroinflammation in SCI models.126 Similarly, pentanoate-labeled chitosan nanoparticles have been shown to repair the blood–spinal cord barrier and reduce astrocyte activation after SCI,131 mirroring findings in TBI models.132 SCFAs also modulate immune responses via G protein–coupled receptor 43 (GPR43), facilitating the differentiation of Th17 cells into Treg cells and mitigating CNS inflammation.133,134 In animal models, oral SCFA administration promotes anti-inflammatory T-cell phenotypes, enhances Treg-derived IL-10 secretion, and facilitates Treg migration to the lesion site, thereby suppressing neuroinflammation via Treg–γδT cell interactions.121 Collectively, these findings suggest that SCFAs serve as pivotal mediators in the gut–spinal cord axis and represent potential therapeutic targets for modulating neuroinflammation in SCI.
Tryptophan Metabolites and AHR Signaling: Microbial Pathways in Neuroimmune Regulation
Tryptophan (TRP) is an essential amino acid whose metabolites play critical roles in numerous neurological disorders.135 TRP is primarily catabolized through the kynurenine pathway (KP), serotonin pathway, and indole pathway, yielding bioactive compounds such as kynurenine (KYN), 5-hydroxytryptamine (5-HT), and various indole derivatives.136 Gut microbes are indispensable in mediating these metabolic routes.137 TRP metabolism is enriched among taxa such as Actinobacteria, Firmicutes, Bacteroidetes, Proteobacteria, and Clostridia, with genera such as Clostridium, Burkholderia, Streptomyces, Pseudomonas, and Bacillus exhibiting robust metabolic capacities.138 Consequently, alterations in the composition of the gut microbiota substantially impact TRP metabolic outputs and associated immune regulatory functions.
In the indole pathway, approximately 4–6% of dietary TRP is converted into indoles (eg, indole-3-acetic acid [IAA] and indole-3-propionic acid [IPA]) by commensal bacteria such as Escherichia coli, Bacteroides, and Clostridia via the enzyme tryptophanase (TnaA).139,140 IPA is synthesized predominantly by Lactobacillus spp. and other members of Firmicutes.141 Clostridium sporogenes is capable of producing both IAA and IPA via oxidative and reductive routes, processes that are crucial for maintaining intestinal homeostasis and mitigating inflammation.142 Microbial dysbiosis can disrupt these pathways, thereby exacerbating neuroinflammatory responses. Within the KP, probiotics such as Lactobacillus and Bifidobacterium downregulate the expression of indoleamine 2,3-dioxygenase (IDO), a key enzyme that catalyzes the formation of neurotoxic metabolites, including 3-hydroxykynurenine (3-HK) and quinolinic acid (QUIN), thus offering neuroprotection141,143 For example, Roseburia intestinalis has been reported to effectively suppress the accumulation of KYN and QUIN,144 whereas species such as Clostridium and Burkholderia promote the conversion of TRP to QUIN.138,145 This skewed metabolic direction has been closely linked to the pathogenesis of several neurodegenerative and neuropsychiatric disorders, including Alzheimer’s disease,146 major depressive disorder,147 and Parkinson’s disease.148 Within the 5-HT pathway, Lactobacillus and Bifidobacterium species have been shown to facilitate the conversion of TRP into 5-HT, modulating both central and peripheral serotonergic signaling. They also increase 5-HT synthesis by upregulating the expression of tryptophan hydroxylase 1 (TPH1), a rate-limiting enzyme in 5-HT biosynthesis.149,150 A reduced abundance of lactic acid bacteria has been associated with decreased plasma and intestinal 5-HT levels.151,152 Specific strains, such as Lactobacillus plantarum DR7 and Roseburia intestinalis, have been demonstrated to increase TPH1/2 expression and alleviate depression-like behaviors in animal models.144,153 However, some studies also suggest that the ingestion of certain Lactobacillus strains may lead to reductions in both 5-HT and brain-derived neurotrophic factor (BDNF) levels, indicating that the neuromodulatory effects of these bacteria are likely strain-specific.154
In addition to metabolic regulation, tryptophan-derived metabolites also exert potent anti-inflammatory effects via activation of AHR, forming the basis of a “tryptophan–AHR–anti-inflammatory signaling axis”. Rothhammer et al155 reported that TRP metabolites, in concert with type I interferons (IFN-Is), activate AHR in astrocytes, thereby inducing the expression of suppressor of cytokine signaling 2 (SOCS2). SOCS2, in turn, downregulates the NF-κB signaling pathway, effectively dampening neuroinflammation.150 Notably, SOCS2-deficient mice exhibit exaggerated NF-κB activation upon LPS stimulation, underscoring the essential role of SOCS2 in mediating AHR-dependent anti-inflammatory responses.156 Thus, the TRP metabolite–AHR–SOCS2–NF-κB signaling cascade not only elucidates the molecular crosstalk between the gut microbiota and its metabolic products and the central immune system but also presents a promising therapeutic target for interventions along the gut–spinal cord axis.157
Bile Acid and the FXR/TGR5 Pathways: Microbial Modulators of Neuroimmune Homeostasis
Bile acids, steroid metabolites synthesized in the liver and modified by the gut microbiota, extend their role beyond lipid digestion to crucial immune and inflammatory regulation.158 Primary bile acids such as cholic acid are transformed by intestinal bacteria into secondary bile acids such as deoxycholic acid and lithocholic acid.159 The composition of this bile acid pool depends heavily on gut microbial communities, including genera such as Lactobacillus, Clostridium, and Bacteroides, which express bile salt hydrolases and 7α-dehydroxylases that modulate bile acid metabolism and signaling.160,161 In SCI animal models, gut dysbiosis and bile acid metabolism disturbances occur alongside altered signaling of bile acid receptors—TGR5, a G protein-coupled receptor, and FXR, a nuclear receptor.162–164 TGR5 activation inhibits spinal microglial and macrophage NF-κB signaling and NLRP3 inflammasome-mediated pyroptosis, reducing secondary inflammation and facilitating recovery.163 FXR regulates bile acid homeostasis, maintains intestinal barrier integrity, modulates the gut microbiota, and suppresses proinflammatory cytokines, thereby mitigating neuroinflammation and neuropathic pain.162 Bile acid derivatives such as tauroursodeoxycholic acid (TUDCA) and ursodeoxycholic acid (UDCA) exert neuroprotective effects by activating antioxidant pathways, inhibiting MAPK signaling, and supporting bone marrow mesenchymal stem cell survival and differentiation, which together promote tissue repair and functional recovery in SCI.165–168 Additionally, bile acids suppress proinflammatory glycolytic pathways (eg, PKM2), further attenuating neuroinflammation.169 Overall, bile acid metabolism and FXR/TGR5-mediated signaling form a key mechanistic axis linking gut microbiota alterations to central nervous system inflammation via the gut–spinal cord axis.
Systemic Pathway Regulatory Mechanisms: Multilevel Crosstalk Among Metabolic, Neural, and Immune Pathways
The gut–spinal cord axis constitutes a complex bidirectional communication system, primarily orchestrated through neural, immune, and metabolic pathways that enable dynamic regulation between the gut and the CNS.33 Under physiological conditions, the gut microbiota interacts with the vagus nerve, the intestinal immune system, and host metabolic networks via microbial metabolites, thereby jointly maintaining neurohomeostasis, immune balance, and metabolic assimilation.170 Following SCI, disruptions in neural regulation, microbial dysbiosis, and compromised intestinal barrier integrity synergistically drive inflammatory cascades and metabolic dysregulation, exacerbating the process of secondary injury.171 Elucidating the integrative neuroimmune-metabolic mechanisms of this axial system could offer novel insights into SCI pathogenesis and lay the theoretical foundation for microbiota-targeted therapeutic strategies (Figure 3).
![]() |
Figure 3 Mechanistic insights into gut microbiota–mediated exacerbation of spinal cord inflammation. Gut dysbiosis acts as an initiating factor that drives pathological changes along the gut–spinal cord axis. Through neural, metabolic, and immune pathways, microbial imbalance disrupts systemic homeostasis and promotes inflammatory responses within the injured spinal cord, ultimately exacerbating neuroinflammation and impeding recovery. Orange arrows indicate neural signaling via the enteric nervous system and vagus nerve; blue arrows represent the transport and effects of microbial metabolites; green arrows indicate immune signaling pathways, including cytokine release and immune cell activation. Abbreviations: GALT, gut-associated lymphoid tissue; MLN, mesenteric lymph nodes; ENS, enteric nervous system.
|
Neural Regulatory Mechanisms
The vagus nerve serves as a central conduit for bidirectional communication within the gut–spinal cord axis. Anatomically, it comprises approximately 80% afferent and 20% efferent fibers, enabling direct transmission of information between the gut and CNS.172 Physiologically, the vagus nerve regulates gastrointestinal motility, peristaltic reflexes, and emptying functions, thereby preserving digestive homeostasis.173 It is also sensitive to changes in the intestinal environment, as it can detect microbial metabolites such as SCFAs, γ-aminobutyric acid, and 5-HT.174 These metabolites interact with receptors expressed on vagal afferents, including free fatty acid receptors (FFARs) and 5-HT receptors (eg, 5-HT3 and 5-HT4), thereby modulating CNS activity.175 Notably, SCFAs such as butyrate significantly increase vagal afferent firing frequency, strengthening gut-to-brain signaling.176 Furthermore, the vagus nerve plays an essential role in immune regulation through the α7 subunit of nicotinic acetylcholine receptors (α7nAChRs), which mediate the cholinergic anti-inflammatory pathway by promoting the shift of M1-polarized Iba-1+/CD86+ microglia to M2-polarized Iba-1+/CD206+ microglia.177,178 Upon detection of peripheral inflammatory signals by afferent fibers, the dorsal motor nucleus integrates this information and activates efferent cholinergic output, thereby suppressing the release of TNF-α, IL-1β, and IL-6 and limiting systemic inflammation.178,179 Pathologically, SCI-induced autonomic dysfunction diminishes vagal tone, leading to gastrointestinal dysmotility, increased permeability, and microbial imbalance.180 Pathogenic metabolites (eg, aberrant α-synuclein) may enter the vagus nerve through a compromised intestinal barrier, initiating CNS inflammatory cascades—a mechanism supported by Parkinson’s disease models.181 Preserving vagal integrity is therefore critical: vagotomy abolishes the anxiolytic effects of probiotics (eg, Bifidobacterium and Lactobacillus) and exacerbates inflammation and endothelial apoptosis after SCI.179,182 In contrast, vagus nerve stimulation mitigates blood–spinal cord barrier disruption and promotes functional neurological recovery.183
In parallel, the ENS—a semi-autonomous network of enteric neurons and glial cells—modulates gut barrier function and mucosal immunity through close interaction with epithelial and immune cells.184 ENS neurons regulate intestinal permeability by influencing tight junction protein expression, mucus secretion, and epithelial cell turnover.185 Moreover, they can sense microbial signals and release neuropeptides such as vasoactive intestinal peptide (VIP) and calcitonin gene-related peptide (CGRP), which exert anti-inflammatory effects locally.186 Enteric glial cells also contribute to intestinal homeostasis by producing cytokines and neurotrophic factors that influence immune cell behavior and epithelial repair.187 Disruption of these neuroimmune interactions after SCI may aggravate gut dysbiosis, barrier dysfunction, and systemic inflammatory responses.53 Overall, neural circuits such as the vagus nerve and ENS function as central integrators of neural, immune, and metabolic signals, playing pivotal roles in maintaining gut–CNS homeostasis and modulating the pathophysiological progression of SCI.
