1
|
Pan M, Qian C, Huo S, Wu Y, Zhao X, Ying Y, Wang B, Yang H, Yeerken A, Wang T, Fu M, Wang L, Wei Y, Zhao Y, Shao C, Wang H, Zhao C. Gut-derived lactic acid enhances tryptophan to 5-hydroxytryptamine in regulation of anxiety via Akkermansia muciniphila. Gut Microbes 2025; 17:2447834. [PMID: 39782002 PMCID: PMC11730363 DOI: 10.1080/19490976.2024.2447834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/28/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025] Open
Abstract
The gut microbiota plays a pivotal role in anxiety regulation through pathways involving neurotransmitter production, immune signaling, and metabolic interactions. Among these, gut-derived serotonin (5-hydroxytryptamine, 5-HT), synthesized from tryptophan metabolism, has been identified as a key mediator. However, it remains unclear whether specific microbial factors regulate tryptophan metabolism to influence 5-HT production and anxiety regulation. In this study, we analyzed 110 athletes undergoing closed training and found that fecal lactate levels were significantly associated with anxiety indicators. We observed a significant negative correlation between Akkermansia abundance and anxiety levels in athletes. Co-supplementation with lactate and Akkermansia muciniphila (A. muciniphila) modulated tryptophan metabolism by increasing key enzyme TPH1 and reducing IDO1, thus shifting metabolism from kynurenine (Kyn) to 5-HT. In addition, lactate enhanced the propionate production capacity of A. muciniphila, potentially contributing to anxiety reduction in mice. Taken together, these findings suggest that enteric lactate and A. muciniphila collaboratively restore the imbalance in tryptophan metabolism, leading to increased 5-HT activity and alleviating anxiety phenotypes. This study highlights the intricate interplay between gut metabolites and anxiety regulation, offering potential avenues for microbiota-targeted therapeutic strategies for anxiety.
Collapse
Affiliation(s)
- Miaomiao Pan
- MOE/NHC/CAMS Key Lab of Medical Molecular Virology, School of Basic Medical Sciences, & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chenglang Qian
- MOE/NHC/CAMS Key Lab of Medical Molecular Virology, School of Basic Medical Sciences, & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shaoye Huo
- Department of Clinical Nutrition, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Yuchen Wu
- Institute of Wound Prevention and Treatment, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | | | | | - Boyu Wang
- MOE/NHC/CAMS Key Lab of Medical Molecular Virology, School of Basic Medical Sciences, & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hao Yang
- MOE/NHC/CAMS Key Lab of Medical Molecular Virology, School of Basic Medical Sciences, & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Anaguli Yeerken
- MOE/NHC/CAMS Key Lab of Medical Molecular Virology, School of Basic Medical Sciences, & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tongyao Wang
- MOE/NHC/CAMS Key Lab of Medical Molecular Virology, School of Basic Medical Sciences, & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mengwei Fu
- MOE/NHC/CAMS Key Lab of Medical Molecular Virology, School of Basic Medical Sciences, & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lihong Wang
- Department of Clinical Nutrition, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Yuhuan Wei
- Department of Clinical Nutrition, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Yunhua Zhao
- Department of Clinical Nutrition, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Chunhai Shao
- Department of Clinical Nutrition, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
- Department of Clinical Nutrition, Huashan Hospital, Fudan University, Shanghai, China
| | - Huijing Wang
- Institute of Wound Prevention and Treatment, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Chao Zhao
- MOE/NHC/CAMS Key Lab of Medical Molecular Virology, School of Basic Medical Sciences, & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Holthuijsen DDB, van Roekel EH, Bours MJL, Ueland PM, Breukink SO, Janssen-Heijnen MLG, Konsten JL, Keulen ETP, McCann A, Brezina S, Gigic B, Kok DE, Ulrich CM, Weijenberg MP, Eussen SJPM. Modeling how iso-caloric macronutrient substitutions are longitudinally associated with plasma kynurenines in colorectal cancer survivors up to 12 months post-treatment. J Nutr Biochem 2025; 141:109910. [PMID: 40158742 DOI: 10.1016/j.jnutbio.2025.109910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 03/08/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
Dietary intake of several macronutrients is associated with plasma kynurenines after colorectal cancer (CRC), and kynurenines have been linked to health-related outcomes. It is unknown how macronutrient substitution affects plasma kynurenines, which may be relevant for developing guidelines to improve post-CRC quality of life through dietary changes. Using iso-caloric substitution models, we investigated how substituting one macronutrient with another is longitudinally associated with plasma tryptophan, kynurenines, and kynurenine ratios in CRC survivors. Measurements were performed at 6-weeks, 6-months, and 12-months post-treatment in 247 stage I-III CRC survivors. Macronutrient intake was measured by 7-d dietary records and plasma kynurenines by LC/MS-MS. For analysis, we applied linear mixed models with false discovery rate (FDR) to adjust for multiple testing. After FDR adjustment, substituting 100 kcal/d of total carbohydrates with 100 kcal/d of total protein was associated with higher plasma concentrations of kynurenic acid (KA), xanthurenic acid (XA), and a higher kynurenic acid-to-quinolinic acid (KA/QA) ratio. Substituting 100 kcal/d of total carbohydrates with 100 kcal/d of total fat was associated with higher tryptophan concentrations, higher KA/QA ratio, and a lower kynurenine-to-tryptophan ratio (KTR) and hydroxykynurenine ratio (HKr). Substituting 100 kcal/d of total fat with 100 kcal/d of total protein was associated with higher XA concentrations. Altogether, iso-caloric macronutrient substitutions, particularly substituting carbohydrates with protein or fat, were longitudinally associated with higher concentrations of potentially favourable kynurenines and ratios (i.e., KA, XA, and KA/QA ratio) and lower ratios with pro-inflammatory or neurotoxic properties (i.e., KTR and HKr) in CRC survivors up to 12-months post-treatment.
