Abstract Background Cognitive dysfunction in diabetes significantly impairs quality of life, yet effective therapies remain limited. Our previous work showed that α-methyltryptophan (α-MT), an inhibitor of the amino acid transporter SLC6A14 and indoleamine 2,3-dioxygenase 1 (IDO1), ameliorates diabetic nephropathy by modulating renal metabolism. Given its role in metabolic regulation, we hypothesized that α-MT may also protect against cognitive decline in diabetes by influencing gut-brain metabolic crosstalk. Methods Db/db mice were used as an in vivo model of type 2 diabetes, complemented by high-glucose–treated Caco-2 cells in vitro. Cognitive function was assessed using the Morris water maze. Gene expression related to synaptic plasticity (c-FOS, ARC, BDNF, EGR1) was measured by RT-qPCR. Colon morphology and SLC6A14 expression were evaluated by H&E, AB-PAS, immunohistochemistry, and Western blotting. ¹H NMR–based metabolomics was applied to profile metabolic changes in the colon and hippocampus. Results α-MT treatment significantly reduced hyperglycemia, restored intestinal barrier integrity, and improved cognitive performance in db/db mice. It also downregulated SLC6A14 overexpression under hyperglycemic conditions in both models. Metabolomic analysis revealed that α-MT induced significant metabolic reprogramming in the colon, with amino acid metabolism as the most affected pathway. Notably, metabolite alterations in the colon were positively correlated with those in the hippocampus, and both were negatively associated with increased expression of c-FOS, ARC, BDNF, and EGR1, suggesting coordinated gut-brain metabolic responses. Conclusions These findings indicate that α-MT alleviates diabetes-associated cognitive impairment by promoting gut-brain metabolic remodeling. Targeting intestinal amino acid transport may represent a promising nutritional and therapeutic strategy for neuroprotection in diabetic encephalopathy (DE). Supplementary Information The online version contains supplementary material available at 10.1186/s12986-025-01024-w. Keywords: Cognitive dysfunction, Diabetes, SLC6A14, α-MT, Metabolite profiles Background Diabetic encephalopathy (DE) is a prevalent central nervous system complication of diabetes, characterized by progressive cognitive decline that significantly impairs quality of life [[44]1, [45]2]. Epidemiological evidence suggests that individuals with both type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM) exhibit mild-to-moderate declines in cognitive function compared to non-diabetic controls [[46]3]. While glycemic control remains a cornerstone in the management of diabetes and its associated complications, it may be insufficient to fully prevent or reverse cognitive decline in DE [[47]4]. Although some therapeutic strategies used for Alzheimer’s disease (AD) - a neurodegenerative disorder that shares overlapping pathogenic mechanisms with DE - have been explored in DE, their efficacy in modifying disease progression remains limited [[48]5, [49]6]. Therefore, novel therapeutic approaches specifically targeting the underlying mechanisms of DE are urgently needed. The gut-brain axis has emerged as a key regulator of cognitive function, with gut-derived metabolites increasingly recognized as critical mediators of brain health [[50]7, [51]8]. Metabolites such as short-chain fatty acids (SCFAs) [[52]9–[53]11], produced or modulated by the intestinal microbiota, can influence neuroinflammation, synaptic plasticity, and neuronal metabolism via neural, immune, and endocrine pathways. In diabetes, dysbiosis and intestinal barrier dysfunction alter the colonic metabolic environment, and accumulating evidence links these changes to hippocampal impairment and cognitive decline. For instance, reduced acetate levels have been shown to decrease hippocampal synaptophysin and accelerate cognitive deficits in type 1 diabetes [[54]12], while elevated gut-derived glutamate disrupts hippocampal signaling and correlates with cognitive dysfunction in type 2 diabetes [[55]13].These findings highlight the potential of targeting intestinal metabolism to modulate central nervous system outcomes. α-Methyltryptophan (α-MT) is a promising candidate for targeting gut-brain metabolic axis dysfunction in diabetes-associated cognitive impairment. α-MT is a well-established inhibitor of SLC6A14 (ATB⁰^,⁺), a broad-specificity amino acid transporter that mediates the uptake of nearly all amino acids except glutamate and aspartate [[56]14–[57]16]. This transporter is expressed in colon, where L-type enteroendocrine cells that secrete GLP-1, peptide YY, and pancreatic polypeptides are present [[58]17]. By modulating intestinal amino acid uptake, α-MT may reshape colonic metabolic output and systemic amino acid availability - key factors in gut-brain communication. Dysregulated amino acid homeostasis in diabetes may contribute to central nervous system dysfunction, and targeting intestinal transport offers a strategy to indirectly influence brain metabolism. Additionally, α-MT also inhibits indoleamine-2,3-dioxygenase-1 (IDO1) [[59]18], an enzyme catalyzing the initial step in tryptophan degradation via the kynurenine pathway. Increased IDO1 activity elevates kynurenine metabolites, which are implicated in neuroinflammation, oxidative stress, and neurotoxicity [[60]19–[61]21]. While both mechanisms may contribute to neuroprotection, the gut-restricted expression of SLC6A14 makes it a compelling target for probing gut-initiated metabolic influences on cognition. Notably, α-MT has demonstrated beneficial effects in diabetic nephropathy through modulation of renal and urinary metabolism, with modest improvements in glycemic control [[62]22], underscoring its broader metabolic regulatory properties. Preclinical studies