Metabolic Regulatory Mechanisms
The gut microbiota plays a critical role in regulating host energy metabolism and lipid homeostasis, all of which are profoundly disrupted following SCI.71,188 Patients with paraplegia or quadriplegia often exhibit features of metabolic syndrome, including visceral adiposity and insulin resistance, which are consistent with the metabolic disturbances induced by microbiota transplantation in animal models.64 Specific alterations in microbial composition are closely associated with these metabolic abnormalities. For example, Yu et al60 reported that an enrichment of Synergistetes may contribute to increased susceptibility to metabolic dysregulation in CTSCI patients. Synergistetes is an opportunistic bacterial phylum implicated in the pathogenesis of periodontitis and peri-implantitis,189 and previous studies have shown positive correlations between Synergistetes abundance and cholesterol, nicotinic acid, and selenium intake.190 Changes in Synergistetes levels, such as increased low-density lipoprotein (LDL) and decreased high-density lipoprotein (HDL) cholesterol, may be linked to dysregulated lipid profiles in SCI patients.191 Conversely, nicotinamide derivatives have demonstrated neuroprotective effects in SCI models [38]. Heller et al192 and Seelig et al193 reported that patients with traumatic SCI presented with elevated serum selenium (Se) at admission, followed by a rapid decline within 24 hours. This redistribution of Se may facilitate anti-inflammatory responses and neuronal regeneration, contributing to improved clinical outcomes. Moreover, SCI-associated dysbiosis—characterized by reduced Clostridiales and increased Akkermansia—correlates with metabolic inflammation and aberrant glucose and lipid metabolism. Notably, the role of Akkermansia appears context dependent: while it is typically beneficial in metabolic disorders owing to its low abundance,194,195 its function in the SCI setting may differ owing to gut environmental adaptations.196 Additional dysbiosis, such as increased Bifidobacterium and reduced Prevotella, along with altered systemic metabolites (eg, decreased HDL), further supports the notion that the gut microbiota modulates neuroinflammation through complex metabolic pathways.59,60,64 In summary, microbiota dysregulation following SCI is intricately linked to systemic metabolic alterations, ultimately contributing to the amplification of neuroinflammatory processes.
Immunomodulatory Mechanisms
The gut microbiota plays a pivotal role in maintaining immune homeostasis. As the largest immune organ, GALT comprises approximately 70–80% of the body’s immune cells, encompassing both innate and adaptive immune populations.197 Commensal microbes present antigenic epitopes to immune cells within the GALT via pattern recognition receptors (PRRs), subsequently activating downstream signaling pathways to orchestrate the balance of local and systemic immune responses.198 Notably, the sympathetic innervation of the GALT positions it as a critical interface for neuroimmune communication. SCI-induced dysregulation of sympathetic tone can disrupt the immunological microenvironment of the GALT, impairing mucosal immune surveillance over the intestinal microbiota.199 This autonomic dysfunction initiates a vicious cycle of microbial dysbiosis and immune disarray, ultimately compromising host immune defense and leading to increased susceptibility to infections and impaired tissue repair.103 Rong et al112 elucidated the molecular basis of microbiota–immune axis disruption in a murine model of SCI. Postinjury dysbiosis was accompanied by the upregulation of proinflammatory cytokines (TNF-α, IL-1β, and IL-6), which was driven primarily by sustained activation of the TLR4/MyD88 signaling cascade. Concurrently, the suppression of anti-inflammatory mediators (IL-4, TGF-β, and IL-10) exacerbates the imbalance of immune regulatory networks. Importantly, SCI-induced gastrointestinal motility disorders further impair microbiota–immune interactions, leading to dysfunction of immune organs such as mesenteric lymph nodes (mLNs). This is characterized by aberrant activation of T and B lymphocytes, which in turn aggravate neuroinflammation and hinder functional recovery.200 As a key hub for intestinal immune surveillance, the mLN system utilizes dendritic cells (DCs) to continuously sample gut antigens and maintain a delicate equilibrium between immune tolerance and activation under physiological conditions.201 These findings reveal how localized intestinal immune disruption can propagate systemic inflammation and exacerbate neural tissue damage, providing a rationale for multitargeted therapeutic strategies.
As central modulators of immune homeostasis, the gut microbiota can profoundly influence the differentiation and function of various lymphocyte subsets, including γδ T cells and regulatory T (Treg) cells—both of which play key roles in SCI pathogenesis.121,202,203 Pang et al65 reported associations between gut microbial taxa and lymphocyte subsets in SCI patients. Specifically, five bacterial genera—Lachnospiraceae UCG-008, Lachnoclostridium 12, Tyzzerella 3, Trueperella, and Ruminococcaceae UCG-002—were correlated with T lymphocyte subpopulations and NK cells. Additionally, Prevotella 9, Lachnospiraceae NC2004 group, Veillonella, and Sutterella were positively correlated with B cells, whereas Clostridium and Akkermansia were negatively correlated. Moreover, Roseburia and Ruminococcaceae UCG-003 were positively associated with cytokine-induced killer (CIK) cells. These observations underscore the close interdependence between the gut microbial composition and immune cell distribution, where dysbiosis may directly precipitate immune surveillance failure and uncontrolled inflammation.204,205 Mechanistically, microbial homeostasis is critical for maintaining basophil populations and guiding T-cell differentiation.206 Certain strains, such as Lactobacillus reuteri, can modulate tryptophan metabolism to induce the generation of CD4+CD8αα+ double-positive T cells in the intestinal epithelium, thereby reprogramming immunoregulatory pathways and enhancing anti-inflammatory responses.141,207 While Treg cells are traditionally considered protective through peripheral immunosuppression, their functionality is highly dependent on a dynamically balanced intestinal immune milieu.208,209 Treg cells can directly influence the activation state of γδ T cells,210 which are central players in the gut–spinal cord immune axis. Owing to their unique migratory capacity across organ systems, γδ T cells may serve as a crucial link between intestinal and spinal pathophysiology.121 Neuroinflammation following SCI is largely driven by the proinflammatory cytokine IL-17, which recruits neutrophils and other inflammatory cells to exacerbate tissue damage and neurodegeneration.211,212 Therapeutically targeting IL-17 signaling—either by inhibiting its production or function—significantly alleviates pathological inflammation and promotes neurofunctional recovery.213 Activated γδ T cells, as a primary source of IL-17, proliferate and secrete both IL-17 and IFN-γ, thereby amplifying inflammatory cascades.214–216 The immunopathology of SCI appears to be predominantly fueled by the infiltration of peripheral γδ T cells, as these cells rarely reside in the uninjured CNS, suggesting that peripheral organs—especially the gut—may regulate cross-organ injury responses by circulating immune networks.217,218 Indeed, analogous to the “gut–brain axis” described in ischemic stroke models,219 SCI prompts the targeted recruitment of gut-derived γδ T cells to the injury site, a process tightly regulated by the composition of the gut microbiota.220 The CCL2/CCR2 signaling axis has been identified as a key mechanism mediating γδ T-cell homing to spinal cord lesions, suggesting that the CCL2/CCR2 signaling axis is a promising therapeutic target for SCI.202 Taken together, these findings not only highlight potential immunotherapeutic strategies for SCI but also emphasize the fundamental role of the gut–spinal cord axis in the immunopathogenesis of neural trauma.
Therapeutic Strategies Targeting the Gut–Spinal Cord Axis
As described above, gut dysbiosis following SCI contributes to the exacerbation of secondary spinal inflammation via multiple pathways. Consequently, modulation of the gut microbiota represents a promising therapeutic avenue to ameliorate the inflammatory microenvironment of the injured spinal cord—an approach termed gut microbial therapy. Current interventions, such as probiotics and FMT, have demonstrated potential in the treatment of SCI (Figure 4).
![]() |
Figure 4 Therapeutic Strategies Targeting the Gut–Spinal Cord Axis. Therapeutic strategies such as probiotic supplementation and FMT aim to restore gut microbial balance and reestablish gut–spinal cord axis homeostasis. By modulating systemic signaling and reducing neuroinflammation, these interventions offer promising avenues for improving outcomes after SCI. Purple arrows illustrate the gut microbiota–metabolite–spinal cord signaling axis, which promotes anti-inflammatory responses in the injured spinal cord.
|
Probiotics
Probiotics are defined as live microorganisms that, when administered in adequate amounts, confer health benefits to the host. When these effects extend to behavior and mental states, such microbes are further classified as “psychobiotics”.10,221,222 Among the most studied genera are Lactobacillus, Bifidobacterium, Streptococcus, Escherichia, Enterococcus, and Clostridium butyricum. Much of the research in this field has focused on the influence of these microbes on the nervous, endocrine, humoral, and immune systems, particularly in the context of anxiety, stress, and depression.223–225 Through modulation of the gut microbial composition and immune homeostasis, probiotics have been shown to promote host health.226 In the context of SCI, probiotic administration has been shown to normalize the gut microbial architecture and mitigate the detrimental feedback loop between intestinal barrier damage and central neuroinflammation via bidirectional signaling along the microbiota–CNS axis.227 Experimental models indicate that certain strains can normalize microbial architecture, repair intestinal barrier integrity, and attenuate central neuroinflammation via bidirectional microbiota–CNS signaling. For example, oral administration of the multistrain formulation VSL#3 (5 × 109 CFU/day for 35 days) improved locomotor coordination, reduced lesion volume, and enhanced myelin preservation in SCI model mice.93 Similarly, local injection of conditioned medium from Lactobacillus rhamnosus GG (30 mg/kg, post-injury) promoted motor function recovery and microglial M2 polarization, likely through inhibition of the NF-κB pathway.228 Another strain, Limosilactobacillus reuteri DSM 17938, improved spinal pathology by reducing M1 microglial activation and inflammatory cytokine production, while restoring intestinal barrier function via AhR signaling and tight junction reinforcement.229 These findings underscore the potential of probiotics not only as microbial modulators but also as immunoregulatory and barrier-protective agents in SCI recovery.
Clinical data further support that such interventions can significantly increase patients’ health indices.230,231 In the ECLISP randomized controlled trial, daily supplementation with Lactobacillus casei Shirota (≥6.5 × 109 CFU) reduced the incidence of antibiotic-associated diarrhea (AAD) in SCI patients who regularly used proton pump inhibitors, suggesting a protective effect in at-risk populations.232 However, no overall benefit was observed in unselected patients, and a separate study revealed that a commercial probiotic cocktail failed to improve functional outcomes and even delayed microbiota normalization.233 These inconsistencies highlight the importance of strain specificity, host conditions, and treatment context. While no major adverse effects have been reported, rigorous evaluation of safety, including donor screening and patient immune status, remains essential.234 Overall, probiotics represent promising adjuncts in SCI management, but further investigations are needed to refine strain selection, dosage regimens, and clinical applicability.
