Abstract
Metabolic and bariatric surgery induces metabolic benefits beyond weight loss, including improved insulin sensitivity, type 2 diabetes (T2D) remission, and reduced inflammation. Recent metabolomics research highlights amino acid metabolites—branched-chain amino acids, aromatic amino acids, and tryptophan-derived compounds—as key biomarkers for predicting surgical outcomes. Elevated preoperative levels of isoleucine, phenylalanine, levodopa, and 3-hydroxyanthranilic acid are associated with improved glycemic control and T2D remission. Gut microbiota-derived metabolites, including indole-3-propionic acid and indole-3-pyruvic acid, contribute to insulin sensitivity and lean mass preservation. Additionally, serotonin-related indicators predict postoperative weight loss rates. These metabolites reflect individual metabolic heterogeneity and may outperform conventional clinical models in predicting surgical responsiveness. Integration of metabolomics into preoperative assessment may enhance precision medicine approaches and identify new therapeutic targets.
Keywords: Bariatric surgery, Metabolomics, Amino acid
INTRODUCTION
Metabolic and bariatric surgery (MBS) is known as the most effective method for treating various metabolic diseases, including severe obesity and type 2 diabetes (T2D). Beyond weight loss, systemic metabolic effects such as increased insulin sensitivity, improved glycemic control, reduced hepatic fat, and regulated blood lipid levels have been reported after surgery. However, the molecular mechanisms that induce these clinical effects are not yet clearly understood. Recent advancements in metabolomics technology provide important clues to understanding these mechanisms by quantitatively analyzing changes in metabolites before and after MBS. In particular, among amino acid metabolites, branched-chain amino acids (BCAAs), aromatic amino acids (AAAs), and tryptophan-derived metabolites show a close association with metabolic health and play a key role in predicting and understanding surgical effects.
ASSOCIATION BETWEEN OBESITY AND AMINO ACID METABOLITES
1. BCAA (isoleucine, leucine, valine)
BCAAs consist of isoleucine, leucine, and valine [1], and play important roles in energy metabolism, muscle growth, and the regulation of insulin action [2,3]. In obese patients, blood concentrations of BCAAs are consistently high, which is strongly associated with insulin resistance [4]. This accumulation of BCAAs is related to a decrease in oxidative capacity in the liver and muscles, meaning a reduction in the efficiency of catabolic pathways, and leads to the inhibition of insulin signaling through the mammalian target of rapamycin complex 1 pathway [5,6]. Furthermore, BCAAs contribute to the progression of metabolic diseases through mechanisms such as inducing inflammation in adipocytes, disrupting the tricarboxylic acid cycle, and causing glucose metabolic imbalance [7,8].
Blood BCAA concentrations significantly decrease after MBS, which is interpreted as an indicator reflecting the recovery of metabolic function in the liver and muscles, beyond a simple weight loss effect [9,10]. Especially for isoleucine, studies have shown that higher preoperative concentrations are associated with greater postoperative weight loss, demonstrating its potential as a prognostic biomarker [11]. Additionally, gut microbiota have also been found to influence BCAA metabolism, with reports indicating that changes in microbial communities after surgery affect BCAA concentrations [12].
2. Phenylalanine and tyrosine
Phenylalanine and tyrosine are AAAs that are closely associated with liver function and neurotransmitter biosynthesis [13]. In obese conditions, blood concentrations of these amino acids increase, reflecting impaired liver detoxification capacity and metabolic stress [14,15]. Phenylalanine, in particular, is linked to systemic inflammatory markers, and persistent high concentrations can lead to metabolic diseases such as impaired liver function, increased insulin resistance, and worsening cardiovascular risk factors [16,17]. Tyrosine acts as a precursor for dopamine and norepinephrine, and these neurotransmitters are linked to the regulation of energy intake, reward behavior, and pancreatic beta-cell function [18,19]. Preoperative tyrosine and levodopa (L-DOPA) concentrations showed a correlation with postoperative T2D remission, suggesting that this pathway directly influences insulin secretion and sensitivity regulation [20,21]. Furthermore, dopamine can inhibit insulin secretion through anti-incretin action, so a decrease in dopamine production may improve insulin response [22]. Therefore, these AAAs can be considered causative agents of metabolic diseases and biological indicators that can predict surgical outcomes.
