Copyright
©The Author(s) 2025.
World J Gastroenterol. Dec 28, 2025; 31(48): 111301
Published online Dec 28, 2025. doi: 10.3748/wjg.v31.i48.111301
Published online Dec 28, 2025. doi: 10.3748/wjg.v31.i48.111301
Table 1 Summary of animal model studies on the association between inflammatory bowel disease and Alzheimer’s disease
| Animal model | Induced condition | Key observation (outcome measures) | Relevance to IBD-AD link | Ref. |
| C57BL/6J mice | DSS-induced colitis | Elevated NLRP3-driven neuroinflammation, cognitive deficits, impaired glymphatic clearance | Links colitis-associated inflammation to neurodegeneration via immune signaling | [42] |
| Increased IL-1β, caspase-1, gasdermin D, Aβ, HMGB1, BBB disruption, TNF-α, IL-6 | Systemic inflammation impairs BBB and enhances neuroinflammation | [45] | ||
| Upregulated IDO-1, increased kynurenine and kynurenic acid, worsened cognitive decline | Systemic inflammation contributes to neuroinflammation and cognitive impairment | [46] | ||
| Tg2576 Transgenic mice | DSS-induced colitis | Altered gut microbiota, increased GFAP and microglial activation | Demonstrates that chronic gut dysbiosis worsens cognitive dysfunction and AD pathology | [43] |
| Gut dysbiosis before AD | Increased gut permeability, reduced tight junction proteins, and fluorescein isothiocyanate-dextran leakage | Gut barrier dysfunction precedes and contributes to AD pathology | [50] | |
| 3XTg-AD transgenic mice | DSS-induced colitis + vagotomy | C/EBPβ/δ-secretase activation, Aβ and Tau fibril propagation to the brain via the vagus nerve | Highlights gut-brain signaling in AD exacerbation due to gut inflammation, with the vagus nerve as the conduit | [44] |
| Transgenerational effects on antibiotic-induced dysbiosis | Delayed cognitive decline, reduced Aβ aggregation in the brain | Gut microbiota influences AD pathology via immune modulation across generations | [40] | |
| APP/PS1 transgenic mice | Antibiotic-induced gut dysbiosis | Altered gut microbiota, reduced amyloid pathology, increased T-reg cells, and modified microglial activation | Highlights the gut microbiota's role in AD via immune modulation, even without direct colitis | [53] |
| Fecal microbiota transplant | Restoration of microbial balance, reduced amyloid load, and cognitive improvement | Supports microbiota-targeting therapy for AD mitigation | [54] | |
| Gut dysbiosis | Microglial activation, elevated TNF-α, IL-6, and early gut microbiota changes | Gut dysbiosis precedes and contributes to AD pathology | [8] | |
| Intestinal barrier alterations | Excessive Aβ accumulation in gut epithelium, increased permeability, inflammatory changes, and mucin2 upregulation[59] | Gut barrier dysfunction linked to AD pathology[59] | [50] | |
| Germ-free condition | Reduced amyloid plaque deposition compared to control mice | Suggests gut microbiota influence on AD pathogenesis | [39] | |
| SAMP8 mice | Gut dysfunction, colonic motility | Delayed colonic motility, enteric neurodegeneration, early ENS Aβ and Tau aggregates | ENS dysfunction may precede brain AD pathology | [52] |
| APPNL-G-F transgenic mice | DSS-induced colitis | Increased neutrophil infiltration into the hippocampus and cortex, clustering around Aβ plaques, worsening neuroinflammation | Neutrophils migrate through the GBA and exacerbate AD pathology | [48] |
| Significant exacerbation of Aβ plaque deposition, altered CD68, and increased systemic inflammation | Gut inflammation triggers neuroinflammation and worsens AD pathology | [19] | ||
| DSS-induced colitis + probiotics | Reduced colitis severity, partially improved gut permeability, limited effects on neuroinflammation and Aβ | Probiotics have mild protective effects on the gut and brain | [55] | |