In addition to live microbial supplementation, prebiotics—nondigestible dietary fibers such as inulin and oat bran—support gut health by selectively promoting the growth of beneficial microbes and enhancing the metabolic activity of commensal bacteria.235 While they do not directly inhibit pathogenic species, prebiotics improve microbial diversity and contribute to intestinal barrier integrity, indirectly modulating host immunity.236 Synbiotics, defined as synergistic combinations of probiotics and prebiotics, aim to maximize microbial engraftment and functionality.237 Although most studies on SCI focus on probiotics alone, early-stage clinical evidence suggests the feasibility and safety of both prebiotic and synbiotic interventions in individuals with SCI.72 A randomized crossover trial in elite wheelchair athletes with SCI demonstrated the feasibility and safety of both probiotic (3 g/day) and prebiotic (5 g/day oat bran) supplementation over a 4-week period. Although both interventions were well tolerated, probiotics elicited greater reductions in systemic inflammatory markers and improved gut microbiota diversity.231 These findings suggest that synbiotics, which combine probiotics and their preferred substrates, may offer additive or synergistic benefits by enhancing microbial colonization and immune regulation. Despite encouraging preclinical and pilot data, the therapeutic utility of prebiotics and synbiotics in SCI requires further investigation to clarify strain–substrate compatibility, optimal dosing, and long-term efficacy across diverse patient populations.
Fmt
FMT, an emerging therapeutic strategy, aims to restore the gut microbial balance and reshape metabolite profiles to regulate host immunity and neurological function. It has shown promise across a range of CNS disorders.238,239 In SCI models, FMT has been shown to reconstruct dysbiotic microbial communities and promote neuronal survival and axonal regeneration. For example, Jing et al79 demonstrated that probiotic transplantation in SCI mice significantly elevated motor-evoked potential (MEP) amplitudes and increased the expression of NeuN+ neurons, NF-200+ axons, and synaptic proteins, indicating the restoration of neural function. Rodenhouse et al102 further confirmed that preoperative FMT could prevent post-SCI functional deficits and reverse antibiotic-induced dysbiosis. Mechanistically, FMT appears to modulate the gut–immune–brain axis to ameliorate the chronic inflammatory microenvironment, for example, by suppressing proinflammatory IL-1β/NF-κB signaling and restoring neurotrophic factor expression (eg, BDNF, NT-3, NGF), thereby enhancing synaptic plasticity and neurorepair.240,241 Moreover, Jing et al242 reported that FMT inhibited the overactivation of microglia and macrophages in the CNS, promoted their shift toward anti-inflammatory phenotypes, and reduced aberrant astrocyte activation. Additionally, microbial-derived metabolites such as β-alanine were shown to inhibit matrix metalloproteinase-9 (MMP-9) activity, thus preserving the integrity of the blood‒spinal cord barrier and mitigating oxidative stress and secondary damage. Notably, FMT has shown therapeutic benefits in a range of CNS diseases, including Parkinson’s disease, Alzheimer’s disease, stroke, autism spectrum disorders, and multiple sclerosis.243–248 For example, germ-free mice receiving FMT from Parkinson’s disease patients developed motor deficits, whereas healthy donor FMT reduced neuroinflammation and TLR/TNF-α signaling in Parkinson’s disease mice, demonstrating a neuroprotective effect.243,244 In APP/PS1 transgenic mice, FMT improved cognitive function and synaptic plasticity while attenuating neuroinflammation, thereby alleviating Alzheimer’s disease-like pathology.245 In a middle cerebral artery occlusion (MCAO) rat model, FMT reduced infarct volume, alleviated cerebral edema, and lowered blood lipid levels.247
Despite these encouraging results, the clinical translation of FMT for SCI remains challenging. One major concern is safety: although most adverse events (eg, abdominal discomfort, bloating) are mild and self-limiting, serious complications such as transmission of multidrug-resistant organisms (eg, drug-resistant Escherichia coli) have been reported, prompting FDA safety alerts.249–251 Rigorous donor screening and standardized protocols are therefore essential. Encouragingly, a recent study in SCI patients colonized with carbapenemase-producing Enterobacteriaceae or vancomycin-resistant enterococci revealed that FMT achieved decolonization in most cases without adverse effects, supporting its feasibility in high-risk populations.252 Collectively, these findings provide compelling evidence for the therapeutic potential of FMT in SCI. Nonetheless, further research is needed to refine treatment protocols, ensure safety, and explore long-term efficacy across diverse patient populations.
Conclusion and Future Perspectives
SCI is a complex neurological disorder characterized by localized neuroinflammation, immune dysregulation, and dysfunction of distal organs. In recent years, the regulatory role of the gut microbiota in SCI has garnered increasing attention. The emerging concept of the “gut–spinal cord axis” offers a novel framework for understanding the pathophysiology of SCI and developing new therapeutic strategies. This review summarizes the dynamic alterations in the gut microbiota following SCI, elaborates on their critical roles in modulating inflammation, promoting neuroregeneration, and maintaining metabolic homeostasis, and explores the underlying mechanisms through which microbiota-derived metabolites—such as LPS, SCFAs and tryptophan metabolites—affect the spinal microenvironment. Additionally, we systematically evaluated microbiota-targeted interventions, including probiotics and FMT, highlighting their therapeutic potential and current progress in SCI recovery. Despite promising preclinical and early clinical evidence supporting the role of gut microbiota modulation in SCI, several key questions remain unresolved. The precise mechanisms of action require further elucidation, and microbial interventions must be optimized and rigorously tested in translational settings to advance precision therapy in SCI.
As research into the gut–spinal cord axis has progressed, the critical influence of the gut microbiota on SCI pathogenesis and recovery has become increasingly evident. However, current investigations face considerable challenges, including interindividual variability in microbial composition, the complexity of microbiota–host interactions, and the need for precise modulation strategies. Future research should prioritize the following areas. First, the integration of multiomics technologies—particularly metagenomics, metabolomics, and transcriptomics—will be essential for comprehensively characterizing the dynamic shifts in the gut microbiota after SCI and unraveling how metabolic–immune–neural networks shape spinal homeostasis. Second, to address limitations associated with existing microbial interventions—such as strain selection, dosage control, and individual variability—synthetic biology and bioengineering approaches could be harnessed to develop next-generation, highly targeted and stable microbiota-based therapies. Moreover, the neuroprotective and immunomodulatory effects of microbiota-derived metabolites, particularly SCFAs and tryptophan catabolites, warrant further investigation. These molecules may serve not only as biomarkers for disease progression and therapeutic response but also as the basis for novel metabolite-based treatment strategies. Finally, translating microbiome-based interventions into clinical practice will require large-scale, longitudinal studies to evaluate long-term efficacy and safety, alongside the application of artificial intelligence and big data analytics to personalize treatment protocols. We suggest that human cohort studies include follow-up durations of at least 12 months to capture stable microbial reconstitution and delayed neurological responses. In conclusion, with the continued convergence of microbiology, neuroscience, and artificial intelligence, microbiota modulation holds promise as an innovative and precise therapeutic approach for SCI. Harnessing this axis may lead to more effective strategies for improving patient outcomes and advancing the field of neuroregenerative medicine.
Consent for publication
All the authors agree with the publication of this work.
Author Contributions
All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.
Funding
This work was supported by the Xi’an Innovation Capability Strong Foundation Plan – Medical Research Project (22XYJ0002).
Disclosure
The authors report no conflicts of interest in this work.
References
1. Ahuja CS, Wilson JR, Nori S, et al. Traumatic spinal cord injury. Nat Rev Dis Primers. 2017;3:17018. doi:10.1038/nrdp.2017.18
2. Quadri SA, Farooqui M, Ikram A, et al. Recent update on basic mechanisms of spinal cord injury. Neurosurg Rev. 2020;43(2):425–441. doi:10.1007/s10143-018-1008-3
3. Kong X, Gao J. Macrophage polarization: a key event in the secondary phase of acute spinal cord injury. J Cell Mol Med. 2017;21(5):941–954. doi:10.1111/jcmm.13034
4. Perrouin-Verbe B, Lefevre C, Kieny P, Gross R, Reiss B, Le Fort M. Spinal cord injury: a multisystem physiological impairment/dysfunction. Rev Neurol. 2021;177(5):594–605. doi:10.1016/j.neurol.2021.02.385
5. Sweis R, Biller J. Systemic complications of spinal cord injury. Curr Neurol Neurosci Rep. 2017;17(2):8. doi:10.1007/s11910-017-0715-4
6. Hu X, Xu W, Ren Y, et al. Spinal cord injury: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther. 2023;8(1):245. doi:10.1038/s41392-023-01477-6
7. Badhiwala JH, Wilson JR, Witiw CD, et al. The influence of timing of surgical decompression for acute spinal cord injury: a pooled analysis of individual patient data. Lancet Neurol. 2021;20(2):117–126. doi:10.1016/S1474-4422(20)30406-3
8. Huang H, Young W, Skaper S, et al. Clinical neurorestorative therapeutic guidelines for spinal cord injury (IANR/CANR version 2019). J Orthop Translat. 2020;20:14–24. doi:10.1016/j.jot.2019.10.006
9. Huang H, Chen L, Chopp M, et al. The 2020 yearbook of neurorestoratology. J Neurorestoratol. 2021;9(1):1–12. doi:10.26599/JNR.2021.9040002
10. Cryan JF, O’Riordan KJ, Cowan C, et al. The microbiota-gut-brain axis. Physiol Rev. 2019;99(4):1877–2013. doi:10.1152/physrev.00018.2018
11. Agirman G, Hsiao EY. SnapShot: the microbiota-gut-brain axis. Cell. 2021;184(9):2524. doi:10.1016/j.cell.2021.03.022
12. Mayer EA, Nance K, Chen S. The gut-brain axis. Annu Rev Med. 2022;73:439–453. doi:10.1146/annurev-med-042320-014032
13. Bannerman CA, Douchant K, Sheth PM, Ghasemlou N. The gut-brain axis and beyond: microbiome control of spinal cord injury pain in humans and rodents. Neurobiol Pain. 2021;9:100059.
14. Zhu L, Wang F, Xing J, et al. Modulatory effects of gut microbiota on innate and adaptive immune responses following spinal cord injury. Exp Neurol. 2024;379:114866.
15. Yuan B, Lu X, Wu Q. Gut microbiota and acute central nervous system injury: a new target for therapeutic intervention. Front Immunol. 2021;12:800796. doi:10.3389/fimmu.2021.800796
16. Guha L, Agnihotri TG, Jain A, Kumar H. Gut microbiota and traumatic central nervous system injuries: insights into pathophysiology and therapeutic approaches. Life Sci. 2023;334:122193. doi:10.1016/j.lfs.2023.122193
17. Singh S, Sahu K, Singh C, Singh A. Lipopolysaccharide induced altered signaling pathways in various neurological disorders. Naunyn Schmiedebergs Arch Pharmacol. 2022;395(3):285–294. doi:10.1007/s00210-021-02198-9
18. Liang W, Han B, Hai Y, et al. The role of microglia/macrophages activation and TLR4/NF-κB/MAPK pathway in distraction spinal cord injury-induced inflammation. Front Cell Neurosci. 2022;16:926453. doi:10.3389/fncel.2022.926453
19. Lukacova N, Kisucka A, Kiss Bimbova K, et al. Glial-neuronal interactions in pathogenesis and treatment of spinal cord injury. Int J Mol Sci. 2021;22(24):13577. doi:10.3390/ijms222413577
20. Kigerl KA, Mostacada K, Popovich PG. Gut microbiota are disease-modifying factors after traumatic spinal cord injury. Neurotherapeutics. 2018;15(1):60–67. doi:10.1007/s13311-017-0583-2
21. Alizadeh A, Dyck SM, Karimi-Abdolrezaee S. Traumatic spinal cord injury: an overview of pathophysiology, models and acute injury mechanisms. Front Neurol. 2019;10:282.