further support its safety, with no significant adverse effects reported on behavior, pharmacokinetics, or organ function [[63]23–[64]25]. Together, these properties justify its investigation as a modulator of gut-brain metabolic crosstalk in diabetic cognitive dysfunction. Despite these pharmacological properties, a critical knowledge gap remains: whether α-MT–induced changes in colonic metabolism translate into coordinated metabolic remodeling in the brain and ultimately improve cognitive function in diabetes. The hippocampus, a brain region essential for spatial learning, memory formation, and integration [[65]26], is particularly vulnerable in DE. To address this gap, we employed ¹H NMR–based metabolomics to comprehensively profile metabolite signatures in both the colon and hippocampus of db/db mice. We hypothesized that α-MT treatment would induce metabolic reprogramming in the gut that correlates with improvements in hippocampal biochemistry and cognitive behavior, thereby providing mechanistic insights into the gut-brain metabolic crosstalk underlying DE. methods Cell cultures and treatments The human colonic epithelial cell line Caco-2 was cultured in DMEM supplemented with 10% FBS and 1% penicillin/streptomycin. For experiments, cells were treated for 24 h under the following conditions: (I) Control (5.5 mM glucose); (II) High glucose (HG, 25 mM glucose); (III) Control + 1.25 mM α-MT; (IV) Control + 2.5 mM α-MT; (V) HG + 1.25 mM α-MT; (VI) HG + 2.5 mM α-MT. The high-glucose treatment conditions were established based on previously reported studies [[66]27–[67]29]. Animals and experimental design Eight-week-old male db/db mice (C57BLKS/J-Lepr^db/db) and their wild-type (WT) littermates (C57BLKS/J-Lepr+/+) were obtained from Model Animal Research Center of Nanjing University. Mice were housed under specific pathogen-free (SPF) conditions with a 12-hour light/dark cycle and ad libitum access to food and water. All experimental procedures were approved by the Institutional Animal Care and Use Committee of Wenzhou Medical University (Approval No.: xmsq2022-1008). After one week of acclimatization, mice were randomly assigned to four groups (n = 7/group): (1) WT + vehicle (0.9% saline, 0.2 mL, i.p.); (2) WT + α-MT (DL-α-methyltryptophan, 2.5 mg per mouse in 0.2 mL saline, i.p.); (3) db/db + vehicle; (4) db/db + α-MT. The DL-form of α-MT (CAS No.: 153-91−3; C₁₂H₁₄N₂O₂: 218.25 g/mol; Shenzhen Qimeike Biotechnology Co., Ltd.) was administered every 48 h for 8 weeks. This treatment duration was designed to cover the critical period of diabetes-associated cognitive decline in db/db mice, which typically occurs between 16 and 20 weeks of age [[68]30, [69]31]. The dose and regimen were selected based on previous studies demonstrating its efficacy in modulating glucose metabolism without overt toxicity [[70]22]. Injections were performed between 9:00 AM and 11:00 AM to minimize circadian variation. We designed and illustrated a schematic diagram of the experimental workflow, as shown in Fig. [71]1A. Fig. 1. [72]Fig. 1 [73]Open in a new tab α-MT improves glucose homeostasis in db/db mice (n = 7). (A) Experimental Design Arrow Diagram. (B) Random blood glucose level; (C) IPGTT; (D): ITT. The treatment α-MT (i.p., 2.5 mg every other day) began at 9 weeks of age and was continued until 17 weeks of age. **p < 0.01, and ***p < 0.001 versus WT mice; ^###p < 0.001versus db/db mice. Statistical analysis: two-way repeated-measures ANOVA with Bonferroni post hoc tests Biochemical analysis Body weight and random blood glucose were monitored weekly. Intraperitoneal glucose tolerance tests (IPGTT) and insulin tolerance tests (ITT) were performed at week 17 after 4–6 h of fasting. For IPGTT, mice were injected with glucose (2 g/kg, i.p.), and blood glucose was measured at 0, 30, 60, and 120 min. For ITT, mice received insulin (0.75–1.0 U/kg, i.p.), and blood glucose was monitored at the same time point. Morris water maze test Spatial learning and memory were assessed using the Morris water maze as previously described [[74]32]. The assay was conducted in a circular pool (120 cm diameter) filled with opaque water (22 ± 1 °C), with a hidden platform (10 cm diameter) submerged 1 cm below the surface in a fixed quadrant. Mice underwent four training days with four trials per day. Each trial had a maximum duration of 60 s, during which mice were allowed to locate the platform. If unsuccessful, they were guided to the platform and allowed to remain for 15 s. Inter-trial intervals were approximately 15 min. Starting positions varied pseudorandomly across trials to avoid bias. On the fifth day, a probe trial was conducted with the platform removed; mice were released from a novel starting point and allowed to swim freely for 60 s. Performance was video-tracked and analyzed using EthoVision XT (Noldus Information Technology). Key metrics included escape latency, path efficiency, time spent in the target quadrant, and number of platform crossings. The experimenter was blinded to genotype and treatment groups throughout all behavioral procedures. Tissue collection and histopathology Mice were anesthetized with isoflurane. For histology, a subset of mice (n = 3/group) underwent transcardial perfusion with saline followed by 4% paraformaldehyde (PFA). Colon and brain tissues were post-fixed, paraffin-embedded, and sectioned for staining with Hematoxylin and Eosin (H&E), Alcian Blue/Periodic Acid-Schiff (AB-PAS), and immunohistochemistry (IHC) for ZO-1 and Occludin. Histopathological scoring of colon sections was performed by a blinded investigator using a established system evaluating inflammatory infiltration, tissue damage, crypt architecture, and goblet cell depletion (scores 0–3 per parameter, total score 0–12) [[75]33]. For molecular and metabolomic