| APP/PS1 and AppNL-G-F mice | Studied for gut-brain interactions | Increased amyloid plaque deposition in the intestines and brain, gut motility and permeability changes | Supports the hypothesis that gut inflammation precedes neurodegeneration | [41] |
| 5XFAD transgenic mice | Genetic overexpression of human APP & PS1 mutations (presymptomatic stage) | Reduced IL-17 protein production/secretion in PP and MLN cells of 5xFAD mice, significantly lower miR-155 expression in MLN cells of 5xFAD mice | The observed GALT changes, especially reduced IL-17 (linked to ↓miR-155), mirror AD progression. This might reflect inadequate immune surveillance in the gut, potentially leading to enhanced AD pathology (e.g., via altered microbiota or Aβ clearance) | [49] |
Table 2 Key inflammatory and immune signaling pathways shared between inflammatory bowel disease and Alzheimer’s disease
| Pathway | Key molecules | Role in IBD | Role in AD | Therapeutic target | Ref. |
| NLRP3 inflammasome | NLRP3, ASC, caspase-1, IL-1β, IL-18, GSDMD | Drives mucosal inflammation and epithelial barrier damage via IL-1β/IL-18 maturation and pyroptosis | Promotes microglial activation, synaptic dysfunction, and Aβ/Tau pathology via IL-1β/IL-18; pyroptosis amplifies neuroinflammation | NLRP3 inhibitors (e.g., MCC950), IL-1 blockers (anakinra), caspase-1/GSDMD inhibitors | [42,44,47,101] |
| Proinflammatory cytokines | TNF-α, IL-6, IL-1β, IFN-γ | Central mediators of flare activity; correlate with severity; disrupt tight junctions | Sustain neuroinflammation, impair synaptic plasticity, associate with cognitive decline; disrupt BBB | Anti-TNF (infliximab, adalimumab), anti-IL-6 (tocilizumab), IL-1 blockade; JAK inhibitors | [61,76,80,81] |
| Tryptophan–kynurenine | IDO1, Tryptophan 2,3-dioxygenase, Kynurenine, 3-Hydroxykynurenine, QA, KA | Inflammation upregulates IDO1; shifts TRP metabolism; impacts gut immunity | Kynurenine Pathway metabolites modulate glutamatergic/cholinergic signaling; QA neurotoxic; KA neuromodulatory | IDO1 inhibitors; KP modulators; microbiota-directed TRP metabolism | [46,67] |
| Microglia–astrocyte activation | TLR2/4, NF-κB, C/EBPβ, AEP (δ-secretase), GFAP | TLR activation by microbial products fuels cytokines and tissue damage | Glial priming via TLR/NF-κB; C/EBPβ–AEP axis accelerates Aβ/Tau pathology | TLR antagonists; NF-κB inhibitors; AEP inhibitors; glial modulators | [96,97,100-103] |
| Barrier–adhesion–trafficking | Tight junctions (ZO-1, occludin, claudins), MUC2, HMGB1, ICAM-1/VCAM-1 | Barrier loss, mucus alterations, leukocyte adhesion/extravasation | BBB disruption, HMGB1 as alarmin, leukocyte trafficking into brain | Barrier protectants; HMGB1 antagonists; anti-adhesion strategies | [145-147] |
| Vagus–cholinergic anti-inflammatory | Vagus nerve, α7 nAChR | Modulates intestinal inflammation via vagal tone | Vagal signaling links gut inflammation to brain; α7 nAChR neuroprotective | Vagus nerve stimulation; α7 nAChR agonists | [59,69-72] |
| Mitochondrial stress-ROS | mtROS, NLRP3 activation, mitophagy | Oxidative stress perpetuates mucosal inflammation | mtROS primes microglia; contributes to Aβ/Tau aggregation | Antioxidants; mitophagy enhancers | [119-124] |
Table 3 Therapeutic approaches targeting gut-brain axis dysfunction in inflammatory bowel disease and Alzheimer’s disease
| Intervention | Mechanism | Evidence (model or study) | Outcome on AD | Outcome of IBD | Trial No./Ref. | |
| Anti-inflammatory medications | TNF-α inhibitors (infliximab, adalimumab) | Reduce systemic inflammation and BBB permeability by blocking TNF-α | Clinical data from IBD patients; Phase 3 trials | 63% reduced risk in CD patients, 36% in UC patients; neuroprotective effects | Widely used IBD treatment; reduces gut inflammation and flare activity | NCT00207766, NCT05090124 |
| Immunomodulators (azathioprine, mercaptopurine, methotrexate) | Suppress systemic inflammation through immune modulation | Clinical cohort studies in IBD patients | 37% reduction in AD risk in both CD and UC patients | Controls inflammation and maintains remission | [37] | |
