BPG is committed to discovery and dissemination of knowledge
Review
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
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 miceDSS-induced colitisElevated NLRP3-driven neuroinflammation, cognitive deficits, impaired glymphatic clearanceLinks colitis-associated inflammation to neurodegeneration via immune signaling[42]
Increased IL-1β, caspase-1, gasdermin D, Aβ, HMGB1, BBB disruption, TNF-α, IL-6Systemic inflammation impairs BBB and enhances neuroinflammation[45]
Upregulated IDO-1, increased kynurenine and kynurenic acid, worsened cognitive declineSystemic inflammation contributes to neuroinflammation and cognitive impairment[46]
Tg2576 Transgenic miceDSS-induced colitisAltered gut microbiota, increased GFAP and microglial activationDemonstrates that chronic gut dysbiosis worsens cognitive dysfunction and AD pathology[43]
Gut dysbiosis before ADIncreased gut permeability, reduced tight junction proteins, and fluorescein isothiocyanate-dextran leakageGut barrier dysfunction precedes and contributes to AD pathology[50]
3XTg-AD transgenic miceDSS-induced colitis + vagotomyC/EBPβ/δ-secretase activation, Aβ and Tau fibril propagation to the brain via the vagus nerveHighlights gut-brain signaling in AD exacerbation due to gut inflammation, with the vagus nerve as the conduit[44]
Transgenerational effects on antibiotic-induced dysbiosisDelayed cognitive decline, reduced Aβ aggregation in the brainGut microbiota influences AD pathology via immune modulation across generations[40]
APP/PS1 transgenic miceAntibiotic-induced gut dysbiosisAltered gut microbiota, reduced amyloid pathology, increased T-reg cells, and modified microglial activationHighlights the gut microbiota's role in AD via immune modulation, even without direct colitis[53]
Fecal microbiota transplantRestoration of microbial balance, reduced amyloid load, and cognitive improvementSupports microbiota-targeting therapy for AD mitigation[54]
Gut dysbiosisMicroglial activation, elevated TNF-α, IL-6, and early gut microbiota changesGut dysbiosis precedes and contributes to AD pathology[8]
Intestinal barrier alterationsExcessive Aβ accumulation in gut epithelium, increased permeability, inflammatory changes, and mucin2 upregulation[59]Gut barrier dysfunction linked to AD pathology[59][50]
Germ-free conditionReduced amyloid plaque deposition compared to control miceSuggests gut microbiota influence on AD pathogenesis[39]
SAMP8 miceGut dysfunction, colonic motilityDelayed colonic motility, enteric neurodegeneration, early ENS Aβ and Tau aggregatesENS dysfunction may precede brain AD pathology[52]
APPNL-G-F transgenic miceDSS-induced colitisIncreased neutrophil infiltration into the hippocampus and cortex, clustering around Aβ plaques, worsening neuroinflammationNeutrophils migrate through the GBA and exacerbate AD pathology[48]
Significant exacerbation of Aβ plaque deposition, altered CD68, and increased systemic inflammationGut inflammation triggers neuroinflammation and worsens AD pathology[19]
DSS-induced colitis + probioticsReduced colitis severity, partially improved gut permeability, limited effects on neuroinflammation and AβProbiotics have mild protective effects on the gut and brain[55]
APP/PS1 and AppNL-G-F miceStudied for gut-brain interactionsIncreased amyloid plaque deposition in the intestines and brain, gut motility and permeability changesSupports the hypothesis that gut inflammation precedes neurodegeneration[41]
5XFAD transgenic miceGenetic 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 miceThe 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 inflammasomeNLRP3, ASC, caspase-1, IL-1β, IL-18, GSDMDDrives mucosal inflammation and epithelial barrier damage via IL-1β/IL-18 maturation and pyroptosisPromotes microglial activation, synaptic dysfunction, and Aβ/Tau pathology via IL-1β/IL-18; pyroptosis amplifies neuroinflammationNLRP3 inhibitors (e.g., MCC950), IL-1 blockers (anakinra), caspase-1/GSDMD inhibitors[42,44,47,101]
Proinflammatory cytokinesTNF-α, IL-6, IL-1β, IFN-γCentral mediators of flare activity; correlate with severity; disrupt tight junctionsSustain neuroinflammation, impair synaptic plasticity, associate with cognitive decline; disrupt BBBAnti-TNF (infliximab, adalimumab), anti-IL-6 (tocilizumab), IL-1 blockade; JAK inhibitors[61,76,80,81]
Tryptophan–kynurenineIDO1, Tryptophan 2,3-dioxygenase, Kynurenine, 3-Hydroxykynurenine, QA, KAInflammation upregulates IDO1; shifts TRP metabolism; impacts gut immunityKynurenine Pathway metabolites modulate glutamatergic/cholinergic signaling; QA neurotoxic; KA neuromodulatoryIDO1 inhibitors; KP modulators; microbiota-directed TRP metabolism[46,67]