analyses, tissues from non-perfused mice (n = 4/group) were rapidly dissected, flash-frozen in liquid nitrogen, and stored at −80 °C. Metabolites extraction and metabolomic analysis Metabolites were extracted from colon and hippocampus tissues (~ 20 mg) using a methanol-chloroform-water method [[76]34]. After homogenization and centrifugation (10,000 g, 4 °C), supernatants were lyophilized and reconstituted in 0.6 mL D₂O containing 0.05% TSP-d4. ^1H NMR spectra were acquired at 298 K on a Bruker Avance III 600 MHz spectrometer with a 5 mm TXI probe. Hippocampus and colon samples were analyzed using zgpr and cpmgpr1d sequences, respectively, with 256 scans, 64,000 data points, a spectral width of 12,000 Hz, and a relaxation delay of 6 s. Spectra were processed in TopSpin 3.0: phased manually, baseline-corrected, and referenced to lactate (δ 1.31). Residual water (δ 4.70–5.00) and noisy regions (δ < 0.5, δ >9.5) were excluded. Data were binned (0.01 ppm for multivariate analysis; 0.0015 ppm for quantification), normalized to total spectral sum, and analyzed using PLS-DA in SIMCA-P 12.0. Metabolite concentrations were normalized to tissue weight and expressed relative to TSP in relative units (r.u.). MetaboAnalyst 6.0 ([77]https://www.metaboanalyst.ca/) was used to perform metabolic pathway analysis, integrating both pathway enrichment analysis and pathway topology analysis. RNA extraction and RT-qPCR analysis Total RNA was extracted from Caco-2 cells, colon and hippocampal tissues using TRIzol reagent (Invitrogen, Carlsbad, CA, USA) according to the manufacturer’s instructions [[78]35]. RNA quality was assessed by measuring A260/A280 ratios (>1.8) on a NanoDrop spectrophotometer. cDNA was synthesized from 2 µg of total RNA using the GoScript Reverse Transcription System (Promega) with oligo(dT) primers. Real-time quantitative PCR (RT-qPCR) was performed using SYBR Green Master Mix (Promega) on a CFX Connect Real-Time PCR Detection System (Bio-Rad). Each 20 µL reaction contained 0.2 µM of each primer and 2 µL of cDNA template. Primer efficiencies were validated using standard curves from serial cDNA dilutions. Relative gene expression was calculated using the 2⁻^ΔΔCt method with normalization to β-actin. All primer sequences (designed via NCBI Primer-BLAST and synthesized by Sangon Biotech) are provided in Table [79]1. Table 1. Primer sequences for RT-qPCR Gene Forward primer 5′−3′ Reverse primer 5′−3′ SLC6A14 CAAATCGTCTGGCAAGGTGG GCAACTAAGCCACCCCAA c-FOS AGGGGCAAAGTAGAGCAGCTA CAATCTCAGTCTGCAACGCA EGR1 AACCGGCCCAGCAAGACACC GGGCACAGGGGATGGGAATG ARC ACCATATGACCACCGGCGGC TCCAGCATCTCAGCTCGGCAC BDNF CCATAAGGACGCGGACTTGTAC AGACATGTTTGCGGCATCCAGG β-actin CCTCACTGTCCACCTTCC GGGTGTAAAACGCAGCTC [80]Open in a new tab Western blot analysis Protein lysates were prepared from colon tissues using RIPA lysis buffer supplemented with protease and phosphatase inhibitors. Total protein concentration was determined using a BCA assay, and 30 µg of protein per sample was separated by 10% SDS-PAGE. Proteins were transferred to a PVDF membrane and incubated overnight at 4 °C with a primary antibody against SLC6A14 (ab316193, Abcam). After washing with TBST, the membrane was incubated with an HRP-conjugated secondary antibody (ab6721, Abcam) for 2 h at room temperature. Immunoreactive bands were visualized using Pierce ECL substrate and analyzed with ImageQuant 5.2 software. β-actin (4970 S, Cell Signaling Technology) was used as a loading control. Statistical analysis Data are presented as mean ± standard deviation (SD) or median with interquartile range, as appropriate. Normality and homogeneity of variances were assessed using Shapiro-Wilk and Levene’s tests, respectively. Data meeting assumptions were analyzed by two-way ANOVA (factors: genotype and treatment) or two-way repeated-measures ANOVA (for longitudinal data), followed by Bonferroni’s post hoc test. Non-parametric data were analyzed by Kruskal-Wallis test with Bonferroni correction. Correlation analyses were performed using Pearson’s correlation with FDR correction. Statistical significance was set at p < 0.05. All analyses were performed using SPSS 19.0. Results α-MT improves glucose homeostasis in db/db mice α-MT treatment significantly influenced various metabolic parameters, with both main effects and interaction effects observed over time (Table [81]S1, Additional file 1). Specifically, α-MT significantly reduced random blood glucose levels in db/db mice compared to untreated diabetic controls at both 13 and 17 weeks (Fig. [82]1B and 13 weeks: p < 0.001, Cohen’s d = 6.955; 17 weeks: p < 0.001, Cohen’s d = 6.881). No significant changes were observed in WT mice, indicating that α-MT selectively targets hyperglycemia in diabetic animals. α-MT also improved glucose tolerance in db/db mice, as evidenced by IPGTT. At all tested time points (30, 60, and 120 min), α-MT-treated diabetic mice exhibited significantly lower blood glucose levels compared to untreated controls (Fig. [83]1C; all p < 0.001, Cohen’s d values ranging from 11.136 to 15.86). Similarly, ITT showed enhanced insulin sensitivity in α-MT-treated db/db mice at 30, 60, and 120 min (Fig. [84]1D; all p < 0.001, Cohen’s d values ranging from 5.230 to 8.077). These findings indicate that α-MT effectively ameliorates hyperglycemia and improves glucose metabolism in db/db mice. The selective improvement in diabetic animals suggests that α-MT may target specific pathways involved in diabetes-associated metabolic dysregulation. α-MT ameliorated cognitive function deficits in db/db mice To assess the impact of α-MT on cognitive function in db/db diabetic mice, we employed Morris water maze tests and RT-qPCR analysis. The swimming trajectories during the water maze test (Fig. [85]2A) revealed that db/db mice exhibited disorganized