| IL-1β blockers (anakinra) | Block IL-1 inflammatory pathway reducing neuroinflammation | ASCOT trial in mild AD patients | Potential cognitive improvement or stabilization | Reduces gut mucosal inflammation and IL-1β-mediated damage | NCT04834388 | |
| IL-6 receptor inhibitors (tocilizumab) | Reduce IL-6 mediated neuroinflammation and amyloidogenesis | Clinical studies in inflammatory conditions | Anti-inflammatory effects; may reduce neuroinflammation | Reduces intestinal IL-6 signaling and inflammation | [151,152] | |
| JAK inhibitors (tofacitinib) | Inhibit JAK/STAT cytokine signaling pathway | Approved for IBD treatment; potential AD therapy | May reduce glial activation-induced neuroinflammation | Effective in decreasing intestinal inflammation and barrier breakdown | [106,107] | |
| XPro™ (selective TNF inhibitor) | Selective TNF inhibition targeting neuroinflammation | MINDFuL trial in early AD and MCI patients | Investigated for neuroinflammation reduction in early AD | Potential anti-inflammatory effects on gut mucosa | NCT05318976 | |
| P2X7 Receptor Antagonists | Modulate P2X7 activation, dampen ATP-driven inflammasome signaling, cut downstream IL-1β/TNF-α release | Pre-clinical IBD and AD models show reduced gut and brain inflammation | Lower microglial activation, curb neuroinflammation, protect neurons | Inhibits EGC hyperactivation and cytokine release, reducing gut inflammation | [94-96] | |
| Gut microbiota modulation | Probiotic supplementation (Lactobacillus, Bifidobacterium) | Restore gut microbial balance; reduce gut permeability and systemic inflammation | Preclinical AD models; Probio-AD trial | Improves cognitive function; reduces neuroinflammation | Decreases colitis severity; partially restores gut barrier | NCT05145881 |
| Fecal microbiota transplantation | Restore healthy gut microbiota and improve gut-brain communication | Animal models (APP/PS1 mice); clinical trial | Decreases Aβ deposition; enhances cognitive performance | Improves gut microbiota composition; reduces inflammation | NCT06920212 | |
| Dietary and lifestyle modifications | Mediterranean diet | Rich in polyphenols and omega-3 fatty acids; modulates gut microbiota and reduces inflammation | Clinical studies | Reduces AD risk; lowers proinflammatory cytokines and neuroinflammation | Beneficially alters gut microbiota composition; reduces systemic inflammation | ISRCTN35739639 |
| Ketogenic diet | Promotes ketone body synthesis; enhances mitochondrial function and reduces oxidative stress | Clinical trials | Neuroprotective effects; potentially slows AD progression | May improve mitochondrial function in gut epithelium | ACTRN12618001450202 | |
| Targeting bacterial amyloids | Anti-bacterial amyloids targeting | Prevent bacterial amyloid formation (E. coli, B. subtilis) that accelerates cerebral amyloid aggregation | Preclinical studies on bacterial amyloids | Reduce amyloid pathology and Aβ aggregation | Reduces gut bacterial-derived amyloid formation and inflammation | [160-162] |
| Neuroimmune modulation | Vagus nerve stimulation and α7nAChR agonists | Enhance CAIP to regulate immune responses | Preclinical and clinical studies in IBD and AD | Neuroprotective by reducing neuroinflammation via α7nAChR activation | Modulates intestinal inflammation through improved vagal tone | [59,68-72] |
- Citation: Durairajan SSK, Singh AK, Sulaiman SM, Patnaik S, Krishnamoorthi S, Iyaswamy A, Vellingiri B, Yang CB, Williams LL. Molecular links between inflammatory bowel disease and Alzheimer’s disease through immune signaling and inflammatory pathways. World J Gastroenterol 2025; 31(48): 111301
- URL: https://www.wjgnet.com/1007-9327/full/v31/i48/111301.htm
- DOI: https://dx.doi.org/10.3748/wjg.v31.i48.111301