3. Tryptophan and its derivatives
Tryptophan is an essential amino acid that is metabolized through three main pathways: the kynurenine pathway, the serotonin pathway, and the indole pathway [23]. These pathways respectively mediate immune regulation, energy storage and behavioral control, and interaction with gut microbiota [24,25]. In obesity, the kynurenine pathway is abnormally activated, and concentrations of kynurenine and its downstream metabolites (e.g. 3-hydroxyanthranilic acid [3-HAA], kynurenic acid) increase due to the overactivity of the inflammation-inducing enzyme indoleamine 2,3-dioxygenase [26]. These metabolites affect immune cell function and insulin sensitivity, and high concentrations can lead to metabolic disorders [27,28].
The serotonin pathway involves the conversion of tryptophan to serotonin via 5-hydroxytryptophan (5-HTP) [29]. Peripheral serotonin induces lipid accumulation in adipocytes and regulates pancreatic insulin secretion [30]. Studies have shown that higher preoperative serotonin concentrations are associated with a slower rate of postoperative weight loss, and the serotonin/5-HTP ratio has been proposed as an indicator for predicting metabolic responsiveness [11].
The indole pathway is formed by the breakdown of tryptophan by gut microbiota and includes indole-3-propionic acid (IPA), indole-3-acetic acid (IAA), and indole-3-lactic acid [31]. In particular, IPA has antioxidant effects, protects the intestinal mucosa, and improves insulin sensitivity, and an increase in IPA concentration after surgery is associated with positive metabolic responses [32,33]. As such, tryptophan-derived metabolites, independently yet interactively, are considered key factors regulating the pathophysiology of obesity and metabolic diseases, as well as surgical responsiveness.
ASSOCIATION BETWEEN MBS AND AMINO ACID METABOLITES
1. Improvement of post-surgical insulin secretion and resistance with amino acid and gut microbiota metabolites
We evaluated the relationship between amino acids and tryptophan-derived gut microbiota metabolites before and after MBS and the improvement of insulin secretion and insulin resistance [34]. Among various amino acid metabolites measured before surgery, phenylalanine, leucine, tryptophan, and tyrosine showed a significant correlation with changes in the insulinogenic index and insulin resistance indices after surgery. In particular, the sum of large neutral amino acids and phenylalanine showed the highest predictive power, with area under the curve (AUC) values of 0.90–0.94 in receiver operating characteristic (ROC) analysis, suggesting their potential as biomarkers to predict improved insulin secretion and sensitivity based solely on preoperative concentrations.
Furthermore, values of 3-HAA, 3-hydroxykynurenine, xanthurenic acid, and anthranilic acid, belonging to the kynurenine pathway of tryptophan metabolism, showed a significant correlation with the improvement of insulin resistance after surgery. These metabolites possess biological properties such as inflammation regulation, oxidative stress alleviation, and pancreatic beta-cell protection [35,36], with 3-HAA in particular demonstrating its potential as a sensitive indicator of surgical responsiveness through its anti-inflammatory and antioxidant functions [37]. Additionally, changes in IAA and IPA among gut microbiota-derived indole metabolites were also associated with improved insulin resistance and secretion, implying that changes in the gut microbiota metabolic environment after MBS can contribute to metabolic improvement [38,39]. This study suggests that various amino acid metabolites and gut microbiota metabolites can act as regulators and predictive indicators of surgical responsiveness, demonstrating the possibility of patient selection based on precision medicine and biomarker-based predictive model development in the future (Table 1).