and less efficient paths compared to WT controls, indicative of cognitive deficits. Notably, α-MT treatment significantly improved the spatial navigation abilities of db/db mice, as evidenced by more organized and efficient motor trajectories. Significant effects of diabetes, time, and a three-way interaction (diabetes × treatment × time) on escape latency were observed (Table [86]S1, Additional file 1). Post hoc comparisons (Fig. [87]2B) further revealed that on day 4 of training, α-MT-treated db/db mice had significantly shorter escape latencies compared to untreated db/db mice (p < 0.001, Cohen’s d = 5.544), approaching levels similar to WT mice (p = 0.688, Cohen’s d=−0.875). This indicates that α-MT effectively ameliorates spatial learning deficits in db/db mice. In the platform crossing test assessing spatial memory retention, a significant interaction between diabetes and treatment was observed (p = 0.034, partial η² = 0.175, Table S2, Additional file 1). Simple effects analysis confirmed that α-MT specifically enhanced performance in db/db mice, with treated db/db mice exhibiting more platform crossings compared to untreated controls (p = 0.007, Cohen’s d=−1.562, Fig. [88]2C). No such effect was observed in WT mice. Fig. 2. [89]Fig. 2 [90]Open in a new tab α-MT improves cognitive function deficits in db/db mice. (A): Swimming trajectory in the water maze (n = 7); (B): Escape latency period (n = 7) (C): Number of platforms crossing (n = 7); mRNA levels for c-FOS (D), EGR1 (E), BDNF (F), and ARC (G) in hippocampus (n = 4). (H): The HE staining image of the hippocampus and a magnified image of the CA1 region. NG: neuronal degeneration. Statistical analysis: repeated-measures ANOVA (B), two-way ANOVA (C–G), with Bonferroni’s post hoc test Further analysis of hippocampal gene expression related to cognitive function provided insights into the molecular mechanisms underlying these improvements. Diabetes and its interaction with α-MT treatment had significant effects on key genes associated with neuroplasticity, including c-FOS, EGR1, BDNF, and ARC (Table S2, Additional file 1). Specifically, α-MT significantly upregulated c-FOS (p = 0.001, Cohen’s d=−3.109, Fig. [91]2D), EGR1 (p < 0.001, Cohen’s d=−5.615, Fig. [92]2E), BDNF (p < 0.001, Cohen’s d=−3.737, Fig. [93]2F), and ARC (p < 0.001, Cohen’s d=−3.739, Fig. [94]2G) expression in db/db mice, without affecting WT mice. Histological examination via HE staining of the hippocampus (Fig. [95]2H) revealed gliosis and neuronal deformities in the CA1 region of db/db mice (indicated by arrows in the figure). Following α-MT treatment, both gliosis and neuronal morphology showed significant improvement. In summary, our results demonstrate that α-MT effectively improves spatial learning and memory retention in db/db diabetic mice, as evidenced by reduced escape latency and increased platform crossings. The improvement in cognitive function may involve the upregulation of neurotrophic factors and early-response genes in the hippocampus. α-MT protects against colonic barrier damage and inhibits the expression of SLC6A14 colon of db/db mice Histological analysis revealed severe colonic damage in db/db mice, including submucosal edema, crypt loss, reduced goblet cells, and inflammatory infiltration (Fig. [96]3A–B). α-MT treatment markedly attenuated these abnormalities, as reflected in significantly improved histological scores (Fig. [97]3H). Goblet cell density and mucus production, assessed by AB-PAS staining, were also reduced in db/db mice but restored by α-MT (Fig. [98]3C–D, I). Statistical analysis confirmed significant main effects of diabetes and treatment, as well as a significant interaction (all p < 0.01, partial η² >0.50, Table S2, Additional file 1), indicating that α-MT specifically ameliorated structural damage in diabetic mice. Fig. 3. [99]Fig. 3 [100]Open in a new tab α-MT ameliorates colon damage and reduces the expression of SLC6A14 in db/db mice. (A) Representative H&E staining images of the colon and (B) a magnified view of the local area. a: submucosal edema; b: crypt injury; c: loss of goblet cells; d: inflammatory cell infiltration. (C) Alcian Blue–Periodic Acid-Schiff (AB-PAS) staining images of the colon and (D) a magnified view of the local area. (E) Representative immunohistochemical (IHC) staining images of ZO-1 and (F) Occludin in the colon (n = 4). (G) Expression levels of SLC6A14 protein in the colon. (H) Histological scores of the colon (n = 4). Quantitative analysis of (I) AB-PAS-positive area (n = 4), (J) ZO-1-positive area (n = 4), and (K) Occludin-positive area (n = 4). (L) Quantification of western blot analysis of SLC6A14 (n = 3). (N) Expression of SLC6A14 mRNA in the colon (n = 3). (M) Expression of SLC6A14 mRNA in Caco-2 cells (n = 4). Note: Con: Normal medium; Con-1.25 α-MT: Normal medium with 1.25 mmol/L α-MT; Con-2.5 α-MT: Normal medium with 2.5 mmol/L α-MT; HG (high glucose medium): Normal medium with 25 mmol/L glucose; HG-1.25 α-MT: High glucose medium with 1.25 mmol/L α-MT; HG-2.5 α-MT: High glucose medium with 2.5 mmol/L α-MT. Statistical analysis: Kruskal-Wallis H tests with Bonferroni correction (L-M), two-way ANOVA with Bonferroni’s post hoc test (H-K) IHC showed decreased expression of tight junction proteins ZO-1 and Occludin in db/db mice, which was reversed by α-MT treatment (Fig. [101]3E–F, J–K). Significant interactions between diabetes and treatment were observed for both proteins (p < 0.001, partial η² >0.70, Table S2, Additional file 1), supporting a protective effect of α-MT on intestinal barrier integrity. Given that α-MT inhibits the amino acid transporter SLC6A14, we examined its expression in the colon. Both protein and mRNA levels of SLC6A14 were elevated in db/db mice and significantly reduced by α-MT (Fig. [102]3G, L, N; p < 0.05, H > 9.0). In vitro, high-glucose conditions upregulated SLC6A14 mRNA in Caco-2 cells, an effect significantly suppressed by α-MT (Fig. [103]3M; p < 0.05, H = 16.322). Collectively, these results demonstrate that α-MT alleviates diabetes-induced colonic barrier dysfunction, associated with downregulation of SLC6A14 and restoration of tight junction and mucosal integrity. α-MT treatment reverses diabetes-induced alterations in colon and hippocampal metabolite profiles To investigate the impact of diabetes and α-MT treatment on metabolic profiles, we performed ^1H-NMR analysis of colon and hippocampus tissues from WT and db/db mice, with or without α-MT treatment. Representative spectra are shown in Fig. [104]4A and B. A total of 37 metabolites were identified, including short-chain fatty acids (acetate, propionate, butyrate, formate), energy-related compounds (glucose, UDP-glucose, lactate, fumarate, succinate, creatine, AMP, IMP, nicotinamide), amino acids (alanine, glycine, taurine, glutamate, glutamine, aspartate, leucine, isoleucine, valine, methionine, phenylalanine, tyrosine, histidine, GABA, 4-aminobutyrate, N-acetylglutamine, 3-hydroxyisovalerate), choline and its derivatives (choline, phosphocholine, dimethylamine, myo-inositol), and purines and pyrimidines (xanthine, adenine, hypoxanthine, inosine, uracil, uridine, pyridoxine). Notably, similar metabolic pathways were altered in both colon and hippocampus, suggesting a potential gut–brain axis involvement in diabetes-related metabolic dysregulation. Fig. 4. [105]Fig. 4 [106]Open in a new tab Metabolic analysis of the colon and hippocampus of WT and db/db mice with and without α-MT treatment. (A-B): Typical 600 MHz ^1H NMR spectra for metabolites from colon (A) and hippocampus (B) of db/db mice. (C-H): The changes in metabolic phenotypes observed in the colon and hippocampus of WT and db/db mice after treatment with α-MT. The PLS-DA score plot was used to identify the variations in the colon metabolomic profile among the four groups (C), between WT and db/db mice (D), and between db/db and α-MT-treated db/db mice (E). The PLS-DA score plot in the hippocampal metabolomic profile among four groups (F), between WT and db/db mice (G), and between db/db and α-MT-treated db/db mice (H). 1, Leucine; 2, Valine; 3, Isoleucine; 4, Propionate; 5, Ethanol; 6, Alanine; 7, Butyrate; 8, Acetate; 9, Glutamate; 10, Glutamine; 11, Methionine; 12, Aspartate; 13, Trimethylamine (TMA); 14, Creatine; 15, Ethanolamine; 16, Choline; 17, Phosphocholine (PC); 18, Taurine; 19, Isocitrate; 20, Glycine; 21, Myo-inositol; 22, Lactate; 23, 3-Hydroxyisovalerate (3-HV); 24, Glucose; 25, Uracil; 26, Uridine; 27, UDP-Glucose; 28, Fumarate; 29, Tyrosine; 30, Phenylalanine; 31, Inosine; 32, Pyridoxine; 33, Xanthine; 34, Adenine; 35, Histidine; 36, Inosine monophosphate (IMP); 37, Formate; 38, Adenosine monophosphate (AMP); 39, Niacinamide; 40, N-Acetylglutamine; 41, 4-Aminobutyrate; 42, Dimethylamine 43. Hypoxanthine. Note: hippo: hippocampus The PLS-DA model was used to determine changes in the metabolite patterns of the WT, db/db, and α-MT-treated groups. Score plots based on the colon and hippocampus metabolomes (Fig. [107]4C and F) clearly showed differences between the WT, db/db, and α-MT-treated db/db groups. Notably, there was a certain degree of overlap in the distribution of the WT and α-MT-treated WT groups in the PLS-DA score map, suggesting that α-MT does not cause significant metabolite profile alterations in mice under normal physiological conditions. The metabolite profile of the α-MT-treated db/db group was closer to that of the WT group than that of the db/db group, suggesting that α-MT treatment reverses the diabetes-induced changes in colon and hippocampal metabolite profiles. Subsequently, we performed PLS-DA analysis by comparing the db/db group with the WT group, and the db/db + α-MT group with the WT group. Significant differences were observed between the db/db and WT groups in both colon (Fig. [108]4D) and hippocampal (Fig. [109]4G) metabolomes. Additionally, distinct metabolic profiles were detected between the db/db and α-MT-treated db/db groups in both colon (Fig. [110]4E) and hippocampus (Fig. [111]4H). These results indicate that α-MT treatment ameliorates the metabolic profiles in the colon and hippocampus induced by diabetes, affecting multiple metabolic pathways. Amino acid metabolism is a key target of α-MT in diabetic colon and hippocampus To further investigate the key metabolic pathways modulated by α-MT in diabetic mice, we analyzed metabolite profiles in the colon and hippocampus. Heatmap visualization revealed widespread elevations of multiple metabolites in both colon (Fig. [112]5A) and hippocampus (Fig. [113]5B) of db/db mice compared to WT. These aberrant metabolite levels were broadly attenuated following α-MT treatment, suggesting a significant metabolic regulatory effect of α-MT. Fig. 5. [114]Fig. 5 [115]Open in a new tab Heatmap for variations in metabolites after α-MT treatment. The heatmap for the colon (A) and hippocampus (B) of the four groups of mice (WT, WT + α-MT, db/db, db/db + α-MT). The shades of red (color scale > 0) represent a relative higher concentration of metabolites, while the shades of blue (color scale < 0) indicate a relative lower concentration of metabolites. Abbreviation: 3-HV: 3-Hydroxyisovalerate; AMP, Adenosine monophosphate; IMP: Inosine monophosphate; DMA, Dimethylamine Pathway enrichment analysis was subsequently performed. In the colon (Fig. [116]6A), key differential metabolites included taurine, phenylalanine, tyrosine, alanine, aspartate, glutamate, and glutamine—all of which are associated with amino acid metabolism. In the hippocampus (Fig. [117]6B), key metabolites comprised