Collapse
Affiliation(s)
- Daniëlle D B Holthuijsen
- Department of Epidemiology, CARIM Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands; Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands.
| | - Eline H van Roekel
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Martijn J L Bours
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | | | - Stéphanie O Breukink
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands; Department of Surgery, Maastricht University Medical Centre+, Maastricht, The Netherlands; NUTRIM Institute of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Maryska L G Janssen-Heijnen
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands; Department of Clinical Epidemiology, VieCuri Medical Centre, Venlo, The Netherlands
| | - Joop L Konsten
- Department of Surgery, VieCuri Medical Centre, Venlo, The Netherlands
| | - Eric T P Keulen
- Department of Internal Medicine and Gastroenterology, Zuyderland Medical Centre Sittard-Geleen, Geleen, The Netherlands
| | | | - Stefanie Brezina
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Biljana Gigic
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Dieuwertje E Kok
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Cornelia M Ulrich
- Huntsman Cancer Institute and Department of Population Health Sciences, University of Utah, Salt Lake City, Utah, USA
| | - Matty P Weijenberg
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Simone J P M Eussen
- Department of Epidemiology, CARIM Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands; Department of Epidemiology, CAPHRI Care and Public Health Research Institute, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
3
|
Lawrence AB, Brown SM, Bradford BM, Mabbott NA, Bombail V, Rutherford KMD. Non-neuronal brain biology and its relevance to animal welfare. Neurosci Biobehav Rev 2025; 173:106136. [PMID: 40185375 DOI: 10.1016/j.neubiorev.2025.106136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Non-neuronal cells constitute a significant portion of brain tissue and are seen as having key roles in brain homeostasis and responses to challenges. This review illustrates how non-neuronal biology can bring new perspectives to animal welfare through understanding mechanisms that determine welfare outcomes and highlighting interventions to improve welfare. Most obvious in this respect is the largely unrecognised relevance of neuroinflammation to animal welfare which is increasingly found to have roles in determining how animals respond to challenges. We start by introducing non-neuronal cells and review their involvement in affective states and cognition often seen as core psychological elements of animal welfare. We find that the evidence for a causal involvement of glia in cognition is currently more advanced than the corresponding evidence for affective states. We propose that translational research on affective disorders could usefully apply welfare science derived approaches for assessing affective states. Using evidence from translational research, we illustrate the involvement of non-neuronal cells and neuroinflammatory processes as mechanisms modulating resilience to welfare challenges including disease, pain, and social stress. We review research on impoverished environments and environmental enrichment which suggests that environmental conditions which improve animal welfare also improve resilience to challenges through balancing pro- and anti-inflammatory non-neuronal processes. We speculate that non-neuronal biology has relevance to animal welfare beyond neuro-inflammation including facilitating positive affective states. We acknowledge the relevance of neuronal biology to animal welfare whilst proposing that non-neuronal biology provides additional and relevant insights to improve animals' lives.
Collapse
Affiliation(s)
- Alistair B Lawrence
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK; Scotland's Rural College (SRUC), Edinburgh EH9 3JG, UK.
| | - Sarah M Brown
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Barry M Bradford
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Neil A Mabbott
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | | | | |
Collapse
|
4
|
Huang F, Sun K, Zhou J, Bao J, Xie G, Lu K, Fan Y. Decoding tryptophan: Pioneering new frontiers in systemic lupus erythematosus. Autoimmun Rev 2025; 24:103809. [PMID: 40158642 DOI: 10.1016/j.autrev.2025.103809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/26/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease that affects multiple organ systems, with its pathogenesis intricately tied to genetic, environmental, and immune regulatory factors. In recent years, the aberration of tryptophan metabolism has emerged as a key player in the disease, particularly through the activation of the kynurenine pathway and its influence on immune regulation. This review delves into the critical pathways of tryptophan metabolism and its profound impact on the multi-system manifestations of SLE, including its connections to the nervous system, kidneys, skin, and other organs. Additionally, it examines how tryptophan metabolism modulates the function of various immune cell types. The review also explores potential therapeutic avenues targeting tryptophan metabolism, such as dietary interventions, probiotic modulation, IDO expression inhibition, and immunoadsorption techniques. While current research has underscored the pivotal role of tryptophan metabolism in the onset and progression of SLE, its full therapeutic potential remains to be fully elucidated. This review aims to provide a solid scientific foundation for therapeutic strategies based on modulating tryptophan metabolism in SLE, offering a comprehensive overview of both clinical and basic research in this rapidly evolving field.
Collapse
Affiliation(s)
- Fugang Huang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Ke Sun
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Jiawang Zhou
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Jie Bao
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Guanqun Xie
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, PR China.
| | - Keda Lu
- The Third Affiliated Hospital of Zhejiang Chinese Medical University (Zhongshan Hospital of Zhejiang Province), Hangzhou 310005, Zhejiang, China.
| | - Yongsheng Fan
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, PR China.