Table 1. Amino acid metabolites and clinical relevance.
| Metabolite | Metabolic role | Clinical insight |
|---|---|---|
| Isoleucine | Energy, insulin sensitivity | ↑ Pre-op → ↑ post-op weight loss (%EWL) |
| Phenylalanine | Oxidative stress, insulin resistance | Marker of metabolic stress, predictor of T2D improvement |
| L-DOPA | β-cell signaling, insulin secretion | ↑ Pre-op → predictor of T2D remission |
| 3-HAA | Antioxidant, β-cell protection | Strongest predictor of insulin resistance improvement |
| Serotonin/5-HTP ratio | Lipogenesis, energy intake | ↑ Pre-op → slower weight loss post-op |
| IPA | Antioxidant, mucosal barrier, insulin sensitizer | ↑ Post-op → better insulin function |
| IPyA | Muscle regeneration, anti-atrophy | ↑ Pre-op → predicts FFM preservation post-surgery |
%EWL = percentage of excess weight loss, T2D = type 2 diabetes, L-DOPA = levodopa, 3-HAA = 3-hydroxyanthranilic acid, 5-HTP = 5-hydroxytryptophan, IPA = indole-3-propionic acid, IPyA = indole-3-pyruvic acid, FFM = fat-free mass.
2. Prediction of post-surgical diabetes remission based on amino acid metabolites
We evaluated whether preoperative blood amino acid metabolite concentrations can predict remission of T2D and compared their performance with existing clinical predictive models [40]. A total of 16 glucose metabolism-related amino acid metabolites were analyzed, among which L-DOPA (tyrosine pathway) and 3-HAA (kynurenine pathway) were identified as the best metabolites for predicting T2D remission at 12 months post-surgery. The baseline concentrations of these two metabolites were significantly higher in the remission group, and their AUC values were 0.92 and 0.85, respectively, showing superior performance compared to existing clinical predictive models such as ABCD (0.81), DiaRem, and IMS.
L-DOPA, as a dopamine precursor, is involved in the auto-inhibitory signals of pancreatic beta-cells, and higher preoperative concentrations were associated with better recovery of blood glucose control and insulin secretion after surgery. On the other hand, 3-HAA, an anti-inflammatory metabolite of the tryptophan-kynurenine pathway, is interpreted to have a clearer effect in reducing metabolic stress and protecting beta-cells at higher concentrations. The levels of these metabolites maintained their predictive power up to 3 months post-surgery, suggesting that the initial metabolic state can influence long-term glycemic control outcomes after surgery. In particular, this study demonstrated that metabolites can provide 'qualitative' information about metabolic status that existing clinical indicators do not capture, highlighting the potential of metabolites as unique indicators reflecting physiological heterogeneity at the molecular level, independent of quantitative values (age, body mass index, diabetes duration). In conclusion, this study provides strong evidence that L-DOPA and 3-HAA can be used as preoperative predictive biomarkers, supporting the need for metabolite-based precision predictive model development.
3. Prediction of early weight loss rate using metabolites
The rate of weight loss among patients undergoing sleeve gastrectomy varies significantly, and preoperative metabolic status can influence the outcome. This study showed that some of the 20 obesity-related amino acid metabolites measured preoperatively were significantly associated with the percentage of excess weight loss (%EWL) at 3 and 6 months post-surgery [11]. Specifically, isoleucine among BCAAs showed a significant positive correlation with postoperative %EWL, while metabolites of the serotonin pathway, namely serotonin, 5-hydroxyindoleacetic acid (5-HIAA), and the serotonin/5-HTP ratio, showed strong performance in predicting slow weight loss after surgery. Higher serotonin concentrations and higher serotonin/5-HTP ratios were associated with slower weight loss, while conversely, higher 5-HIAA and 5-HIAA/serotonin ratios tended to be associated with faster weight loss. This result is interpreted as being related to the energy storage promotion, increased insulin secretion, and fat accumulation promotion functions of peripheral serotonin. In ROC analysis, serotonin and the serotonin/5-HTP ratio showed high predictive power with AUC values of 0.79–0.81, suggesting that preoperative serotonin metabolic profiles can be valid biomarkers for predicting early weight loss rates.