nicotinamide, fumarate, alanine, glutamate, glutamine, 4-aminobutyrate, glycine, choline, creatine, and taurine. Notably, amino acid metabolism emerged as a significantly enriched pathway in both tissues, indicating it as a central node targeted by α-MT (Fig. [118]6A–B). Fig. 6. [119]Fig. 6 [120]Open in a new tab Identification of Key Metabolic Pathways. Metabolic pathways in the colon (A) and hippocampus (B) were identified using MetaboAnalyst, based on p-values from pathway enrichment analysis and pathway impact values from pathway topology analysis. (C) Metabolic network diagram of selected metabolites from the colon and hippocampus as analyzed by Metabo Analyst. Bars with black borders represent metabolites identified in the colon; bars with yellow borders represent metabolites identified in the hippocampus. Black font indicates metabolites specific to the colon; Yellow font indicates metabolites specific to the hippocampus. Red font indicates metabolites present in both the hippocampus and the colon. Statistical notations: *p < 0.05, **p < 0.01, ***p < 0.001 versus WT mice; ^#p < 0.05, ^##p < 0.01, ^###p < 0.001 versus db/db mice. Abbreviation: GABA, γ-aminobutyric acid. Statistical analysis: Kruskal-Wallis H tests with Bonferroni correction (C) To integrate and visualize the metabolic crosstalk between gut and brain, we constructed a network mapping of the key metabolites identified in both colon and hippocampus (Fig. [121]6C). All metabolites in this network—except aspartate—showed an overall p-value < 0.05, with H-values ranging from 7.691 to 11.868. Compared to WT mice, all selected key metabolites (except aspartate) were significantly elevated in db/db mice (all p < 0.05) and markedly reduced following α-MT intervention (all p < 0.05). Furthermore, network analysis revealed that taurine, alanine, glutamate, and glutamine were overlapping key metabolites shared between colon and hippocampus, whose levels were effectively normalized by α-MT treatment. These findings suggest that amino acid metabolism in the colon and hippocampus may contribute to the beneficial effects of α-MT in DE. Gut–brain metabolic coordination is evident in diabetic mice To investigate functional coupling between the gut and brain in diabetes, we performed Pearson correlation analysis between key metabolites in the colon and hippocampus (Fig. [122]7A). A widespread positive correlation was observed, indicating coordinated metabolic dysregulation across the gut–brain axis. Notably, colonic taurine—a metabolite elevated in db/db mice and normalized by α-MT - showed strongest associations with hippocampal metabolism, including glycine (r = 0.81, p < 0.001), niacinamide (r = 0.80, p < 0.001), and fumarate (r = 0.81, p = 0.001). It also correlated significantly with multiple other hippocampal metabolites (alanine, glutamate, GABA, creatine; r = 0.70–0.77, p < 0.01). Other colonic amino acids, including alanine, tyrosine, and phenylalanine, similarly showed broad positive correlations with hippocampal metabolites involved in neurotransmission and energy metabolism (e.g., glutamine, fumarate, niacinamide; r = 0.70–0.79, p < 0.01), suggesting a systemic gut–brain metabolic linkage in diabetes. Fig. 7. [123]Fig. 7 [124]Open in a new tab Pearson correlation analysis involving colonic and hippocampal metabolites and cognitive-related genes. (A) Heatmap of correlations between colonic metabolites (columns) and hippocampal metabolites (rows). (B) Heatmap of correlations between colonic metabolites (columns) and gene expression (c-FOS, EGR1, BDNF, ARC; rows). (C) Heatmap of correlations between hippocampal metabolites (columns) and gene expression (c-FOS, EGR1, BDNF, ARC; rows). Cell values represent Pearson correlation coefficients (range: −1 [blue] to 1 [red]); asterisks denote statistical significance (*p < 0.05, **p < 0.01, ***p < 0.001) Metabolic dysregulation is inversely associated with cognitive gene expression We next examined the relationship between metabolite levels and hippocampal expression of neuroplasticity-related genes (BDNF, c-FOS, EGR1, ARC). Both colonic and hippocampal metabolites showed predominantly negative correlations with these cognition-associated transcripts (Fig. [125]7B–C). In the colon (Fig. [126]7B), glutamine, taurine, and phenylalanine were strongly and negatively correlated with cognitive gene expression. Glutamine exhibited the most consistent associations (r = − 0.71 to − 0.75, p < 0.01 across all four genes), while taurine and phenylalanine showed moderate-to-strong negative correlations (r = − 0.52 to − 0.74, p < 0.05). In the hippocampus (Fig. [127]7C), the negative associations were even more pronounced. BDNF expression was widely and strongly inversely correlated with metabolites such as alanine (r = − 0.85), glycine (r = − 0.82), glutamate (r = − 0.84), GABA (r = − 0.81), and choline (r = − 0.83; all p < 0.001). Similarly, ARC and c-FOS showed significant negative correlations with multiple key metabolites, reinforcing the link between metabolic imbalance and impaired synaptic plasticity. EGR1 exhibited moderate negative correlations with key hippocampal metabolites, excluding glutamine, with Pearson’s r values between − 0.5 and − 0.6, all statistically significant (p < 0.05). Taken together, the widespread negative correlations between gut and hippocampal metabolite levels and cognition-associated gene expression suggest that modulating gut–brain metabolism may enhance the expression of neuroplasticity-related genes and improve cognitive function. Proposed mechanism of α-MT in ameliorating cognitive dysfunction Collectively, these findings support a gut–brain metabolic axis in diabetic cognitive impairment (summarized in Fig. [128]8). α-MT treatment downregulates colonic SLC6A14, reducing excessive amino acid uptake and restoring