| |
Collapse
|
5
|
He X, Zhang J, Zhang Y, Li H, Chen Y, Zhang H, Pan J, Zhou Y, Zhang S, Cheng L. L-Kynurenine regulates immune response in ICIs-associated myocarditis via JAK/STAT pathway. Int Immunopharmacol 2025; 156:114676. [PMID: 40267722 DOI: 10.1016/j.intimp.2025.114676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/24/2025] [Accepted: 04/12/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Immune checkpoint inhibitor associated myocarditis (ICIAM) is a drug-induced myocarditis characterized by the infiltration of immune cells into cardiac. However, the mechanisms are unknown and effective drug therapies are lacking in clinical practice. This study aims to explore the relationship between plasma metabolites and the treatment of ICIAM. METHODS Human plasma metabolites were analyzed using untargeted metabolomics for characteristic metabolites. For in vivo experiments, Male Balb/c mice were divided into six groups: PBS, L-Kynurenine, cTNI+PD-1 inhibitor (ICIs), ICIs+L-Kynurenine, ICIs+RO8191, ICIs+RO8191+L-Kynurenine. On day 21 post-modeling, echocardiography, ELISA and histopathology were employed to evaluate the therapeutic effect of L-Kynurenine. Flow cytometry was used to determine the proportion of immune cells in the heart and spleen. Bulk-RNAseq was conducted to analyze differential genes, and q-PCR, immunofluorescence and western blot were performed for validation experiments. RESULTS Untargeted metabolomics verified that indoleamine 2,3 dioxygenase-1 (IDO1)-derived L-Kynurenine level was higher in the serum of ICIAM compared to non-ICIAM patients. Meanwhile, in vitro and in vivo experiments showed that L-Kynurenine exhibited a therapeutic ability in ICIAM by inhibiting the pro-inflammatory polarization of immune cells and the secretion of pro-inflammatory cytokines. Mechanistically, L-Kynurenine improved cardiac functions majorly by the inhibition of the JAK1/STAT3 signaling pathway. CONCLUSION L-Kynurenine exhibits significant therapeutic potential in ICIAM. The multi-roles of L-Kynurenine in regulating immune responses make it possible to be used as a targeted drug for ICIAM therapy.
Collapse
Affiliation(s)
- Xiaozhen He
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, China
| | - Jian Zhang
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, China
| | - Yerui Zhang
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, China
| | - Huishan Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yifan Chen
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, China
| | - Hui Zhang
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, China
| | - Jianan Pan
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, China
| | - Yan Zhou
- State Key Laboratory of Oncogenes and Related Genes Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shilong Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Leilei Cheng
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, China.
| |
Collapse
|
6
|
Li L, Haijun W, Tianyu C, Wenjing W, Zi W, Yibing C. Metabolomics and machine learning identify urine metabolic characteristics and potential biomarkers for severe Mycoplasma pneumoniae pneumonia. Sci Rep 2025; 15:17090. [PMID: 40379752 PMCID: PMC12084370 DOI: 10.1038/s41598-025-01895-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 05/08/2025] [Indexed: 05/19/2025] Open
Abstract
To study the differences in the urine metabolome between pediatric patients with severe Mycoplasma pneumoniae pneumonia (SMPP) and those with general Mycoplasma pneumoniae pneumonia (GMPP) via non-targeted metabolomics method, and potential biomarkers were explored through machine learning (ML) algorithms. The urine metabonomics data of 48 children with SMPP and 85 children with GMPP were collected via high performance liquid chromatography‒mass spectrometry (HPLC-MS/MS). The differential metabolites between the two groups were obtained via principal component analysis (PCA) and partial least squares-discriminant analysis (PLS-DA), and the significant metabolic pathways were screened via enrichment analysis. Potential biomarkers were identified using the random forest algorithm, and their relationships with clinical indicators were subsequently analyzed. A total of 136 significantly differential metabolites were identified in the urine samples from SMPP and GMPP. Of these, 68 metabolites were upregulated, and 68 were downregulated, predominantly belonging to the amino acid group. A total of 6 differential metabolic pathways were enriched, including Galactose metabolism, Pantothenate and CoA biosynthesis, Cysteine and methionine metabolism, Biotin metabolism, Glycine, serine and threonine metabolism, Arginine biosynthesis. Three significant potential biomarkers were identified through machine learning: 3-Hydroxyanthranilic acid (3-HAA), L-Kynurenine, and 16(R)-HETE. The area under the receiver operating characteristic curve (AUC) for this three-metabolite panel was 0.9142. There are great differences in the urine metabolome between SMPP and GMPP children, with multiple metabolic pathways being abnormally expressed. Three metabolites have been identified as potential biomarkers for the early detection of SMPP.
Collapse
Affiliation(s)
- Lin Li
- Department of Emergency, Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Wang Haijun
- Department of Emergency, Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Chen Tianyu
- JIUHE Diagnostics Co., Ltd, Zhengzhou, 450016, China
| | - Wang Wenjing
- JIUHE Diagnostics Co., Ltd, Zhengzhou, 450016, China
| | - Wang Zi
- Department of Emergency, Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Cheng Yibing
- Department of Emergency, Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China.
| |
Collapse
|
7
|
Xiong L, Huang YX, Mao L, Xu Y, Deng YQ. Targeting gut microbiota and its associated metabolites as a potential strategy for promoting would healing in diabetes. World J Diabetes 2025; 16:98788. [DOI: 10.4239/wjd.v16.i5.98788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 01/03/2025] [Accepted: 03/05/2025] [Indexed: 04/25/2025] Open
Abstract
Impaired healing of diabetic wounds is one of the most important complications of diabetes, often leading to lower limb amputations and incurring significant economic and psychosocial costs. Unfortunately, there are currently no effective prevention or treatment strategies available. Recent research has reported that an imbalance in the gut microbiota, known as dysbiosis, was linked to the onset of type 2 diabetes, as well as the development and progression of diabetic complications. Indeed, the gut microbiota has emerged as a promising therapeutic approach for treating type 2 diabetes and related diseases. However, there is few of literatures specifically discussing the relationship between gut microbiota and diabetic wounds. This review aims to explore the potential role of the gut microbiota, especially probiotics, and its associated byproducts such as short chain fatty acids, bile acids, hydrogen sulfide, and tryptophan metabolites on wound healing to provide fresh insights and novel perspectives for the treatment of chronic wounds in diabetes.