22. Sterner RC, Sterner RM. Immune response following traumatic spinal cord injury: pathophysiology and therapies. Front Immunol. 2023;13:1084101. doi:10.3389/fimmu.2022.1084101
23. Fan B, Wei Z, Yao X, et al. Microenvironment imbalance of spinal cord injury. Cell Transplant. 2018;27(6):853–866. doi:10.1177/0963689718755778
24. Anjum A, Yazid MDI, Fauzi Daud M, et al. Spinal cord injury: pathophysiology, multimolecular interactions, and underlying recovery mechanisms. Int J Mol Sci. 2020;21(20):7533. doi:10.3390/ijms21207533
25. Chio J, Xu KJ, Popovich P, David S, Fehlings MG. Neuroimmunological therapies for treating spinal cord injury: evidence and future perspectives. Exp Neurol. 2021;341:113704. doi:10.1016/j.expneurol.2021.113704
26. Bajwa E, Pointer CB, Klegeris A. The role of mitochondrial damage-associated molecular patterns in chronic neuroinflammation. Mediators Inflamm. 2019;2019:4050796. doi:10.1155/2019/4050796
27. Kigerl KA, de Rivero VJ, Dietrich WD, Popovich PG, Keane RW. Pattern recognition receptors and central nervous system repair. Exp Neurol. 2014;258:5–16. doi:10.1016/j.expneurol.2014.01.001
28. Ahuja CS, Nori S, Tetreault L, et al. Traumatic spinal cord injury-repair and regeneration. Neurosurgery. 2017;80(3S):S9–S22. doi:10.1093/neuros/nyw080
29. Zhang Y, Yang S, Liu C, Han X, Gu X, Zhou S. Deciphering glial scar after spinal cord injury. Burns Trauma. 2021;9:b35. doi:10.1093/burnst/tkab035
30. Hellenbrand DJ, Quinn CM, Piper ZJ, Morehouse CN, Fixel JA, Hanna AS. Inflammation after spinal cord injury: a review of the critical timeline of signaling cues and cellular infiltration. J Neuroinflammation. 2021;18(1):284. doi:10.1186/s12974-021-02337-2
31. Zivkovic S, Ayazi M, Hammel G, Ren Y. For better or for worse: a look into neutrophils in traumatic spinal cord injury. Front Cell Neurosci. 2021;15:648076. doi:10.3389/fncel.2021.648076
32. Li C, Wu Z, Zhou L, et al. Temporal and spatial cellular and molecular pathological alterations with single-cell resolution in the adult spinal cord after injury. Signal Transduct Target Ther. 2022;7(1):65. doi:10.1038/s41392-022-00885-4
33. Ortega MA, Fraile-Martinez O, Garcia-Montero C, et al. A comprehensive look at the psychoneuroimmunoendocrinology of spinal cord injury and its progression: mechanisms and clinical opportunities. Mil Med Res. 2023;10(1):26. doi:10.1186/s40779-023-00461-z
34. Li H, Wang P, Tang L, et al. Distinct polarization dynamics of microglia and infiltrating macrophages: a novel mechanism of spinal cord ischemia/reperfusion injury. J Inflamm Res. 2021;14:5227–5239. doi:10.2147/JIR.S335382
35. Kopper TJ, Gensel JC. Myelin as an inflammatory mediator: myelin interactions with complement, macrophages, and microglia in spinal cord injury. J Neurosci Res. 2018;96(6):969–977. doi:10.1002/jnr.24114
36. Xu L, Ye X, Wang Q, et al. T-cell infiltration, contribution and regulation in the central nervous system post-traumatic injury. Cell Prolif. 2021;54(8):e13092. doi:10.1111/cpr.13092
37. Maheshwari S, Dwyer LJ, Sirbulescu RF. Inflammation and immunomodulation in central nervous system injury – B cells as a novel therapeutic opportunity. Neurobiol Dis. 2023;180:106077. doi:10.1016/j.nbd.2023.106077
38. Bannerman CA, Douchant K, Segal JP, et al. Spinal cord injury in mice affects central and peripheral pathology in a severity-dependent manner. Pain. 2022;163(6):1172–1185. doi:10.1097/j.pain.0000000000002471
39. Pruss H, Tedeschi A, Thiriot A, et al. Spinal cord injury-induced immunodeficiency is mediated by a sympathetic-neuroendocrine adrenal reflex. Nat Neurosci. 2017;20(11):1549–1559. doi:10.1038/nn.4643
40. Maldonado-Bouchard S, Peters K, Woller SA, et al. Inflammation is increased with anxiety- and depression-like signs in a rat model of spinal cord injury. Brain Behav Immun. 2016;51:176–195. doi:10.1016/j.bbi.2015.08.009
41. Besecker EM, Deiter GM, Pironi N, Cooper TK, Holmes GM. Mesenteric vascular dysregulation and intestinal inflammation accompanies experimental spinal cord injury. Am J Physiol Regul Integr Comp Physiol. 2017;312(1):R146–R156. doi:10.1152/ajpregu.00347.2016
42. Chikuda H, Yasunaga H, Takeshita K, et al. Mortality and morbidity after high-dose methylprednisolone treatment in patients with acute cervical spinal cord injury: a propensity-matched analysis using a nationwide administrative database. Emerg Med J. 2014;31(3):201–206. doi:10.1136/emermed-2012-202058
43. Margo TE, McMullin PR, Kaddouh F. An interval of clinically silent gastrointestinal bleed in dysautonomic spinal cord injury: a case report. Bmc Neurol. 2023;23(1):70. doi:10.1186/s12883-023-03114-9
44. Durney P, Stillman M, Montero W, Goetz L. A primary care provider’s guide to neurogenic bowel dysfunction in spinal cord injury. Top Spinal Cord Inj Rehabil. 2020;26(3):172–176. doi:10.46292/sci2603-172
45. Cheng J, Wang X, Guo J, et al. Effects of electroacupuncture on the daily rhythmicity of intestinal movement and circadian rhythmicity of colonic per2 expression in rats with spinal cord injury. Biomed Res Int. 2016;2016:9860281. doi:10.1155/2016/9860281
46. Qi Z, Middleton JW, Malcolm A. Bowel dysfunction in spinal cord injury. Curr Gastroenterol Rep. 2018;20(10):47. doi:10.1007/s11894-018-0655-4
47. Emmanuel A. Neurogenic bowel dysfunction. F1000Research. 2019;8:1800. doi:10.12688/f1000research.20529.1
48. Hakim S, Gaglani T, Cash BD. Neurogenic bowel dysfunction: the impact of the central nervous system in constipation and fecal incontinence. Gastroenterol Clin North Am. 2022;51(1):93–105. doi:10.1016/j.gtc.2021.10.006
49. Gungor B, Adiguzel E, Gursel I, Yilmaz B, Gursel M. Intestinal microbiota in patients with spinal cord injury. Plos One. 2016;11(1):e145878. doi:10.1371/journal.pone.0145878
50. Rueckert H, Ganz J. How to heal the gut’s brain: regeneration of the enteric nervous system. Int J Mol Sci. 2022;23(9):4799. doi:10.3390/ijms23094799
51. White AR, Holmes GM. Anatomical and functional changes to the colonic neuromuscular compartment after experimental spinal cord injury. J Neurotrauma. 2018;35(9):1079–1090. doi:10.1089/neu.2017.5369
52. White AR, Holmes GM. Investigating neurogenic bowel in experimental spinal cord injury: where to begin?. Neural Regen Res. 2019;14(2):222–226. doi:10.4103/1673-5374.244779
53. Hamilton AM, Blackmer-Raynolds L, Li Y, et al. Diet-microbiome interactions promote enteric nervous system resilience following spinal cord injury. NPJ Biofilms Microbiomes. 2024;10(1):75. doi:10.1038/s41522-024-00556-y
54. Faber W, Stolwijk-Swuste J, van Ginkel F, et al. Faecal microbiota in patients with neurogenic bowel dysfunction and spinal cord injury or multiple sclerosis—A systematic review. J Clin Med. 2021;10(8):1598. doi:10.3390/jcm10081598
55. Savic G, DeVivo MJ, Frankel HL, Jamous MA, Soni BM, Charlifue S. Causes of death after traumatic spinal cord injury-a 70-year British study. Spinal Cord. 2017;55(10):891–897. doi:10.1038/sc.2017.64
56. Ture SD, Ozkaya G, Sivrioglu K. Relationship between neurogenic bowel dysfunction severity and functional status, depression, and quality of life in individuals with spinal cord injury. J Spinal Cord Med. 2023;46(3):424–432. doi:10.1080/10790268.2021.2021043
57. Khadour FA, Khadour YA, Xu J, Meng L, Cui L, Xu T. Effect of neurogenic bowel dysfunction symptoms on quality of life after a spinal cord injury. J Orthop Surg Res. 2023;18(1):458. doi:10.1186/s13018-023-03946-8
58. Johns JS, Krogh K, Ethans K, Chi J, Querée M, Eng JJ. Pharmacological Management of neurogenic bowel dysfunction after spinal cord injury and multiple sclerosis: a systematic review and clinical implications. J Clin Med. 2021;10(4):882. doi:10.3390/jcm10040882
59. Zhang C, Zhang W, Zhang J, et al. Gut microbiota dysbiosis in male patients with chronic traumatic complete spinal cord injury. J Transl Med. 2018;16(1):353.
60. Yu B, Qiu H, Cheng S, et al. Profile of gut microbiota in patients with traumatic thoracic spinal cord injury and its clinical implications: a case-control study in a rehabilitation setting. Bioengineered. 2021;12(1):4489–4499. doi:10.1080/21655979.2021.1955543
61. Tang Q, Jin G, Wang G, et al. Current sampling methods for gut microbiota: a call for more precise devices. Front Cell Infect Microbiol. 2020;10:151. doi:10.3389/fcimb.2020.00151
62. Muhamad Rizal NS, Neoh H, Ramli R, et al. Advantages and limitations of 16S rRNA next-generation sequencing for pathogen identification in the diagnostic microbiology laboratory: perspectives from a middle-income country. Diagnostics. 2020;10(10):816. doi:10.3390/diagnostics10100816
63. Valido E, Bertolo A, Frankl GP, et al. Systematic review of the changes in the microbiome following spinal cord injury: animal and human evidence. Spinal Cord. 2022;60(4):288–300. doi:10.1038/s41393-021-00737-y
64. Zhang C, Jing Y, Zhang W, et al. Dysbiosis of gut microbiota is associated with serum lipid profiles in male patients with chronic traumatic cervical spinal cord injury. Am J Transl Res. 2019;11(8):4817–4834.
65. Pang R, Wang J, Xiong Y, et al. Relationship between gut microbiota and lymphocyte subsets in Chinese Han patients with spinal cord injury. Front Microbiol. 2022;13:986480. doi:10.3389/fmicb.2022.986480
66. Li J, Van Der Pol W, Eraslan M, et al. Comparison of the gut microbiome composition among individuals with acute or long-standing spinal cord injury vs. able-bodied controls. J Spinal Cord Med. 2022;45(1):91–99. doi:10.1080/10790268.2020.1769949
67. O’Connor G, Jeffrey E, Madorma D, et al. Investigation of microbiota alterations and intestinal inflammation post-spinal cord injury in rat model. J Neurotrauma. 2018;35(18):2159–2166. doi:10.1089/neu.2017.5349
68. Schmidt E, Torres-Espin A, Raposo P, et al. Fecal transplant prevents gut dysbiosis and anxiety-like behaviour after spinal cord injury in rats. Plos One. 2020;15(1):e226128. doi:10.1371/journal.pone.0226128
69. Schmidt EKA, Raposo PJF, Madsen KL, Fenrich KK, Kabarchuk G, Fouad K. What makes a successful donor? Fecal transplant from anxious-like rats does not prevent spinal cord injury-induced dysbiosis. Biology. 2021;10(4):254. doi:10.3390/biology10040254
70. Lin R, Xu J, Ma Q, et al. Alterations in the fecal microbiota of patients with spinal cord injury. Plos One. 2020;15(8):e236470.