This study also analyzed ratio indicators reflecting enzyme activity of the serotonin pathway (tryptophan hydroxylase, aromatic L-amino acid decarboxylase, monoamine oxidase A), emphasizing the importance of sophisticated metabolite analysis reflecting even the enzyme activity status in metabolic pathways for predicting surgical responsiveness. This approach, by utilizing information on pathway-based functional metabolic flow in addition to single metabolite concentrations, provides important evidence for the establishment of future metabolite-based precision medicine strategies.
4. Preservation of lean body mass after surgery with tryptophan-derived metabolites
Rapid weight loss after MBS inevitably accompanies the loss of fat-free mass (FFM), which can lead to a decrease in basal metabolic rate and musculoskeletal function. Consequently, recent studies have highlighted the importance of FFM preservation in addition to the amount of weight loss as a clinically significant indicator. This study analyzed the effect of indole-3-pyruvic acid (IPyA) concentrations, measured in preoperative serum from 42 patients who underwent sleeve gastrectomy, on changes in FFM after surgery [41]. The patient group with relatively preserved FFM at 3–6 months showed significantly higher preoperative IPyA concentrations, and in ROC analysis, IPyA showed a predictive power of AUC 0.763 (P=0.006) for predicting FFM gain.
IPyA is a tryptophan-derived metabolite produced by gut microbiota and muscle stem cells, and it activates the aryl hydrocarbon receptor (AhR) to regulate inflammation and contribute to muscle regeneration. In particular, this study suggested the possibility that IPyA can not only preserve FFM but also reduce the risk of postoperative weight regain. This is based on a physiological mechanism where inflammation suppression and atrophy inhibition functions act together through AhR signaling in muscles. Furthermore, the FFM preservation group also had a higher rate of weight loss, showing significant differences at both 6 and 12 months post-surgery. As such, IPyA can function as a precise biomarker reflecting muscle regeneration and energy metabolism regulation, beyond simple body composition changes, and suggests the possibility of therapeutic intervention through tryptophan-IPA metabolic pathway modulation in the future. This study is one of the first cases to emphasize the role of metabolites in the novel clinical goal of FFM preservation, expanding the potential of biological indicators reflecting preoperative metabolic status.
CONCLUSIONS
MBS induces various metabolic effects beyond simple weight loss, including remission of T2D, improvement of insulin resistance, and inflammation suppression. Metabolomics is gaining attention as a tool to understand the molecular basis of these effects and to predict patient-specific surgical responsiveness. In particular, amino acid metabolites such as BCAAs, AAAs, and tryptophan derivatives are closely associated with obesity and metabolic diseases, and their preoperative concentrations can be useful in predicting the extent of metabolic improvement after surgery. In the future, it is expected that personalized surgical strategies based on metabolomics will become possible through multi-omics integration analysis and machine learning-based predictive model development. Ultimately, such research can extend beyond predicting postoperative prognosis to the discovery of non-surgical alternative therapeutic targets.
Footnotes
Funding: This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government(MSIT) (RS-2025-00522643).
Conflict of Interest: None of the authors have any conflict of interest.
- Conceptualization:Lee HS, Kwon J, Kwon Y.
- Investigation:Lee HS, Kwon Y.
- Methodology:Lee HS, Kwon Y.
- Project administration:Kwon Y.
- Supervision:Kwon Y.
- Writing - original draft:Lee HS, Kwon Y.
- Writing - review & editing:Lee HS, Kwon Y.