Collapse
Affiliation(s)
- Ling Xiong
- Department of Dermatology & STD, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Ya-Xin Huang
- Department of Dermatology & STD, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Lan Mao
- Department of Dermatology & STD, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Yong Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Yong-Qiong Deng
- Department of Dermatology & STD, Chengdu Integrated TCM & Western Medicine Hospital, Chengdu 610000, Sichuan Province, China
- Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| |
Collapse
|
8
|
Yi L, Wang K, Liufu S, Chen W, Chen B, Liu X, Liu C, Liu J, Xu X, Ma H. LC-MS/MS based metabolomics reveals the mechanism of skeletal muscle regeneration. BMC Musculoskelet Disord 2025; 26:457. [PMID: 40346552 PMCID: PMC12065226 DOI: 10.1186/s12891-025-08703-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/25/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Skeletal muscle possesses a robust regenerative capacity and can effectively repair itself following injury. However, research on the metabolic changes during skeletal muscle regeneration in large animals remains relatively limited. Therefore, in this study, we used pigs as a model and applied non-targeted LC-MS/MS metabolomic technology to reveal the metabolic changes during skeletal muscle regeneration, and conducted an in-depth exploration of important signaling pathways, which can provide a reference for further research on the mechanisms promoting skeletal muscle regeneration. METHODS In this study, we used 18 piglets aged 35 days and weighing 7.10 ± 0.90 kg to construct a skeletal muscle regeneration model. These piglets were randomly divided into three treatment groups (n = 6) and injected with cardiotoxins (CTX) in the right longissimus dorsi muscle. They were euthanized on the 1st, 4th, and 16th days post-injection to collect right longissimus dorsi muscle samples as the treatment group. Additionally, the left longissimus dorsi muscle of piglets on the 4th day post-injection was selected as the control group. Phenotypic changes in skeletal muscle regeneration were determined through H&E staining, immunofluorescence, and Western Blot analysis, and LC-MS/MS untargeted metabolomics technology was utilized to explore the differential expressed metabolites (DEMs) involved in skeletal muscle regeneration. RESULTS Phenotyping results showed that the regeneration model showed 3 stages of inflammation, regeneration and remodeling, which indicated successful model construction. Non-targeted LC-MS/MS metabolomics analysis showed significant differences in the structure of metabolites in these 3 stages. (1) In the inflammatory stage, a total of 198 DEMs were identified, which were mainly enriched in the pathways regulating the inflammatory response. (2) in the repair stage, 264 DEMs were identified, which were mainly enriched in pathways that inhibit inflammatory response and promote protein synthesis. (3) During the remodeling stage, 102 DEMs were identified, which were mainly enriched in the pathways that inhibit protein depletion and promote protein deposition. Temporal expression analysis revealed metabolites consistent with changes in the skeletal muscle regeneration process and found that these metabolite functions were mainly enriched in inhibiting inflammatory responses, alleviating myofibrillar lysis, and promoting muscle growth. Among them, (R)-Lipoic acid, 8-Hydroxyguanosine, and Uridine 5'-monophosphate maybe key metabolites associated with skeletal muscle regeneration. CONCLUSION The skeletal muscle regeneration mechanism was systematically explored, and the metabolite time series analysis during skeletal muscle regeneration revealed some key metabolites that reflect the degree of skeletal muscle damage.
Collapse
Affiliation(s)
- Lei Yi
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, PR, 410128, China
| | - Kaiming Wang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, PR, 410128, China
| | - Sui Liufu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, PR, 410128, China
| | - Wenwu Chen
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, PR, 410128, China
| | - Bohe Chen
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, PR, 410128, China
| | - Xiaolin Liu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, PR, 410128, China
| | - Caihong Liu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, PR, 410128, China
| | - Jingwen Liu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, PR, 410128, China
| | - Xin Xu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, PR, 410128, China
| | - Haiming Ma
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, PR, 410128, China.
- Key Laboratory of Livestock and Poultry Resources (Pig) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Changsha, PR, 410128, China.
- Yuelushan Laboratory, Changsha, PR, 410128, China.
| |
Collapse
|
9
|
Lee MH, Menezes TCF, Reisz JA, Cendali FI, Ferreira EVM, Ota-Arakaki JS, Sperandio PA, Kumar R, Mickael C, Ieong MM, Santos JL, Duarte ACB, Fonseca Balladares DC, Nolan K, Tuder RM, Hassoun PM, D’Alessandro A, Oliveira RKF, Graham BB. Physiologic relevance of the transpulmonary metabolome in connective tissue disease-associated pulmonary vascular disease. JCI Insight 2025; 10:e187911. [PMID: 40337861 PMCID: PMC12070491 DOI: 10.1172/jci.insight.187911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 03/19/2025] [Indexed: 05/09/2025] Open
Abstract
Pathologic implications of dysregulated pulmonary vascular metabolism to pulmonary arterial hypertension (PAH) are increasingly recognized, but their clinical applications have been limited. We hypothesized that metabolite quantification across the pulmonary vascular bed in connective tissue disease-associated (CTD-associated) PAH would identify transpulmonary gradients of pathobiologically relevant metabolites, in an exercise stage-specific manner. Sixty-three CTD patients with established or suspected PAH underwent exercise right heart catheterization. Using mass spectrometry-based metabolomics, metabolites were quantified in plasma samples simultaneously collected from the pulmonary and radial arteries at baseline and during resistance-free wheeling, peak exercise, and recovery. We identified uptake and excretion of metabolites across the pulmonary vascular bed, unique and distinct from single vascular site analysis. We demonstrated the physiological relevance of metabolites previously shown to promote disease in animal models and end-stage human lung tissues, including acylcarnitines, glycolytic intermediates, and tryptophan catabolites. Notably, pulmonary vascular metabolite handling was exercise stage specific. Transpulmonary metabolite gradients correlated with hemodynamic endpoints largely during free-wheeling. Glycolytic intermediates demonstrated physiologic significance at peak exercise, including net uptake of lactate in those with more advanced disease. Contribution of pulmonary vascular metabolism to CTD-PAH pathogenesis and therapeutic candidacy of metabolism modulation must be considered in the context of physiologic stress.