71. Kong G, Zhang W, Zhang S, et al. The gut microbiota and metabolite profiles are altered in patients with spinal cord injury. Mol Brain. 2023;16(1):26. doi:10.1186/s13041-023-01014-0
72. Glisic M, Flueck JL, Ruettimann B, et al. The feasibility of a crossover, randomized controlled trial design to assess the effect of probiotics and prebiotics on health of elite Swiss para-athletes: a study protocol. Pilot Feasibility Stud. 2022;8(1):94. doi:10.1186/s40814-022-01048-6
73. Kulecka M, Fraczek B, Mikula M, et al. The composition and richness of the gut microbiota differentiate the top Polish endurance athletes from sedentary controls. Gut Microbes. 2020;11(5):1374–1384. doi:10.1080/19490976.2020.1758009
74. Zang Y, Lai X, Li C, Ding D, Wang Y, Zhu Y. The role of gut microbiota in various neurological and psychiatric disorders-an evidence mapping based on quantified evidence. Mediators Inflamm. 2023;2023:5127157. doi:10.1155/2023/5127157
75. Koliada A, Moseiko V, Romanenko M, et al. Sex differences in the phylum-level human gut microbiota composition. Bmc Microbiol. 2021;21(1):131. doi:10.1186/s12866-021-02198-y
76. Tyler PT, Grandhi R. Gut microbiota and neurologic diseases and injuries. Adv Exp Med Biol. 2020;1238:73–91.
77. Yin Y, Yang T, Tian Z, et al. Progress in the investigation of the Firmicutes/Bacteroidetes ratio as a potential pathogenic factor in ulcerative colitis. J Med Microbiol. 2025;74(1). doi:10.1099/jmm.0.001966
78. Kang J, Sun Z, Li X, et al. Alterations in gut microbiota are related to metabolite profiles in spinal cord injury. Neural Regen Res. 2023;18(5):1076. doi:10.4103/1673-5374.355769
79. Jing Y, Yu Y, Bai F, et al. Effect of fecal microbiota transplantation on neurological restoration in a spinal cord injury mouse model: involvement of brain-gut axis. Microbiome. 2021;9(1):59. doi:10.1186/s40168-021-01007-y
80. Myers SA, Gobejishvili L, Saraswat OS, et al. Following spinal cord injury, PDE4B drives an acute, local inflammatory response and a chronic, systemic response exacerbated by gut dysbiosis and endotoxemia. Neurobiol Dis. 2019;124:353–363. doi:10.1016/j.nbd.2018.12.008
81. Du J, Zayed AA, Kigerl KA, Zane K, Sullivan MB, Popovich PG. Spinal Cord injury changes the structure and functional potential of gut bacterial and viral communities. Msystems. 2021;6(3):e1320–e1356. doi:10.1128/mSystems.01356-20
82. Hsu C, Ghannoum M, Cominelli F, Martino LD. Mycobiome and inflammatory bowel disease: role in disease pathogenesis, current approaches and novel nutritional-based therapies. Inflamm Bowel Dis. 2023;29(3):470–479. doi:10.1093/ibd/izac156
83. Wu Y, Cheng R, Lin H, et al. Gut virome and its implications in the pathogenesis and therapeutics of inflammatory bowel disease. Bmc Med. 2025;23(1):183. doi:10.1186/s12916-025-04016-y
84. Schmidt EKA, Raposo PJF, Torres-Espin A, Fenrich KK, Fouad K. Beyond the lesion site: minocycline augments inflammation and anxiety-like behavior following SCI in rats through action on the gut microbiota. J Neuroinflammation. 2021;18(1):144. doi:10.1186/s12974-021-02123-0
85. Doelman A, Tigchelaar S, McConeghy B, et al. Characterization of the gut microbiome in a porcine model of thoracic spinal cord injury. Bmc Genomics. 2021;22(1):775. doi:10.1186/s12864-021-07979-3
86. Jing Y, Yang D, Bai F, et al. Melatonin treatment alleviates spinal cord injury-induced gut dysbiosis in mice. J Neurotraum. 2019;36(18):2646–2664. doi:10.1089/neu.2018.6012
87. Gur AA, Toren I, Hadar R, et al. Lack of gut microbiome recovery with spinal cord injury rehabilitation. Gut Microbes. 2024;16(1):2309682. doi:10.1080/19490976.2024.2309682
88. Zhang Y, Lang R, Guo S, et al. Intestinal microbiota and melatonin in the treatment of secondary injury and complications after spinal cord injury. Front Neurosci. 2022;16:981772. doi:10.3389/fnins.2022.981772
89. Bazzocchi G, Turroni S, Bulzamini MC, et al. Changes in gut microbiota in the acute phase after spinal cord injury correlate with severity of the lesion. Sci Rep-Uk. 2021;11(1):12743. doi:10.1038/s41598-021-92027-z
90. Lefevre C, Bessard A, Aubert P, et al. Enteric nervous system remodeling in a rat model of spinal cord injury: a pilot study. Neurotrauma Rep. 2020;1(1):125–136. doi:10.1089/neur.2020.0041
91. White AR, Werner CM, Holmes GM. Diminished enteric neuromuscular transmission in the distal colon following experimental spinal cord injury. Exp Neurol. 2020;331:113377. doi:10.1016/j.expneurol.2020.113377
92. Wallace DJ, Sayre NL, Patterson TT, Nicholson SE, Hilton D, Grandhi R. Spinal cord injury and the human microbiome: beyond the brain-gut axis. Neurosurg Focus. 2019;46(3):E11. doi:10.3171/2018.12.FOCUS18206
93. Kigerl KA, Hall JC, Wang L, Mo X, Yu Z, Popovich PG. Gut dysbiosis impairs recovery after spinal cord injury. J Exp Med. 2016;213(12):2603–2620. doi:10.1084/jem.20151345
94. Chow AK, Gulbransen BD. Potential roles of enteric glia in bridging neuroimmune communication in the gut. Am J Physiol Gastrointest Liver Physiol. 2017;312(2):G145–G152. doi:10.1152/ajpgi.00384.2016
95. Kuo WT, Odenwald MA, Turner JR, Zuo L. Tight junction proteins occludin and ZO-1 as regulators of epithelial proliferation and survival. Ann N Y Acad Sci. 2022;1514(1):21–33. doi:10.1111/nyas.14798
96. Feldman GJ, Mullin JM, Ryan MP. Occludin: structure, function and regulation. Adv Drug Deliv Rev. 2005;57(6):883–917. doi:10.1016/j.addr.2005.01.009
97. Liu X, Liang F, Zhang J, Li Z, Yang J, Kang N. Hyperbaric oxygen treatment improves intestinal barrier function after spinal cord injury in rats. Front Neurol. 2020;11:563281. doi:10.3389/fneur.2020.563281
98. Liu X, Liang F, Song W, Diao X, Zhu W, Yang J. Effect of Nrf2 signaling pathway on the improvement of intestinal epithelial barrier dysfunction by hyperbaric oxygen treatment after spinal cord injury. Cell Stress Chaperones. 2021;26(2):433–441. doi:10.1007/s12192-020-01190-1
99. Ouyang S, Wang X, Chen Y, et al. Swimming training combined with fecal microbial transplantation protects motor functions in rats with spinal cord injury by improving the intestinal system. Neurosci Lett. 2023;799:137104. doi:10.1016/j.neulet.2023.137104
100. Wu WL, Adame MD, Liou CW, et al. Microbiota regulate social behaviour via stress response neurons in the brain. Nature. 2021;595(7867):409–414. doi:10.1038/s41586-021-03669-y
101. Skelton F, Suda K, Evans C, Trautner B. Effective antibiotic stewardship in spinal cord injury: challenges and a way forward. J Spinal Cord Med. 2019;42(2):251–254. doi:10.1080/10790268.2017.1396183
102. Rodenhouse A, Talukder MAH, Lee JI, et al. Altered gut microbiota composition with antibiotic treatment impairs functional recovery after traumatic peripheral nerve crush injury in mice: effects of probiotics with butyrate producing bacteria. BMC Res Notes. 2022;15(1):80. doi:10.1186/s13104-022-05967-8
103. Brommer B, Engel O, Kopp MA, et al. Spinal cord injury-induced immune deficiency syndrome enhances infection susceptibility dependent on lesion level. Brain. 2016;139(3):692–707. doi:10.1093/brain/awv375
104. Mohammad S, Thiemermann C. Role of metabolic endotoxemia in systemic inflammation and potential interventions. Front Immunol. 2020;11:594150. doi:10.3389/fimmu.2020.594150
105. Camara-Lemarroy CR, Metz L, Meddings JB, Sharkey KA, Wee YV. The intestinal barrier in multiple sclerosis: implications for pathophysiology and therapeutics. Brain. 2018;141(7):1900–1916. doi:10.1093/brain/awy131
106. Stephens M, Von der weid PY. Lipopolysaccharides modulate intestinal epithelial permeability and inflammation in a species-specific manner. Gut Microbes. 2020;11(3):421–432. doi:10.1080/19490976.2019.1629235
107. Di Vincenzo F, Del GA, Petito V, Lopetuso LR, Scaldaferri F. Gut microbiota, intestinal permeability, and systemic inflammation: a narrative review. Intern Emerg Med. 2024;19(2):275–293. doi:10.1007/s11739-023-03374-w
108. Kayagaki N, Wong MT, Stowe IB, et al. Noncanonical inflammasome activation by intracellular LPS independent of TLR4. Science. 2013;341(6151):1246–1249. doi:10.1126/science.1240248
109. Aschenbach JR, Zebeli Q, Patra AK, Greco G, Amasheh S, Penner GB. Symposium review: the importance of the ruminal epithelial barrier for a healthy and productive cow. J Dairy Sci. 2019;102(2):1866–1882. doi:10.3168/jds.2018-15243
110. Nehra G, Bauer B, Hartz A. Blood-brain barrier leakage in Alzheimer’s disease: from discovery to clinical relevance. Pharmacol Ther. 2022;234:108119. doi:10.1016/j.pharmthera.2022.108119
111. Chen WK, Feng LJ, Liu QD, et al. Inhibition of leucine-rich repeats and calponin homology domain containing 1 accelerates microglia-mediated neuroinflammation in a rat traumatic spinal cord injury model. J Neuroinflammation. 2020;17(1):202. doi:10.1186/s12974-020-01884-4
112. Rong Z, Huang Y, Cai H, et al. Gut microbiota disorders promote inflammation and aggravate spinal cord injury through the TLR4/MyD88 signaling pathway. Frontiers in Nutrition. 2021;8:702659. doi:10.3389/fnut.2021.702659
113. Li Z, Bai H, Zhang R, et al. Systematic analysis of critical genes and pathways identified a signature of neuropathic pain after spinal cord injury. Eur J Neurosci. 2022;56(2):3991–4008.