References
- 1.Neinast M, Murashige D, Arany Z. Branched chain amino acids. Annu Rev Physiol. 2019;81:139–164. doi: 10.1146/annurev-physiol-020518-114455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Richardson NE, Konon EN, Schuster HS, Mitchell AT, Boyle C, Rodgers AC, et al. Lifelong restriction of dietary branched-chain amino acids has sex-specific benefits for frailty and lifespan in mice. Nat Aging. 2021;1:73–86. doi: 10.1038/s43587-020-00006-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Jang C, Oh SF, Wada S, Rowe GC, Liu L, Chan MC, et al. A branched-chain amino acid metabolite drives vascular fatty acid transport and causes insulin resistance. Nat Med. 2016;22:421–426. doi: 10.1038/nm.4057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Zhou M, Shao J, Wu CY, Shu L, Dong W, Liu Y, et al. Targeting BCAA catabolism to treat obesity-associated insulin resistance. Diabetes. 2019;68:1730–1746. doi: 10.2337/db18-0927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ericksen RE, Lim SL, McDonnell E, Shuen WH, Vadiveloo M, White PJ, et al. Loss of BCAA catabolism during carcinogenesis enhances mTORC1 activity and promotes tumor development and progression. Cell Metab. 2019;29:1151–1165.e6. doi: 10.1016/j.cmet.2018.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Sun EJ, Wankell M, Palamuthusingam P, McFarlane C, Hebbard L. Targeting the PI3K/Akt/mTOR pathway in hepatocellular carcinoma. Biomedicines. 2021;9:1639. doi: 10.3390/biomedicines9111639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Herman MA, She P, Peroni OD, Lynch CJ, Kahn BB. Adipose tissue branched chain amino acid (BCAA) metabolism modulates circulating BCAA levels. J Biol Chem. 2010;285:11348–11356. doi: 10.1074/jbc.M109.075184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Neinast MD, Jang C, Hui S, Murashige DS, Chu Q, Morscher RJ, et al. Quantitative analysis of the whole-body metabolic fate of branched-chain amino acids. Cell Metab. 2019;29:417–429.e4. doi: 10.1016/j.cmet.2018.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Bozadjieva Kramer N, Evers SS, Shin JH, Silverwood S, Wang Y, Burant CF, et al. The role of elevated branched-chain amino acids in the effects of vertical sleeve gastrectomy to reduce weight and improve glucose regulation. Cell Reports. 2020;33:108239. doi: 10.1016/j.celrep.2020.108239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Pakiet A, Wilczynski M, Rostkowska O, Korczynska J, Jabłonska P, Kaska L, et al. The effect of one anastomosis gastric bypass on branched-chain fatty acid and branched-chain amino acid metabolism in subjects with morbid obesity. Obes Surg. 2020;30:304–312. doi: 10.1007/s11695-019-04157-z. [DOI] [PubMed] [Google Scholar]
- 11.Kwon Y, Jang M, Lee Y, Ha J, Park S. Amino acid metabolites and slow weight loss in the early postoperative period after sleeve gastrectomy. J Clin Med. 2020;9:2348. doi: 10.3390/jcm9082348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Bohm MS, Joseph SC, Sipe LM, Kim M, Leathem CT, Mims TS, et al. The gut microbiome enhances breast cancer immunotherapy following bariatric surgery. JCI Insight. 2025;10:e187683. doi: 10.1172/jci.insight.187683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Dejong CH, van de Poll MC, Soeters PB, Jalan R, Olde Damink SW. Aromatic amino acid metabolism during liver failure. J Nutr. 2007;137:1579S–1585S. doi: 10.1093/jn/137.6.1579S. [DOI] [PubMed] [Google Scholar]