Collapse
Affiliation(s)
- Michael H. Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, UCSF, San Francisco, California, USA
- Lung Biology Center, Department of Medicine, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Thaís C. F. Menezes
- Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Hospital São Paulo, São Paulo, Brazil
| | | | | | - Eloara V. M. Ferreira
- Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Hospital São Paulo, São Paulo, Brazil
| | - Jaquelina S. Ota-Arakaki
- Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Hospital São Paulo, São Paulo, Brazil
| | - Priscila A. Sperandio
- Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Hospital São Paulo, São Paulo, Brazil
| | - Rahul Kumar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, UCSF, San Francisco, California, USA
- Lung Biology Center, Department of Medicine, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Claudia Mickael
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Martin M. Ieong
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, UCSF, San Francisco, California, USA
- Lung Biology Center, Department of Medicine, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Juliana Lucena Santos
- Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Hospital São Paulo, São Paulo, Brazil
| | - Ana Carolina B. Duarte
- Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Hospital São Paulo, São Paulo, Brazil
| | - Dara C. Fonseca Balladares
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, UCSF, San Francisco, California, USA
- Lung Biology Center, Department of Medicine, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Kevin Nolan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, UCSF, San Francisco, California, USA
- Lung Biology Center, Department of Medicine, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Rubin M. Tuder
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Paul M. Hassoun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Rudolf K. F. Oliveira
- Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo (UNIFESP), Hospital São Paulo, São Paulo, Brazil
| | - Brian B. Graham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, UCSF, San Francisco, California, USA
- Lung Biology Center, Department of Medicine, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| |
Collapse
|
10
|
Bakker L, Ramakers IHGB, van Greevenbroek MMJ, Backes WH, Jansen JFA, Schram MT, van der Kallen CJH, Schalkwijk CG, Wesselius A, Ulvik A, Ueland PM, Verhey FRJ, Eussen SJPM, Köhler S. The kynurenine pathway and markers of neurodegeneration and cerebral small vessel disease: The Maastricht Study. J Neurol Sci 2025; 474:123522. [PMID: 40367835 DOI: 10.1016/j.jns.2025.123522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 04/24/2025] [Accepted: 04/29/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND The kynurenine pathway, the main metabolic pathway of tryptophan degradation, has been mostly studied in neurodegenerative disorders, while its role in cerebrovascular pathology is less clear. We investigated whether kynurenines are associated with markers of neurodegeneration and cerebrovascular pathology in the general population. METHODS Cross-sectional data was used from 1589 individuals (60.0 ± 8.0 years, 54.3 % men) who participated in The Maastricht Study, an observational population-based cohort study. Plasma concentrations of tryptophan, kynurenines, and neopterin were quantified. Neurodegeneration was measured by volumes of intracranial cerebrospinal fluid (CSF), while cerebrovascular pathology was measured by white matter hyperintensity (WMH) volume and presence of cerebral small vessel disease (cSVD), defined as the presence of lacunar infarcts, cerebral microbleeds or a Fazekas score ≥ 2, all derived from 3 T MRI. Associations of kynurenines with these markers were investigated using linear, logistic, and restricted cubic spline regression models adjusted for confounders. RESULTS Fully adjusted analyses indicated that higher levels of 3-hydroxyanthranilic acid, kynurenine, kynurenic acid, quinolinic acid, and neopterin were associated with lower CSF volume. For the latter four, associations were non-linear and restricted to participants with already below average concentrations. Higher levels of tryptophan and anthranilic acid were associated with higher CSF volumes in participants with above-average levels. Higher levels of the kynurenine-tryptophan ratio were associated with a lower WMH volume. No evidence was found for associations between individual kynurenines and WMH volume or cSVD presence. CONCLUSIONS These findings suggest that several kynurenines are associated with neurodegeneration in community-dwelling older adults, but not with cerebrovascular damage.
Collapse
Affiliation(s)
- Lieke Bakker
- Mental Health and Neuroscience Research Institute (MHeNs), Maastricht University, 6229 ER Maastricht, the Netherlands; Department of Psychiatry and Neuropsychology, Alzheimer Center Limburg, Maastricht University, 6229 ER Maastricht, the Netherlands.
| | - Inez H G B Ramakers
- Mental Health and Neuroscience Research Institute (MHeNs), Maastricht University, 6229 ER Maastricht, the Netherlands; Department of Psychiatry and Neuropsychology, Alzheimer Center Limburg, Maastricht University, 6229 ER Maastricht, the Netherlands.
| | - Marleen M J van Greevenbroek
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, the Netherlands; Department of Internal Medicine, Maastricht University Medical Center, 6229 HX Maastricht, the Netherlands.
| | - Walter H Backes
- Mental Health and Neuroscience Research Institute (MHeNs), Maastricht University, 6229 ER Maastricht, the Netherlands; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6229 HX Maastricht, the Netherlands.