Microglia–astrocyte activationTLR2/4, NF-κB, C/EBPβ, AEP (δ-secretase), GFAPTLR activation by microbial products fuels cytokines and tissue damageGlial priming via TLR/NF-κB; C/EBPβ–AEP axis accelerates Aβ/Tau pathologyTLR antagonists; NF-κB inhibitors; AEP inhibitors; glial modulators[96,97,100-103]
Barrier–adhesion–traffickingTight junctions (ZO-1, occludin, claudins), MUC2, HMGB1, ICAM-1/VCAM-1Barrier loss, mucus alterations, leukocyte adhesion/extravasationBBB disruption, HMGB1 as alarmin, leukocyte trafficking into brainBarrier protectants; HMGB1 antagonists; anti-adhesion strategies[145-147]
Vagus–cholinergic anti-inflammatoryVagus nerve, α7 nAChRModulates intestinal inflammation via vagal toneVagal signaling links gut inflammation to brain; α7 nAChR neuroprotectiveVagus nerve stimulation; α7 nAChR agonists[59,69-72]
Mitochondrial stress-ROSmtROS, NLRP3 activation, mitophagyOxidative stress perpetuates mucosal inflammationmtROS primes microglia; contributes to Aβ/Tau aggregationAntioxidants; 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 medicationsTNF-α inhibitors (infliximab, adalimumab)Reduce systemic inflammation and BBB permeability by blocking TNF-αClinical data from IBD patients; Phase 3 trials63% reduced risk in CD patients, 36% in UC patients; neuroprotective effectsWidely used IBD treatment; reduces gut inflammation and flare activityNCT00207766, NCT05090124
Immunomodulators (azathioprine, mercaptopurine, methotrexate)Suppress systemic inflammation through immune modulationClinical cohort studies in IBD patients37% reduction in AD risk in both CD and UC patientsControls inflammation and maintains remission[37]
IL-1β blockers (anakinra)Block IL-1 inflammatory pathway reducing neuroinflammationASCOT trial in mild AD patientsPotential cognitive improvement or stabilizationReduces gut mucosal inflammation and IL-1β-mediated damageNCT04834388
IL-6 receptor inhibitors (tocilizumab)Reduce IL-6 mediated neuroinflammation and amyloidogenesisClinical studies in inflammatory conditionsAnti-inflammatory effects; may reduce neuroinflammationReduces intestinal IL-6 signaling and inflammation[151,152]
JAK inhibitors (tofacitinib)Inhibit JAK/STAT cytokine signaling pathwayApproved for IBD treatment; potential AD therapyMay reduce glial activation-induced neuroinflammationEffective in decreasing intestinal inflammation and barrier breakdown[106,107]
XPro™ (selective TNF inhibitor)Selective TNF inhibition targeting neuroinflammationMINDFuL trial in early AD and MCI patientsInvestigated for neuroinflammation reduction in early ADPotential anti-inflammatory effects on gut mucosaNCT05318976
P2X7 Receptor AntagonistsModulate P2X7 activation, dampen ATP-driven inflammasome signaling, cut downstream IL-1β/TNF-α releasePre-clinical IBD and AD models show reduced gut and brain inflammationLower microglial activation, curb neuroinflammation, protect neuronsInhibits EGC hyperactivation and cytokine release, reducing gut inflammation[94-96]
Gut microbiota modulationProbiotic supplementation (Lactobacillus, Bifidobacterium)Restore gut microbial balance; reduce gut permeability and systemic inflammationPreclinical AD models; Probio-AD trialImproves cognitive function; reduces neuroinflammationDecreases colitis severity; partially restores gut barrierNCT05145881
Fecal microbiota transplantationRestore healthy gut microbiota and improve gut-brain communicationAnimal models (APP/PS1 mice); clinical trialDecreases Aβ deposition; enhances cognitive performanceImproves gut microbiota composition; reduces inflammationNCT06920212
Dietary and lifestyle modificationsMediterranean dietRich in polyphenols and omega-3 fatty acids; modulates gut microbiota and reduces inflammationClinical studiesReduces AD risk; lowers proinflammatory cytokines and neuroinflammationBeneficially alters gut microbiota composition; reduces systemic inflammationISRCTN35739639
Ketogenic dietPromotes ketone body synthesis; enhances mitochondrial function and reduces oxidative stressClinical trialsNeuroprotective effects; potentially slows AD progressionMay improve mitochondrial function in gut epitheliumACTRN12618001450202
Targeting bacterial amyloidsAnti-bacterial amyloids targetingPrevent bacterial amyloid formation (E. coli, B. subtilis) that accelerates cerebral amyloid aggregationPreclinical studies on bacterial amyloidsReduce amyloid pathology and Aβ aggregationReduces gut bacterial-derived amyloid formation and inflammation[160-162]
Neuroimmune modulationVagus nerve stimulation and α7nAChR agonistsEnhance CAIP to regulate immune responsesPreclinical and clinical studies in IBD and ADNeuroprotective by reducing neuroinflammation via α7nAChR activationModulates intestinal inflammation through improved vagal tone[59,68-72]