- 14.Wang TJ, Larson MG, Vasan RS, Cheng S, Rhee EP, McCabe E, et al. Metabolite profiles and the risk of developing diabetes. Nat Med. 2011;17:448–453. doi: 10.1038/nm.2307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Würtz P, Soininen P, Kangas AJ, Rönnemaa T, Lehtimäki T, Kähönen M, et al. Branched-chain and aromatic amino acids are predictors of insulin resistance in young adults. Diabetes Care. 2013;36:648–655. doi: 10.2337/dc12-0895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Wyse ATS, Dos Santos TM, Seminotti B, Leipnitz G. Insights from animal models on the pathophysiology of hyperphenylalaninemia: role of mitochondrial dysfunction, oxidative stress and inflammation. Mol Neurobiol. 2021;58:2897–2909. doi: 10.1007/s12035-021-02304-1. [DOI] [PubMed] [Google Scholar]
- 17.Zhou Q, Sun WW, Chen JC, Zhang HL, Liu J, Lin Y, et al. Phenylalanine impairs insulin signaling and inhibits glucose uptake through modification of IRβ. Nat Commun. 2022;13:4291. doi: 10.1038/s41467-022-32000-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Tam SY, Elsworth JD, Bradberry CW, Roth RH. Mesocortical dopamine neurons: high basal firing frequency predicts tyrosine dependence of dopamine synthesis. J Neural Transm Gen Sect. 1990;81:97–110. doi: 10.1007/BF01245830. [DOI] [PubMed] [Google Scholar]
- 19.Bromberg-Martin ES, Matsumoto M, Hikosaka O. Dopamine in motivational control: rewarding, aversive, and alerting. Neuron. 2010;68:815–834. doi: 10.1016/j.neuron.2010.11.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Kwon Y, Yoon H, Ha J, Lee HS, Pahk K, Kwon H, et al. Changes in pancreatic levodopa uptake in patients with obesity and new-onset type 2 diabetes: an 18F-FDOPA PET-CT study. Front Endocrinol (Lausanne) 2025;16:1460253. doi: 10.3389/fendo.2025.1460253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Chesler K, Motz C, Vo H, Douglass A, Allen RS, Feola AJ, et al. Initiation of L-DOPA treatment after detection of diabetes-induced retinal dysfunction reverses retinopathy and provides neuroprotection in rats. Transl Vis Sci Technol. 2021;10:8. doi: 10.1167/tvst.10.4.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Chaudhry S, Bernardes M, Harris PE, Maffei A. Gastrointestinal dopamine as an anti-incretin and its possible role in bypass surgery as therapy for type 2 diabetes with associated obesity. Minerva Endocrinol. 2016;41:43–56. [PMC free article] [PubMed] [Google Scholar]
- 23.Yu L, Lu J, Du W. Tryptophan metabolism in digestive system tumors: unraveling the pathways and implications. Cell Commun Signal. 2024;22:174. doi: 10.1186/s12964-024-01552-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Correia AS, Vale N. Tryptophan metabolism in depression: a narrative review with a focus on serotonin and kynurenine pathways. Int J Mol Sci. 2022;23:8493. doi: 10.3390/ijms23158493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Hyland NP, Cavanaugh CR, Hornby PJ. Emerging effects of tryptophan pathway metabolites and intestinal microbiota on metabolism and intestinal function. Amino Acids. 2022;54:57–70. doi: 10.1007/s00726-022-03123-x. [DOI] [PubMed] [Google Scholar]
- 26.Engin AB, Engin A. Tryptophan metabolism in obesity: the indoleamine 2,3-dioxygenase-1 activity and therapeutic options. Adv Exp Med Biol. 2024;1460:629–655. doi: 10.1007/978-3-031-63657-8_21. [DOI] [PubMed] [Google Scholar]
- 27.Berg M, Polyzos KA, Agardh H, Baumgartner R, Forteza MJ, Kareinen I, et al. 3-Hydroxyanthralinic acid metabolism controls the hepatic SREBP/lipoprotein axis, inhibits inflammasome activation in macrophages, and decreases atherosclerosis in Ldlr-/- mice. Cardiovasc Res. 2020;116:1948–1957. doi: 10.1093/cvr/cvz258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Oxenkrug G. Insulin resistance and dysregulation of tryptophan-kynurenine and kynurenine-nicotinamide adenine dinucleotide metabolic pathways. Mol Neurobiol. 2013;48:294–301. doi: 10.1007/s12035-013-8497-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Boadle-Biber MC. Regulation of serotonin synthesis. Prog Biophys Mol Biol. 1993;60:1–15. doi: 10.1016/0079-6107(93)90009-9. [DOI] [PubMed] [Google Scholar]