| | - Jacobus F A Jansen
- Mental Health and Neuroscience Research Institute (MHeNs), Maastricht University, 6229 ER Maastricht, the Netherlands; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6229 HX Maastricht, the Netherlands; Department of Electrical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, the Netherlands.
| | - Miranda T Schram
- Mental Health and Neuroscience Research Institute (MHeNs), Maastricht University, 6229 ER Maastricht, the Netherlands; Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, the Netherlands; Department of Internal Medicine, Maastricht University Medical Center, 6229 HX Maastricht, the Netherlands; Heart and Vascular Center, Maastricht University Medical Center, 6229 HX Maastricht, the Netherlands.
| | - Carla J H van der Kallen
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, the Netherlands; Department of Internal Medicine, Maastricht University Medical Center, 6229 HX Maastricht, the Netherlands.
| | - Casper G Schalkwijk
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, the Netherlands; Department of Internal Medicine, Maastricht University Medical Center, 6229 HX Maastricht, the Netherlands.
| | - Anke Wesselius
- Department of Epidemiology, Maastricht University, 6229 HA Maastricht, the Netherlands; Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6229 ER Maastricht, the Netherlands.
| | | | | | - Frans R J Verhey
- Mental Health and Neuroscience Research Institute (MHeNs), Maastricht University, 6229 ER Maastricht, the Netherlands; Department of Psychiatry and Neuropsychology, Alzheimer Center Limburg, Maastricht University, 6229 ER Maastricht, the Netherlands.
| | - Simone J P M Eussen
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, the Netherlands; Department of Epidemiology, Maastricht University, 6229 HA Maastricht, the Netherlands; Care and Public Health Research Institute (CAPHRI), Maastricht University, 6229 ER Maastricht, the Netherlands.
| | - Sebastian Köhler
- Mental Health and Neuroscience Research Institute (MHeNs), Maastricht University, 6229 ER Maastricht, the Netherlands; Department of Psychiatry and Neuropsychology, Alzheimer Center Limburg, Maastricht University, 6229 ER Maastricht, the Netherlands.
| |
Collapse
|
11
|
Hu W, Garrison C, Prasad R, Boulton M, Grant M. Indole metabolism and its role in diabetic macrovascular and microvascular complications. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2025; 53:100532. [PMID: 40230659 PMCID: PMC11995707 DOI: 10.1016/j.ahjo.2025.100532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/03/2025] [Accepted: 03/21/2025] [Indexed: 04/16/2025]
Abstract
Tryptophan (Trp), an essential amino acid obtained through dietary sources, plays a crucial role in various physiological processes. The metabolism of Trp branches into three principal pathways: the serotonin pathway, the kynurenine pathway, and the indole pathway. The kynurenine and serotonin pathways are host pathways while the indole pathway is solely the result of bacterial metabolism. Trp metabolites extend their influence beyond protein biosynthesis to affect a spectrum of pathophysiological mechanisms including, but not limited to, neuronal function, immune modulation, inflammatory responses, oxidative stress regulation, and maintenance of intestinal health. This review focuses on indole derivatives and their impact on vascular health. Trp-containing dipeptides are highlighted as a targeted nutraceutical approach to modulate Trp metabolism, enhance beneficial metabolite production, and mitigate risk factors for vascular diseases. The importance of optimizing Trp intake and dietary strategies to harness the benefits of Trp-derived metabolites for vascular health is underscored, bringing to light the need for further research to refine these therapeutic approaches.
Collapse
Affiliation(s)
- W. Hu
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Food Science and Technology, National University of Singapore, Singapore
| | - C. Garrison
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - R. Prasad
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - M.E. Boulton
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - M.B. Grant
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
12
|
Hao C, Meng D, Shi W, Xu C, Wang Q, Kuang H. Chiral Gold Nanostructure Monolayers as SERS Substrates for Ultrasensitive Detection of Enantiomer Biomarkers of Alzheimer's Disease. Angew Chem Int Ed Engl 2025; 64:e202502115. [PMID: 40062420 DOI: 10.1002/anie.202502115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/09/2025] [Accepted: 03/09/2025] [Indexed: 03/19/2025]
Abstract
The early diagnosis of neurodegenerative diseases, such as Alzheimer's disease (AD), requires the identification of sensitive and specific biomarkers. Detecting chiral molecules at concentrations relevant to disease states remains challenging. Herein, a new type of chiral gold nanostructure induced by D-/L-cysteine-leucine dipeptides with a g-factor of 0.1 was successfully synthesized for enantiomer biomarker detection. To enhance the discrimination performance, the chiral gold nanostructures were assembled into D-/L-Au monolayers. As surface-enhanced Raman scattering (SERS) substrates, the D-/L-Au monolayers simultaneously deliver molecular structural specificity and enantioselectivity within a single spectrum, which can be a versatile, label-free chiral discrimination strategy for the detection of D-/L-kynurenine (Kyn). The mechanism was unveiled to involve high enantioselective adsorption energies between L- and D-Kyn on the lattice plane (221), resulting in enantioselective sensing. The results showed that the L-Au monolayer reached a limit of detection (LOD) of 3.7 nm for L-Kyn, while the D-Au monolayer reached an LOD of 3.6 nM for D-Kyn. Notably, there was a significant difference in D-Kyn levels between AD patients and healthy individuals in serum samples, a distinction not observed for L-Kyn, which positioned D-Kyn as a potential novel biomarker for clinical prediagnosis of AD patients, marking the first report of its kind worldwide. This study provides a robust tool for advancing biomedical science and clinical diagnostics.