114. Salga M, Samuel SG, Tseng HW, et al. Bacterial lipopolysaccharides exacerbate neurogenic heterotopic ossification development. J Bone Miner Res. 2023;38(11):1700–1717. doi:10.1002/jbmr.4905
115. Zhao J, Bi W, Xiao S, et al. Neuroinflammation induced by lipopolysaccharide causes cognitive impairment in mice. Sci Rep. 2019;9(1):5790.
116. Wang J, Zhang F, Xu H, et al. TLR4 aggravates microglial pyroptosis by promoting DDX3X-mediated NLRP3 inflammasome activation via JAK2/STAT1 pathway after spinal cord injury. Clin Transl Med. 2022;12(6):e894. doi:10.1002/ctm2.894
117. Dong X, Deng L, Su Y, et al. Curcumin alleviates traumatic brain injury induced by gas explosion through modulating gut microbiota and suppressing the LPS/TLR4/MyD88/NF-kappaB pathway. Environ Sci Pollut Res Int. 2024;31(1):1094–1113.
118. Tan JK, Macia L, Mackay CR. Dietary fiber and SCFAs in the regulation of mucosal immunity. J Allergy Clin Immunol. 2023;151(2):361–370.
119. Silva YP, Bernardi A, Frozza RL. The role of short-chain fatty acids from gut microbiota in gut-brain communication. Front Endocrinol. 2020;11:25.
120. Parada VD, De la Fuente MK, Landskron G, et al. Short chain fatty acids (SCFAs)-mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front Immunol. 2019;10:277.
121. Liu P, Liu M, Xi D, et al. Short-chain fatty acids ameliorate spinal cord injury recovery by regulating the balance of regulatory T cells and effector IL-17(+) gammadelta T cells. J Zhejiang Univ Sci B. 2023;24(4):312–325.
122. Jing Y, Yang D, Bai F, et al. Spinal cord injury-induced gut dysbiosis influences neurological recovery partly through short-chain fatty acids. NPJ Biofilms Microbiomes. 2023;9(1):99. doi:10.1038/s41522-023-00466-5
123. Blander JM, Longman RS, Iliev ID, Sonnenberg GF, Artis D. Regulation of inflammation by microbiota interactions with the host. Nat Immunol. 2017;18(8):851–860. doi:10.1038/ni.3780
124. Tang C, Wang C, Wang J, et al. Short-chain fatty acids ameliorate depressive-like behaviors of high fructose-fed mice by rescuing hippocampal neurogenesis decline and blood–brain barrier damage. Nutrients. 2022;14(9):1882. doi:10.3390/nu14091882
125. Socala K, Doboszewska U, Szopa A, et al. The role of microbiota-gut-brain axis in neuropsychiatric and neurological disorders. Pharmacol Res. 2021;172:105840.
126. Chen S, Ye J, Chen X, et al. Valproic acid attenuates traumatic spinal cord injury-induced inflammation via STAT1 and NF-kappaB pathway dependent of HDAC3. J Neuroinflammation. 2018;15(1):150.
127. Lee JY, Kim HS, Choi HY, Oh TH, Ju BG, Yune TY. Valproic acid attenuates blood-spinal cord barrier disruption by inhibiting matrix metalloprotease-9 activity and improves functional recovery after spinal cord injury. J Neurochem. 2012;121(5):818–829. doi:10.1111/j.1471-4159.2012.07731.x
128. Parker A, Fonseca S, Carding SR. Gut microbes and metabolites as modulators of blood-brain barrier integrity and brain health. Gut Microbes. 2020;11(2):135–157. doi:10.1080/19490976.2019.1638722
129. Rooks MG, Garrett WS. Gut microbiota, metabolites and host immunity. Nat Rev Immunol. 2016;16(6):341–352.
130. Jiang Y, Li K, Li X, Xu L, Yang Z. Sodium butyrate ameliorates the impairment of synaptic plasticity by inhibiting the neuroinflammation in 5XFAD mice. Chem Biol Interact. 2021;341:109452.
131. Wang D, Wang K, Liu Z, Wang Z, Wu H. Valproic acid-labeled chitosan nanoparticles promote recovery of neuronal injury after spinal cord injury. Aging. 2020;12(10):8953–8967. doi:10.18632/aging.103125
132. Zheng Z, Wu Y, Li Z, et al. Valproic acid affects neuronal fate and microglial function via enhancing autophagic flux in mice after traumatic brain injury. J Neurochem. 2020;154(3):284–300. doi:10.1111/jnc.14892
133. Aleti G, Troyer EA, Hong S. G protein-coupled receptors: a target for microbial metabolites and a mechanistic link to microbiome-immune-brain interactions. Brain Behav Immun Health. 2023;32:100671. doi:10.1016/j.bbih.2023.100671
134. Husted AS, Trauelsen M, Rudenko O, Hjorth SA, Schwartz TW. GPCR-mediated signaling of metabolites. Cell Metab. 2017;25(4):777–796. doi:10.1016/j.cmet.2017.03.008
135. Huang Y, Zhao M, Chen X, et al. Tryptophan metabolism in central nervous system diseases: pathophysiology and potential therapeutic strategies. Aging Dis. 2023;14(3):858. doi:10.14336/AD.2022.0916
136. Gao K, Mu CL, Farzi A, Zhu WY. Tryptophan metabolism: a link between the gut microbiota and brain. Adv Nutr. 2020;11(3):709–723. doi:10.1093/advances/nmz127
137. Mardinoglu A, Shoaie S, Bergentall M, et al. The gut microbiota modulates host amino acid and glutathione metabolism in mice. Mol Syst Biol. 2015;11(10):834. doi:10.15252/msb.20156487
138. Kaur H, Bose C, Mande SS. Tryptophan metabolism by gut microbiome and gut-brain-axis: an in silico analysis. Front Neurosci. 2019;13:1365. doi:10.3389/fnins.2019.01365
139. Dodd D, Spitzer MH, Van Treuren W, et al. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature. 2017;551(7682):648–652. doi:10.1038/nature24661
140. Gao J, Xu K, Liu H, et al. 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
141. Cervantes-Barragan L, Chai JN, Tianero MD, et al. Lactobacillus reuteri induces gut intraepithelial CD4(+)CD8alphaalpha(+) T cells. Science. 2017;357(6353):806–810. doi:10.1126/science.aah5825
142. He Y, Zhao C, Su N, et al. Disturbances of the gut microbiota-derived tryptophan metabolites as key actors in vagotomy-induced mastitis in mice. Cell Rep. 2024;43(8):114585. doi:10.1016/j.celrep.2024.114585
143. Purton T, Staskova L, Lane MM, et al. Prebiotic and probiotic supplementation and the tryptophan-kynurenine pathway: a systematic review and meta analysis. Neurosci Biobehav Rev. 2021;123:1–13. doi:10.1016/j.neubiorev.2020.12.026
144. Zhou M, Fan Y, Xu L, et al. Microbiome and tryptophan metabolomics analysis in adolescent depression: roles of the gut microbiota in the regulation of tryptophan-derived neurotransmitters and behaviors in human and mice. Microbiome. 2023;11(1):145. doi:10.1186/s40168-023-01589-9
145. Rust C, Malan-Muller S, van den Heuvel LL, et al. Platelets bridging the gap between gut dysbiosis and neuroinflammation in stress-linked disorders: a narrative review. J Neuroimmunol. 2023;382:578155. doi:10.1016/j.jneuroim.2023.578155
146. Guillemin GJ, Brew BJ, Noonan CE, Takikawa O, Cullen KM. Indoleamine 2,3 dioxygenase and quinolinic acid immunoreactivity in Alzheimer’s disease hippocampus. Neuropathol Appl Neurobiol. 2005;31(4):395–404. doi:10.1111/j.1365-2990.2005.00655.x
147. Wang D, Wu J, Zhu P, et al. Tryptophan-rich diet ameliorates chronic unpredictable mild stress induced depression- and anxiety-like behavior in mice: the potential involvement of gut-brain axis. Food Res Int. 2022;157:111289. doi:10.1016/j.foodres.2022.111289
148. Maddison DC, Giorgini F. The kynurenine pathway and neurodegenerative disease. Semin Cell Dev Biol. 2015;40:134–141. doi:10.1016/j.semcdb.2015.03.002
149. Roth W, Zadeh K, Vekariya R, Ge Y, Mohamadzadeh M. Tryptophan metabolism and gut-brain homeostasis. Int J Mol Sci. 2021;22(6):2973. doi:10.3390/ijms22062973
150. Chen C, Xiao Q, Wen Z, et al. Gut microbiome-derived indole-3-carboxaldehyde regulates stress vulnerability in chronic restraint stress by activating aryl hydrocarbon receptors. Pharmacol Res. 2025;213:107654.
151. Li YI, Wong G, Humphrey J, Raj T. Prioritizing Parkinson’s disease genes using population-scale transcriptomic data. Nat Commun. 2019;10(1):994.