colonic metabolic homeostasis. This gut-level correction is positively correlated with improved metabolic profiles in the hippocampus, particularly in pathways linked to amino acids, energy, choline and nicotinamide metabolism. In turn, the normalization of these brain metabolites is inversely associated with the expression of neuroplasticity markers (BDNF, c-FOS, EGR1, ARC), which are upregulated following α-MT treatment. These data suggest that α-MT may indirectly support cognitive function through modulation of gut-brain metabolic crosstalk, although direct causal mechanisms require further investigation. Fig. 8. [129]Fig. 8 [130]Open in a new tab Potential mechanisms by which α-MT ameliorates diabetes-induced cognitive decline in mice Discussion Diabetes mellitus is a complex metabolic disorder characterized by chronic hyperglycemia due to impaired insulin secretion or action. Chronic glucose dysregulation extends beyond peripheral complications, contributing to DE and impairments in learning, memory, and executive function. These cognitive deficits significantly impair quality of life, highlighting the need for neuroprotective strategies. Diabetes-associated cognitive decline involves multiple mechanisms, including metabolic dysregulation, inflammation, oxidative stress, and mitochondrial dysfunction [[131]36, [132]37]. Hyperglycemia promotes the formation of advanced glycation end products (AGEs), activation of protein kinase C (PKC), and oxidative stress, leading to synaptic dysfunction and neuronal damage [[133]38]. Emerging evidence suggests gut barrier disruption in diabetes may allow microbial products like LPS to enter circulation, contributing to systemic inflammation and CNS effects through gut-brain interactions, independent of glycemic control [[134]39, [135]40]. Given that α-MT ameliorates diabetic nephropathy through metabolic modulation [[136]21], we investigated its potential neuroprotective effects in DE. Our findings suggest that α-MT improves cognitive function through mechanisms largely independent of direct glycemic control. Cognitive improvements, assessed by Morris water maze performance and memory-related gene expression (BDNF, c-FOS, ARC, EGR1), were more pronounced than changes in blood glucose levels. Metabolomic analyses identified amino acid metabolism as the most significantly altered pathway in both the colon and hippocampus, with additional changes in energy, choline, and nicotinamide metabolism in the brain. The absence of glycolytic or glucose-related pathways among the significantly enriched pathways supports that α-MT’s effects are not primarily mediated by glucose modulation. α-MT also improved intestinal morphology and barrier integrity in diabetic mice. Positive correlations between colonic and hippocampal amino acid levels, and their association with memory-related gene expression, suggest that α-MT may influence brain function through systemic metabolic changes, potentially involving gut-brain interactions. α-MT attenuated the high-glucose-induced upregulation of SLC6A14 in the colon, and this attenuation was associated with improved colonic and hippocampal amino acid metabolism in db/db mice, suggesting a systemic metabolic effect. Amino acids are critical for neurotransmission, energy metabolism, and neuroplasticity, all of which support cognitive function [[137]41, [138]42]. We observed significant dysregulation of hippocampal amino acids—including glutamate, glutamine, alanine, glycine, GABA, and taurine—in db/db mice, which were partially normalized by α-MT treatment. Glycine, a neurotransmitter and neuromodulator involved in neuroprotection and metabolism, plays an important role in cognitive function, and its dysregulation has been linked to cognitive impairment [[139]43, [140]44]. Glutamate and GABA are the primary excitatory and inhibitory neurotransmitters, respectively, and their metabolic coupling with glutamine is essential for cognitive function; disruption of this cycle is associated with cognitive impairment in diabetic models [[141]45, [142]46], a phenomenon already substantiated in mouse models of diabetes-associated cognitive dysfunction. Taurine modulates the glutamatergic system and synaptic plasticity, and its dysregulation may contribute to learning and memory deficits [[143]47]. Notably, the concentrations of most amino acid metabolites in the hippocampus showed a significant negative correlation with the expression levels of genes such as BDNF, c-FOS, ARC, and EGR1. BDNF, a key regulator of synaptic plasticity [[144]48], was downregulated in db/db mice and negatively correlated with elevated glutamate and glutamine levels, suggesting that metabolic disturbances may impair neurotrophic signaling. c-FOS, a marker of neuronal activation [[145]49], was downregulated in diabetic mice and was upregulated by α-MT treatment. ARC and EGR1, which regulate synaptic remodeling and memory [[146]50, [147]51], were also dysregulated and negatively correlated with amino acid levels, reinforcing the link between metabolic imbalance and cognitive dysfunction. In summary, hippocampal amino acid dysregulation may contribute to diabetes-associated cognitive dysfunction, and α-MT appears to alleviate these deficits by restoring metabolic balance and supporting the expression of neuroplasticity-related genes. Beyond amino acid dysregulation, impaired energy metabolism also contributes to cognitive deficits in diabetic mice. The brain primarily depends on glucose, which is metabolized via glycolysis and oxidative phosphorylation to produce ATP. Efficient energy metabolism supports cognitive functions such as attention, memory, and learning [[148]52]. Cognitive disorders including Alzheimer’s disease [[149]53] and vascular dementia [[150]54] are associated with impaired brain