- 30.Shong KE, Oh CM, Namkung J, Park S, Kim H. Serotonin regulates de novo lipogenesis in adipose tissues through serotonin receptor 2A. Endocrinol Metab (Seoul) 2020;35:470–479. doi: 10.3803/EnM.2020.35.2.470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Xue C, Li G, Zheng Q, Gu X, Shi Q, Su Y, et al. Tryptophan metabolism in health and disease. Cell Metab. 2023;35:1304–1326. doi: 10.1016/j.cmet.2023.06.004. [DOI] [PubMed] [Google Scholar]
- 32.Gao H, Sun M, Li A, Gu Q, Kang D, Feng Z, et al. Microbiota-derived IPA alleviates intestinal mucosal inflammation through upregulating Th1/Th17 cell apoptosis in inflammatory bowel disease. Gut Microbes. 2025;17:2467235. doi: 10.1080/19490976.2025.2467235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.de Mello VD, Paananen J, Lindström J, Lankinen MA, Shi L, Kuusisto J, et al. Indolepropionic acid and novel lipid metabolites are associated with a lower risk of type 2 diabetes in the Finnish Diabetes Prevention Study. Sci Rep. 2017;7:46337. doi: 10.1038/srep46337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Kwon Y, Jang M, Lee Y, Ha J, Park S. Metabolomic analysis of the improvements in insulin secretion and resistance after sleeve gastrectomy: implications of the novel biomarkers. Obes Surg. 2021;31:43–52. doi: 10.1007/s11695-020-04925-2. [DOI] [PubMed] [Google Scholar]
- 35.Krause D, Suh HS, Tarassishin L, Cui QL, Durafourt BA, Choi N, et al. The tryptophan metabolite 3-hydroxyanthranilic acid plays anti-inflammatory and neuroprotective roles during inflammation: role of hemeoxygenase-1. Am J Pathol. 2011;179:1360–1372. doi: 10.1016/j.ajpath.2011.05.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Chobot V, Hadacek F, Weckwerth W, Kubicova L. Iron chelation and redox chemistry of anthranilic acid and 3-hydroxyanthranilic acid: a comparison of two structurally related kynurenine pathway metabolites to obtain improved insights into their potential role in neurological disease development. J Organomet Chem. 2015;782:103–110. doi: 10.1016/j.jorganchem.2015.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Christensen MHE, Fadnes DJ, Røst TH, Pedersen ER, Andersen JR, Våge V, et al. Inflammatory markers, the tryptophan-kynurenine pathway, and vitamin B status after bariatric surgery. PLoS One. 2018;13:e0192169. doi: 10.1371/journal.pone.0192169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Abildgaard A, Elfving B, Hokland M, Wegener G, Lund S. The microbial metabolite indole-3-propionic acid improves glucose metabolism in rats, but does not affect behaviour. Arch Physiol Biochem. 2018;124:306–312. doi: 10.1080/13813455.2017.1398262. [DOI] [PubMed] [Google Scholar]
- 39.Tuomainen M, Lindström J, Lehtonen M, Auriola S, Pihlajamäki J, Peltonen M, et al. Associations of serum indolepropionic acid, a gut microbiota metabolite, with type 2 diabetes and low-grade inflammation in high-risk individuals. Nutr Diabetes. 2018;8:35. doi: 10.1038/s41387-018-0046-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Ha J, Jang M, Kwon Y, Park YS, Park DJ, Lee JH, et al. Metabolomic profiles predict diabetes remission after bariatric surgery. J Clin Med. 2020;9:3897. doi: 10.3390/jcm9123897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Seo E, Kwon Y, Park S. Association between indole-3-pyruvic acid and change in fat-free mass relative to weight loss in patients undergoing sleeve gastrectomy. Metabolites. 2024;14:444. doi: 10.3390/metabo14080444. [DOI] [PMC free article] [PubMed] [Google Scholar]