152. Yaghoubfar R, Behrouzi A, Zare BE, et al. Effect of akkermansia muciniphila, faecalibacterium prausnitzii, and their extracellular vesicles on the serotonin system in intestinal epithelial cells. Probiotics Antimicrob Proteins. 2021;13(6):1546–1556. doi:10.1007/s12602-021-09786-4
153. Zaydi AI, Lew LC, Hor YY, et al. Lactobacillus plantarum DR7 improved brain health in aging rats via the serotonin, inflammatory and apoptosis pathways. Benef Microbes. 2020;11(8):753–766. doi:10.3920/BM2019.0200
154. Wang S, Ishima T, Zhang J, et al. Ingestion of Lactobacillus intestinalis and Lactobacillus reuteri causes depression- and anhedonia-like phenotypes in antibiotic-treated mice via the vagus nerve. J Neuroinflammation. 2020;17(1):241. doi:10.1186/s12974-020-01916-z
155. Rothhammer V, Mascanfroni ID, Bunse L, et al. Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor. Nat Med. 2016;22(6):586–597. doi:10.1038/nm.4106
156. Hu F, Feng AP, Liu XX, et al. Lipoxin A4 inhibits lipopolysaccharide-induced production of inflammatory cytokines in keratinocytes by up-regulating SOCS2 and down-regulating TRAF6. J Huazhong Univ Sci Technolog Med Sci. 2015;35(3):426–431. doi:10.1007/s11596-015-1448-8
157. Rothhammer V, Borucki DM, Tjon EC, et al. Microglial control of astrocytes in response to microbial metabolites. Nature. 2018;557(7707):724–728. doi:10.1038/s41586-018-0119-x
158. Ferrell JM, Chiang J. Bile acid receptors and signaling crosstalk in the liver, gut and brain. Liver Res. 2021;5(3):105–118. doi:10.1016/j.livres.2021.07.002
159. Fogelson KA, Dorrestein PC, Zarrinpar A, Knight R. The gut microbial bile acid modulation and its relevance to digestive health and diseases. Gastroenterology. 2023;164(7):1069–1085. doi:10.1053/j.gastro.2023.02.022
160. Joyce SA, Gahan CG. Disease-associated changes in bile acid profiles and links to altered gut microbiota. Dig Dis. 2017;35(3):169–177. doi:10.1159/000450907
161. Jia B, Park D, Hahn Y, Jeon CO. Metagenomic analysis of the human microbiome reveals the association between the abundance of gut bile salt hydrolases and host health. Gut Microbes. 2020;11(5):1300–1313. doi:10.1080/19490976.2020.1748261
162. Lin J, Zeng X, Su Z, et al. Hyodeoxycholic acid relieves neuropathic pain by activating farnesoid X receptor signaling. J Adv Res. 2025. doi:10.1016/j.jare.2025.07.017
163. Song H, Pang R, Chen Z, et al. Every-other-day fasting inhibits pyroptosis while regulating bile acid metabolism and activating TGR5 signaling in spinal cord injury. Front Mol Neurosci. 2024;17:1466125. doi:10.3389/fnmol.2024.1466125
164. Wu Y, Qiu Y, Su M, Wang L, Gong Q, Wei X. Activation of the bile acid receptors TGR5 and FXR in the spinal dorsal horn alleviates neuropathic pain. Cns Neurosci Ther. 2023;29(7):1981–1998. doi:10.1111/cns.14154
165. Wu S, Romero-Ramirez L, Mey J. Taurolithocholic acid but not tauroursodeoxycholic acid rescues phagocytosis activity of bone marrow-derived macrophages under inflammatory stress. J Cell Physiol. 2022;237(2):1455–1470. doi:10.1002/jcp.30619
166. Hou Y, Luan J, Huang T, et al. Tauroursodeoxycholic acid alleviates secondary injury in spinal cord injury mice by reducing oxidative stress, apoptosis, and inflammatory response. J Neuroinflammation. 2021;18(1):216. doi:10.1186/s12974-021-02248-2
167. Ko WK, Kim SJ, Jo MJ, et al. Ursodeoxycholic acid inhibits inflammatory responses and promotes functional recovery after spinal cord injury in rats. Mol Neurobiol. 2019;56(1):267–277. doi:10.1007/s12035-018-0994-z
168. Chang Y, Yang T, Ding H, Wang Z, Liang Q. Tauroursodeoxycholic acid protects rat spinal cord neurons after mechanical injury through regulating neuronal autophagy. Neurosci Lett. 2022;776:136578. doi:10.1016/j.neulet.2022.136578
169. Romero-Ramirez L, Garcia-Rama C, Wu S, Mey J. Bile acids attenuate PKM2 pathway activation in proinflammatory microglia. Sci Rep. 2022;12(1):1459. doi:10.1038/s41598-022-05408-3
170. Aburto MR, Cryan JF. Gastrointestinal and brain barriers: unlocking gates of communication across the microbiota-gut-brain axis. Nat Rev Gastroenterol Hepatol. 2024;21(4):222–247. doi:10.1038/s41575-023-00890-0
171. Pagan-Rivera LH, Ocasio-Rivera SE, Godoy-Vitorino F, Miranda JD. Spinal cord injury: pathophysiology, possible treatments and the role of the gut microbiota. Front Microbiol. 2024;15:1490855. doi:10.3389/fmicb.2024.1490855
172. 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
173. Besecker EM, Blanke EN, Deiter GM, Holmes GM. Gastric vagal afferent neuropathy following experimental spinal cord injury. Exp Neurol. 2020;323:113092. doi:10.1016/j.expneurol.2019.113092
174. Browning KN, Verheijden S, Boeckxstaens GE. The vagus nerve in appetite regulation, mood, and intestinal inflammation. Gastroenterology. 2017;152(4):730–744. doi:10.1053/j.gastro.2016.10.046
175. Margolis KG, Cryan JF, Mayer EA. The microbiota-gut-brain axis: from motility to mood. Gastroenterology. 2021;160(5):1486–1501. doi:10.1053/j.gastro.2020.10.066
176. Zhao C, Yang X, Su EM, et al. Signals of vagal circuits engaging with AKT1 in alpha7 nAChR(+)CD11b(+) cells lessen E. coli and LPS-induced acute inflammatory injury. Cell Discov. 2017;3:17009. doi:10.1038/celldisc.2017.9
177. Legan TB, Lavoie B, Mawe GM. Direct and indirect mechanisms by which the gut microbiota influence host serotonin systems. Neurogastroenterol Motil. 2022;34(10):e14346. doi:10.1111/nmo.14346
178. Chen H, Feng Z, Min L, et al. Vagus nerve stimulation reduces neuroinflammation through microglia polarization regulation to improve functional recovery after spinal cord injury. Front Neurosci. 2022;16:813472. doi:10.3389/fnins.2022.813472
179. Liu Y, Forsythe P. Vagotomy and insights into the microbiota-gut-brain axis. Neurosci Res. 2021;168:20–27. doi:10.1016/j.neures.2021.04.001
180. Williams EK, Chang RB, Strochlic DE, Umans BD, Lowell BB, Liberles SD. Sensory neurons that detect stretch and nutrients in the digestive system. Cell. 2016;166(1):209–221. doi:10.1016/j.cell.2016.05.011
181. Kim S, Kwon SH, Kam TI, et al. Transneuronal propagation of pathologic alpha-synuclein from the gut to the brain models parkinson’s disease. Neuron. 2019;103(4):627–641. doi:10.1016/j.neuron.2019.05.035
182. Dalton A, Mermier C, Zuhl M. Exercise influence on the microbiome-gut-brain axis. Gut Microbes. 2019;10(5):555–568. doi:10.1080/19490976.2018.1562268
183. Chen H, Feng Z, Min L, et al. Vagus nerve stimulation prevents endothelial necroptosis to alleviate blood-spinal cord barrier disruption after spinal cord injury. Mol Neurobiol. 2023;60(11):6466–6475. doi:10.1007/s12035-023-03477-7
184. Bubeck M, Becker C, Patankar JV. Guardians of the gut: influence of the enteric nervous system on the intestinal epithelial barrier. Front Med. 2023;10:1228938. doi:10.3389/fmed.2023.1228938
185. Julio-Pieper M, López-Aguilera A, Eyzaguirre-Velásquez J, et al. Gut susceptibility to viral invasion: contributing roles of diet, microbiota and enteric nervous system to mucosal barrier preservation. Int J Mol Sci. 2021;22(9):4734. doi:10.3390/ijms22094734
186. Willits AB, Kader L, Eller O, et al. Spinal cord injury-induced neurogenic bowel: a role for host-microbiome interactions in bowel pain and dysfunction. Neurobiol Pain. 2024;15:100156. doi:10.1016/j.ynpai.2024.100156
187. Hansebout CR, Su C, Reddy K, et al. Enteric glia mediate neuronal outgrowth through release of neurotrophic factors. Neural Regen Res. 2012;7(28):2165–2175. doi:10.3969/j.issn.1673-5374.2012.028.001
188. Wang C, Yan D, Huang J, Li Y. Impacts of changes in intestinal flora on the metabolism of Sprague-Dawley rats. Bioengineered. 2021;12(2):10603–10611. doi:10.1080/21655979.2021.2000242
189. McCracken BA, Nathalia GM. Phylum Synergistetes in the oral cavity: a possible contributor to periodontal disease. Anaerobe. 2021;68:102250. doi:10.1016/j.anaerobe.2020.102250
190. Nuli R, Cai J, Kadeer A, Zhang Y, Mohemaiti P. Integrative analysis toward different glucose tolerance-related gut microbiota and diet. Front Endocrinol (Lausanne). 2019;10:295. doi:10.3389/fendo.2019.00295
191. Gilbert O, Croffoot JR, Taylor AJ, Nash M, Schomer K, Groah S. Serum lipid concentrations among persons with spinal cord injury – a systematic review and meta-analysis of the literature. Atherosclerosis. 2014;232(2):305–312. doi:10.1016/j.atherosclerosis.2013.11.028
192. Heller RA, Seelig J, Bock T, et al. Relation of selenium status to neuro-regeneration after traumatic spinal cord injury. J Trace Elem Med Biol. 2019;51:141–149. doi:10.1016/j.jtemb.2018.10.006
193. Seelig J, Heller RA, Hackler J, et al. Selenium and copper status – potential signposts for neurological remission after traumatic spinal cord injury. J Trace Elem Med Biol. 2020;57:126415. doi:10.1016/j.jtemb.2019.126415
194. Tilg H, Zmora N, Adolph TE, Elinav E. The intestinal microbiota fuelling metabolic inflammation. Nat Rev Immunol. 2020;20(1):40–54. doi:10.1038/s41577-019-0198-4
195. Hasani A, Ebrahimzadeh S, Hemmati F, Khabbaz A, Hasani A, Gholizadeh P. The role of Akkermansia muciniphila in obesity, diabetes and atherosclerosis. J Med Microbiol. 2021;70(10). doi:10.1099/jmm.0.001435
196. Li J, Morrow C, Barnes S, et al. Gut microbiome composition and serum metabolome profile among individuals with spinal cord injury and normal glucose tolerance or prediabetes/type 2 diabetes. Arch Phys Med Rehabil. 2022;103(4):702–710. doi:10.1016/j.apmr.2021.03.043
197. Gholami H, Chmiel JA, Burton JP, Maleki Vareki S. The role of microbiota-derived vitamins in immune homeostasis and enhancing cancer immunotherapy. Cancers. 2023;15(4):1300. doi:10.3390/cancers15041300
198. Bello-Gil D, Audebert C, Olivera-Ardid S, et al. The formation of glycan-specific natural antibodies repertoire in GalT-KO mice is determined by gut microbiota. Front Immunol. 2019;10:342. doi:10.3389/fimmu.2019.00342
199. Kigerl KA, Zane K, Adams K, Sullivan MB, Popovich PG. The spinal cord-gut-immune axis as a master regulator of health and neurological function after spinal cord injury. Exp Neurol. 2020;323:113085. doi:10.1016/j.expneurol.2019.113085
200. Saksida T, Koprivica I, Vujicic M, et al. Impaired IL-17 Production in Gut-Residing Immune Cells of 5xFAD Mice with Alzheimer’s Disease Pathology. J Alzheimers Dis. 2018;61(2):619–630. doi:10.3233/JAD-170538
201. Shiokawa A, Kotaki R, Takano T, Nakajima-Adachi H, Hachimura S. Mesenteric lymph node CD11b(-) CD103(+) PD-L1(High) dendritic cells highly induce regulatory T cells. Immunology. 2017;152(1):52–64. doi:10.1111/imm.12747
202. Xu P, Zhang F, Chang MM, et al. Recruitment of gammadelta T cells to the lesion via the CCL2/CCR2 signaling after spinal cord injury. J Neuroinflammation. 2021;18(1):64. doi:10.1186/s12974-021-02115-0