Collapse
Affiliation(s)
- Changlong Hao
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P.R. China
| | - Dan Meng
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P.R. China
| | - Wenxiong Shi
- Institute for New Energy Materials and Low Carbon Technologies, School of Materials Science and Engineering, Tianjin University of Technology, Tianjin, 300384, P.R. China
| | - Chuanlai Xu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P.R. China
| | - Qing Wang
- Department of Neurosurgery, Wuxi Neurosurgical Institute, Jiangnan University, Wuxi, Jiangsu, 214122, P.R. China
| | - Hua Kuang
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P.R. China
| |
Collapse
|
13
|
Gao A, Qi Y, Luo Y, Hu X, Jiang R, Chang S, Zhou X, Liu L, Zhu L, Feng X, Jiang L, Zhong H. Mass spectrometric monitoring of redox transformation and arylation of tryptophan. Anal Chim Acta 2025; 1349:343822. [PMID: 40074454 DOI: 10.1016/j.aca.2025.343822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/27/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025]
Abstract
Tryptophan (Trp) is an essential amino acid obtained from human diet. It is involved not only in de novo biosynthesis of proteins but also in complex metabolic pathways. Redox transformation of tryptophan is under-explored in comparison with kynurenine, serotonin and indole pyruvate pathways. We described herein a mass spectrometric approach that can not only detect electron transfer-associated changes in masses and charges, but also identify electron-directed bond cleavages and radical-radical cross-coupling reactions in redox transformation of tryptophan. Photoactive TiO2 that is widely applied in cosmetic products is used as electron donor and receptor because of the capability to generate photoelectrons and holes. It was demonstrated tryptophan undergoes redox transformation through the removal of an electron from amino nitrogen atom by hole oxidization along with an electron capture in the indole ring. The back and forth electron-shuttle converts electric energy into chemical energy that enforces bond cleavages. Sodium-coupled electron transfer (SCET) was found in complementary with proton-coupled electron transfer in tryptophan. The movement of sodium ions avoids electric charge buildup caused by electron transfer. Various redox products were detected on both light irradiated TiO2 and skins, among which β-carboline shows extensive radical scavenging ability for diverse cross-coupling with indole derivatives. Light-independent redox products have been detected in vivo such as in mouse brain, indicating the presence of in vivo electron transfer-directed redox transformation. It has also been revealed that tryptophan can be arylated on Cα and Cβ atoms in response to the exposure of halogenated aromatics.
Collapse
Affiliation(s)
- Anji Gao
- State Key Laboratory of Magnetic Resonance Spectroscopy and Imaging, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Yinghua Qi
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, PR China
| | - Yixiang Luo
- Medical College of Guangxi University, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Xiaoyuan Hu
- Medical College of Guangxi University, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Ruowei Jiang
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, PR China
| | - Shao Chang
- College of Life Science and Technology, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Xin Zhou
- Medical College of Guangxi University, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Linhui Liu
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, PR China
| | - Luping Zhu
- College of Life Science and Technology, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Xue Feng
- Center for Instrumental Analysis, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Ling Jiang
- State Key Laboratory of Magnetic Resonance Spectroscopy and Imaging, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Hongying Zhong
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, Guangxi University, Nanning, Guangxi, 530004, PR China; College of Life Science and Technology, Guangxi University, Nanning, Guangxi, 530004, PR China; Medical College of Guangxi University, Guangxi University, Nanning, Guangxi, 530004, PR China; Center for Instrumental Analysis, Guangxi University, Nanning, Guangxi, 530004, PR China.
| |
Collapse
|
14
|
Ogger PP, Murray PJ. Dissecting inflammation in the immunemetabolomic era. Cell Mol Life Sci 2025; 82:182. [PMID: 40293552 PMCID: PMC12037969 DOI: 10.1007/s00018-025-05715-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 04/30/2025]
Abstract
The role of immune metabolism, specific metabolites and cell-intrinsic and -extrinsic metabolic states across the time course of an inflammatory response are emerging knowledge. Targeted and untargeted metabolomic analysis is essential to understand how immune cells adapt their metabolic program throughout an immune response. In addition, metabolomic analysis can aid to identify pathophysiological patterns in inflammatory disease. Here, we discuss new metabolomic findings within the transition from inflammation to resolution, focusing on three key programs of immunity: Efferocytosis, IL-10 signaling and trained immunity. Particularly the tryptophan-derived metabolite kynurenine was identified as essential for efferocytosis and inflammation resolution as well as a potential biomarker in diverse inflammatory conditions. In summary, metabolomic analysis and integration with transcriptomic and proteomic data, high resolution imaging and spatial information is key to unravel metabolic drivers and dependencies during inflammation and progression to tissue-repair.
Collapse
Affiliation(s)
- Patricia P Ogger
- Immunoregulation Research Group, Max Planck Institute of Biochemistry, Martinsried, 82152, Germany
| | - Peter J Murray
- Immunoregulation Research Group, Max Planck Institute of Biochemistry, Martinsried, 82152, Germany.