energy metabolism. These include reduced glucose uptake, insulin resistance, and mitochondrial dysfunction, which limit energy production and increase oxidative stress. Such disturbances are linked to neuronal dysfunction, synaptic loss, and neurodegenerative pathology, contributing to cognitive decline [[151]55]. We found elevated hippocampal levels of fumarate and creatine in db/db mice, which were significantly reduced by α-MT treatment. Fumarate, a TCA cycle intermediate, plays a role in energy production and redox homeostasis. Elevated fumarate is associated with mitochondrial dysfunction and oxidative stress, impairing neuronal energy metabolism [[152]56]. The reduction of fumarate by α-MT may reflect improved TCA cycle function and reduced metabolic stress in the diabetic hippocampus. Creatine supports rapid energy buffering through phosphocreatine (PCr), especially during high-energy processes like synaptic transmission [[153]57, [154]58]. The PCr/creatine system acts as a transient ATP buffer, helping maintain energy homeostasis in the brain, particularly in the hippocampus. In db/db mice, elevated hippocampal creatine was not linked to improved cognition and was modestly associated with reduced neuronal activity. Elevated creatine levels may not be functionally beneficial and could instead indicate metabolic dysregulation in the diabetic brain. α-MT treatment significantly reduced creatine levels, normalizing them toward control values; this restoration of metabolic balance may support improved synaptic function in the hippocampus. The functional significance of elevated creatine in db/db mice remains unclear and warrants further investigation to determine whether it is compensatory, neutral, or detrimental. We also observed negative correlations between hippocampal energy metabolites and the expression of the synaptic plasticity genes, suggesting a link between metabolic dysregulation and impaired neuronal plasticity. Nicotinamide, a precursor of NAD+, participates in redox reactions, DNA repair, and cellular signaling. In this study, hippocampal nicotinamide levels were significantly higher in db/db mice than in controls, and α-MT treatment significantly reduced these levels, suggesting improved NAD⁺ metabolism and redox homeostasis. Correlation analysis revealed positive associations between hippocampal nicotinamide levels and colonic concentrations of taurine, phenylalanine, tyrosine, and alanine—amino acids involved in neuroprotection, neurotransmission, and energy metabolism [[155]59–[156]61]. Additionally, hippocampal nicotinamide levels were inversely correlated with BDNF and c-FOS expression, indicating a potential link between elevated nicotinamide and impaired synaptic plasticity. Thus, the reduction in nicotinamide following α-MT treatment might contribute to improved gene expression and cognition, but this causal relationship requires experimental validation. Choline is a precursor of acetylcholine, a key neurotransmitter in cognitive processes, and is particularly important in the hippocampus [[157]62]. Choline metabolism interacts with glutamatergic and GABAergic systems, contributing to excitatory-inhibitory balance in the brain. Dysregulation of choline metabolism disrupt this balance and impair cognitive function [[158]63]. Hippocampal levels of choline and related metabolites were significantly altered in db/db mice, with choline, phosphorylcholine, and dimethylamine all significantly elevated. α-MT treatment significantly reduced these elevated metabolite levels, normalizing them toward control values. Notably, we observed a significant negative correlation between hippocampal choline levels and the expression of neuroplasticity-related genes. The positive correlation between hippocampal choline and colonic taurine suggests a potential gut-brain metabolic link and reflects systemic metabolic changes influencing brain function. Furthermore, hippocampal choline levels were negatively correlated with BDNF and c-FOS expression, indicating that elevated choline may be associated with impaired synaptic plasticity. Collectively, these results suggest that dysregulated hippocampal choline metabolism may contribute to cognitive impairment in diabetic mice. Conclusion Our metabolomic analysis indicates that α-MT is associated with improved cognitive performance in diabetic mice. α-MT reduced the overexpression of the colonic amino acid transporter SLC6A14, which was linked to improved gut barrier integrity and normalized colonic amino acid metabolism. These peripheral changes correlated with improved hippocampal metabolism across multiple pathways, consistent with a potential gut-brain interaction. Metabolic improvements were associated with partial restoration of synaptic plasticity gene expression (BDNF, ARC, EGR1, C-FOS) and cognitive function. Although these findings suggest systemic effects of α-MT, the underlying causal mechanisms require further investigation. This study supports the exploration of intestinal nutrient metabolism as a potential target for addressing DE. Limitations and future perspectives First, the study used only male mice, had a limited sample size, and an 8-week intervention period. Future studies should include both sexes, larger cohorts, and longer durations to enhance generalizability. Second, correlations do not establish causation; thus, mechanistic studies—such as gut microbiota profiling or SLC6A14 knockout models—are required to validate the gut-brain metabolic link. Finally, the pharmacokinetics, safety, and human translatability of α-MT remain poorly characterized. Further research is needed to assess its clinical potential and the translatability of α-MT and related analogs. Supplementary Information [159]Supplementary Material 1.^ (12.9KB, xlsx) Acknowledgements