203. Liu R, Li Y, Wang Z, et al. Regulatory T cells promote functional recovery after spinal cord injury by alleviating microglia inflammation via STAT3 inhibition. Cns Neurosci Ther. 2023;29(8):2129–2144. doi:10.1111/cns.14161
204. Nicholson JK, Holmes E, Kinross J, et al. Host-gut microbiota metabolic interactions. Science. 2012;336(6086):1262–1267. doi:10.1126/science.1223813
205. Yang J, Yang H, Li Y. The triple interactions between gut microbiota, mycobiota and host immunity. Crit Rev Food Sci Nutr. 2023;63(33):11604–11624. doi:10.1080/10408398.2022.2094888
206. Hill DA, Siracusa MC, Abt MC, et al. Commensal bacteria-derived signals regulate basophil hematopoiesis and allergic inflammation. Nat Med. 2012;18(4):538–546. doi:10.1038/nm.2657
207. Shi CW, Cheng MY, Yang X, et al. Probiotic Lactobacillus rhamnosus GG promotes mouse gut microbiota diversity and T cell differentiation. Front Microbiol. 2020;11:607735. doi:10.3389/fmicb.2020.607735
208. Li W, Hang S, Fang Y, et al. A bacterial bile acid metabolite modulates T(reg) activity through the nuclear hormone receptor NR4A1. Cell Host Microbe. 2021;29(9):1366–1377. doi:10.1016/j.chom.2021.07.013
209. Chen P, Tang X. Gut microbiota as regulators of Th17/Treg balance in patients with myasthenia gravis. Front Immunol. 2021;12:803101. doi:10.3389/fimmu.2021.803101
210. Ou Q, Power R, Griffin MD. Revisiting regulatory T cells as modulators of innate immune response and inflammatory diseases. Front Immunol. 2023;14:1287465. doi:10.3389/fimmu.2023.1287465
211. Mills K. IL-17 and IL-17-producing cells in protection versus pathology. Nat Rev Immunol. 2023;23(1):38–54. doi:10.1038/s41577-022-00746-9
212. McGinley AM, Sutton CE, Edwards SC, et al. Interleukin-17A serves a priming role in autoimmunity by recruiting IL-1beta-producing myeloid cells that promote pathogenic T cells. Immunity. 2020;52(2):342–356. doi:10.1016/j.immuni.2020.01.002
213. Sommer A, Marxreiter F, Krach F, et al. Th17 lymphocytes induce neuronal cell death in a human iPSC-based model of parkinson’s disease. Cell Stem Cell. 2018;23(1):123–131. doi:10.1016/j.stem.2018.06.015
214. Fiala GJ, Silva-Santos B. How to develop IL-17-producing gammadelta T cells. Immunol Cell Biol. 2018;96(9):886–887. doi:10.1111/imcb.12196
215. Sun G, Yang S, Cao G, et al. gammadelta T cells provide the early source of IFN-gamma to aggravate lesions in spinal cord injury. J Exp Med. 2018;215(2):521–535. doi:10.1084/jem.20170686
216. McGinley AM, Edwards SC, Raverdeau M, Mills KHG. Th17 cells, γδ T cells and their interplay in EAE and multiple sclerosis. J Autoimmun. 2018;87:97–108. doi:10.1016/j.jaut.2018.01.001
217. Ribot JC, Lopes N, Silva-Santos B. gammadelta T cells in tissue physiology and surveillance. Nat Rev Immunol. 2021;21(4):221–232. doi:10.1038/s41577-020-00452-4
218. Park JH, Kang I, Lee HK. gammadelta T cells in brain homeostasis and diseases. Front Immunol. 2022;13:886397. doi:10.3389/fimmu.2022.886397
219. Benakis C, Brea D, Caballero S, et al. Commensal microbiota affects ischemic stroke outcome by regulating intestinal gammadelta T cells. Nat Med. 2016;22(5):516–523. doi:10.1038/nm.4068
220. Xi D, Liu P, Feng Y, et al. Fecal microbiota transplantation regulates the microbiota-gut-spinal cord axis to promote recovery after spinal cord injury. Int Immunopharmacol. 2024;126:111212. doi:10.1016/j.intimp.2023.111212
221. Bermúdez-Humarán LG, Salinas E, Ortiz GG, Ramirez-Jirano LJ, Morales JA, Bitzer-Quintero OK. From probiotics to psychobiotics: live beneficial bacteria which act on the brain-gut axis. Nutrients. 2019;11(4):890. doi:10.3390/nu11040890
222. Zagorska A, Marcinkowska M, Jamrozik M, Wisniowska B, Pasko P. From probiotics to psychobiotics – the gut-brain axis in psychiatric disorders. Benef Microbes. 2020;11(8):717–732. doi:10.3920/BM2020.0063
223. El DR, Periyasamy AG, de Barros JL, et al. Probiotics for the treatment of depression and anxiety: a systematic review and meta-analysis of randomized controlled trials. Clin Nutr ESPEN. 2021;45:75–90. doi:10.1016/j.clnesp.2021.07.027
224. Chao L, Liu C, Sutthawongwadee S, et al. Effects of probiotics on depressive or anxiety variables in healthy participants under stress conditions or with a depressive or anxiety diagnosis: a meta-analysis of randomized controlled trials. Front Neurol. 2020;11:421. doi:10.3389/fneur.2020.00421
225. Asad A, Kirk M, Zhu S, Dong X, Gao M. Effects of prebiotics and probiotics on symptoms of depression and anxiety in clinically diagnosed samples: systematic review and meta-analysis of randomized controlled trials. Nutr Rev. 2025;83(7):e1504–e1520. doi:10.1093/nutrit/nuae177
226. Jager R, Mohr AE, Carpenter KC, et al. International society of sports nutrition position stand: probiotics. J Int Soc Sports Nutr. 2019;16(1):62. doi:10.1186/s12970-019-0329-0
227. Jogia T, Ruitenberg MJ. Traumatic spinal cord injury and the gut microbiota: current insights and future challenges. Front Immunol. 2020;11:704. doi:10.3389/fimmu.2020.00704
228. Lin F, Zhang B, Shi Q, et al. The conditioned medium of Lactobacillus rhamnoides GG regulates microglia/macrophage polarization and improves functional recovery after spinal cord injury in rats. Biomed Res Int. 2021;2021:3376496. doi:10.1155/2021/3376496
229. Cen Q, Cui Y, Feng J, et al. Limosilactobacillus reuteri DSM17938 attenuates neuroinflammatory responses after spinal cord injury by modulating tryptophan metabolism. Probiotics Antimicro. 2025. doi:10.1007/s12602-025-10545-y
230. Yoo JY, Kim SS. Probiotics and prebiotics: present status and future perspectives on metabolic disorders. Nutrients. 2016;8(3):173. doi:10.3390/nu8030173
231. Valido E, Capossela S, Glisic M, et al. Gut microbiome and inflammation among athletes in wheelchair in a crossover randomized pilot trial of probiotic and prebiotic interventions. Sci Rep. 2024;14(1):12838. doi:10.1038/s41598-024-63163-z
232. Wong S, Hirani SP, Forbes A, et al. Lactobacillus casei Shirota probiotic drinks reduce antibiotic associated diarrhoea in patients with spinal cord injuries who regularly consume proton pump inhibitors: a subgroup analysis of the ECLISP multicentre RCT. Spinal Cord. 2024;62(5):255–263. doi:10.1038/s41393-024-00983-w
233. Raposo P, Nguyen AT, Schmidt E, et al. No beneficial effects of the alfasigma VSL#3 Probiotic treatment after cervical spinal cord injury in rats. Top Spinal Cord Inj Rehabil. 2025;31(1):1–16. doi:10.46292/sci24-00004
234. Jamali F, Mousavi S, Homayouni-Rad A, et al. Exploring Innovative Approaches for Managing Spinal Cord Injury: a Comprehensive Review of Promising Probiotics and Postbiotics. Probiotics Antimicro. 2025. doi:10.1007/s12602-025-10513-6
235. Sanders ME, Merenstein DJ, Reid G, Gibson GR, Rastall RA. Probiotics and prebiotics in intestinal health and disease: from biology to the clinic. Nat Rev Gastroenterol Hepatol. 2019;16(10):605–616. doi:10.1038/s41575-019-0173-3
236. Roy S, Dhaneshwar S. Role of prebiotics, probiotics, and synbiotics in management of inflammatory bowel disease: current perspectives. World J Gastroenterol. 2023;29(14):2078–2100. doi:10.3748/wjg.v29.i14.2078
237. Swanson KS, Gibson GR, Hutkins R, et al. The international scientific association for probiotics and prebiotics (ISAPP) consensus statement on the definition and scope of synbiotics. Nat Rev Gastroenterol Hepatol. 2020;17(11):687–701. doi:10.1038/s41575-020-0344-2
238. Leonardi I, Paramsothy S, Doron I, et al. Fungal trans-kingdom dynamics linked to responsiveness to fecal microbiota transplantation (FMT) therapy in ulcerative colitis. Cell Host Microbe. 2020;27(5):823–829. doi:10.1016/j.chom.2020.03.006
239. Kelly CR, Yen EF, Grinspan AM, et al. Fecal microbiota transplantation is highly effective in real-world practice: initial results from the FMT national registry. Gastroenterology. 2021;160(1):183–192. doi:10.1053/j.gastro.2020.09.038
240. Hernandez-Torres V, Gransee HM, Mantilla CB, Wang Y, Zhan WZ, Sieck GC. BDNF effects on functional recovery across motor behaviors after cervical spinal cord injury. J Neurophysiol. 2017;117(2):537–544. doi:10.1152/jn.00654.2016
241. Li Y, Tran A, Graham L, Brock J, Tuszynski MH, Lu P. BDNF guides neural stem cell-derived axons to ventral interneurons and motor neurons after spinal cord injury. Exp Neurol. 2023;359:114259. doi:10.1016/j.expneurol.2022.114259
242. Jing Y, Bai F, Wang L, et al. Fecal microbiota transplantation exerts neuroprotective effects in a mouse spinal cord injury model by modulating the microenvironment at the lesion site. Microbiol Spectr. 2022;10(3):e122–e177. doi:10.1128/spectrum.00177-22
243. Sampson TR, Debelius JW, Thron T, et al. Gut microbiota regulate motor deficits and neuroinflammation in a model of parkinson’s disease. Cell. 2016;167(6):1469–1480. doi:10.1016/j.cell.2016.11.018
244. Sun MF, Zhu YL, Zhou ZL, et al. Neuroprotective effects of fecal microbiota transplantation on MPTP-induced Parkinson’s disease mice: gut microbiota, glial reaction and TLR4/TNF-alpha signaling pathway. Brain Behav Immun. 2018;70:48–60. doi:10.1016/j.bbi.2018.02.005
245. Sun J, Xu J, Ling Y, 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
246. Nirmalkar K, Qureshi F, Kang D, Hahn J, Adams JB, Krajmalnik-Brown R. Shotgun metagenomics study suggests alteration in sulfur metabolism and oxidative stress in children with autism and improvement after microbiota transfer therapy. Int J Mol Sci. 2022;23(21):13481. doi:10.3390/ijms232113481
247. Chen R, Xu Y, Wu P, et al. Transplantation of fecal microbiota rich in short chain fatty acids and butyric acid treat cerebral ischemic stroke by regulating gut microbiota. Pharmacol Res. 2019;148:104403. doi:10.1016/j.phrs.2019.104403
248. Li K, Wei S, Hu L, et al. Protection of fecal microbiota transplantation in a mouse model of multiple sclerosis. Mediators Inflammation. 2020;2020:1–13.
249. Wang JW, Kuo CH, Kuo FC, et al. Fecal microbiota transplantation: review and update. J Formos Med Assoc. 2019;118 Suppl 1:S23–S31. doi:10.1016/j.jfma.2018.08.011
250. Blaser MJ. Fecal Microbiota Transplantation for Dysbiosis – Predictable Risks. N Engl J Med. 2019;381(21):2064–2066. doi:10.1056/NEJMe1913807
251. DeFilipp Z, Bloom PP, Torres SM, et al. Drug-resistant E. coli bacteremia transmitted by fecal microbiota transplant. N Engl J Med. 2019;381(21):2043–2050. doi:10.1056/NEJMoa1910437
252. Kriz J, Hysperska V, Bebrova E, Roznetinska M. Faecal microbiota transplantation for multidrug-resistant organism decolonization in spinal cord injury patients: a case series. Infect Prev Pract. 2024;6(1):100340. doi:10.1016/j.infpip.2024.100340