| |
Collapse
|
15
|
Zhu H, Zheng S, Xie L, Yun Y, Kwan P, Rollo B, Huang H. Identification and enrichment of potential pathways in the buffy coat of patients with DRE using non-targeted metabolomics integrated with GEO Datasets. Eur J Med Res 2025; 30:332. [PMID: 40287763 PMCID: PMC12032676 DOI: 10.1186/s40001-025-02609-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND This study aims to identify potential biomarkers in the buffy coat of drug-resistant epilepsy (DRE) patients with mesial temporal lobe epilepsy and to elucidate associated pathways. METHODS A comprehensive non-targeted metabolomic and Gene Expression Omnibus (GEO) datasets analysis was first performed on buffy coat from DRE patients and non-epilepsy (CON) patients. Potential enriched biomarkers and pathways were integrated with gene expression profiles from GEO datasets to identify robust biomarkers. RESULTS In the DRE group, there were 15 patients (10 males and 5 females), with an average age of (37.67 ± 15.53) years. In the CON group, there were 10 patients (7 males and 3 females), with an average age of (51.60 ± 18.20) years. A total of 27 potential biomarkers were identified, including 7 down-regulated and 8 up-regulated. Additionally, 9 potential pathways related to DRE were identified. Notably, purine metabolism, tryptophan metabolism and aminoacyl-tRNA metabolism were closely related to DRE. Purine metabolism was up-regulated, while aminoacyl-tRNA and tryptophan metabolism were down-regulated. CONCLUSIONS The integration of metabolomic data with GEO datasets analysis offers a new strategy to identify robust biomarkers and pathways. The findings obtained from the buffy coat analysis offer potential insights for the diagnosis and treatment of DRE.
Collapse
Affiliation(s)
- Hailin Zhu
- School of Pharmacy, Nanchang University, Nanchang, China
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Australia
| | - Suyue Zheng
- Department of Neurosurgery, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Liyuan Xie
- Department of Neurosurgery, the Second Affiliated Hospital of Nanchang University, 566 Xuefu Road, Nanchang, 330000, China
| | - Yi Yun
- Biobank Center, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Patrick Kwan
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Australia
| | - Ben Rollo
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Australia
| | - Hui Huang
- Department of Neurosurgery, the Second Affiliated Hospital of Nanchang University, 566 Xuefu Road, Nanchang, 330000, China.
| |
Collapse
|
16
|
Tang B, Ouyang H, Zheng S, Yu L, Xiao R, Wu L, Wang Z. Doxorubicin promotes NK cell dysfunction and induces acute liver injury through kynurenine-AhR axis. Int Immunopharmacol 2025; 153:114489. [PMID: 40112597 DOI: 10.1016/j.intimp.2025.114489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/04/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
Drug-induced liver injury (DILI) is one of the significant drug-induced diseases and a major cause of clinically unexplained liver injury and unexplained liver diseases. However, the mechanisms underlying doxorubicin (DOX)-induced DILI remain unclear. In this study, we constructed a mouse model of DOX-induced acute liver injury (ALI) and employed a combination of proteomics, metabolomics, and flow cytometry (FCM) to examine the roles of metabolic processes and innate immune responses. Our findings revealed that DOX treatment altered the metabolic profile and innate immune response signals in mouse livers. Specifically, DOX activated the indoleamine 2,3-dioxygenase 2 (IDO2)-mediated L-Tryptophan/L-Kynurenine metabolic pathway. Further in-depth analysis demonstrated that DOX promoted natural killer (NK) cell dysfunction leading to ALI by activating the kynurenine-aryl hydrocarbon receptor (Kyn-AhR) axis. Importantly, targeting the Kyn-AhR axis could reverse DOX-induced ALI. In summary, this study suggests that targeting the Kyn-AhR axis holds promise as an effective strategy to reverse ALI.
Collapse
Affiliation(s)
- Bohuai Tang
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Huan Ouyang
- Department of Cardiovascular, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350004, China
| | - Shuping Zheng
- Public Technology Service Center, Fujian Medical University, Fuzhou, 350122, China
| | - Le Yu
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Ruiying Xiao
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Linqing Wu
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China.
| | - Zengbin Wang
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China.
| |
Collapse
|
17
|
Jackson EF, Riley TB, Overton PG. Serotonin dysfunction in ADHD. J Neurodev Disord 2025; 17:20. [PMID: 40264019 PMCID: PMC12013068 DOI: 10.1186/s11689-025-09610-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 04/04/2025] [Indexed: 04/24/2025] Open
Abstract
It is well accepted that attention deficit hyperactivity disorder (ADHD) is in part driven by dysfunction in the monoaminergic neurotransmitter system, but both the extent of dysfunction and possible therapeutic avenues presented by serotonergic neurotransmission is frequently overlooked. As such, we present key evidence for dysfunction in serotonergic transmission, as seen from biochemical, genetic and pharmacological perspectives. An overall deficit in serotonin availability is a common theme throughout the literature, thus this review aims to explore possible dysfunctions in the serotonin synthesis pathway which result in this reduced bioavailability, and investigate whether such dysfunctions could be loci of change in ADHD. We have identified several steps in transmission, namely the conversion of tryptophan to 5-hydroxytryptophan and its use of cofactor tetrahydrobiopterin, which could present promising avenues for development of novel clinical interventions for ADHD.
Collapse
Affiliation(s)
- Eleanor F Jackson
- Department of Psychology, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
| | - Timothy B Riley
- Department of Psychology, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Paul G Overton
- Department of Psychology, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| |
Collapse
|
18
|
Hu H, Lu X, Wu M, Bai Z, Liu X. Effects of Environmental Pollutants on Tryptophan Metabolism. TOXICS 2025; 13:311. [PMID: 40278627 PMCID: PMC12031123 DOI: 10.3390/toxics13040311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025]
Abstract
Tryptophan (Trp) is an important essential amino acid that plays a variety of physiological functions in the human body, including being a precursor of neurotransmitter and participating in immune regulation. Currently, more and more studies show that some pollutants in the environment can affect the metabolism of Trp and consequently affect human health. The present paper offers a comprehensive overview of prior research investigating the impact of environmental pollutants, including inorganic and organic contaminants, microplastics, and nanoplastics on the nervous system, immune system, digestive system, and maternal-fetal pregnancy, revealing their detrimental effects on Trp metabolism and human well-being.
Collapse
Affiliation(s)
| | | | | | | | - Xiaoshan Liu
- School of Public Health, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China; (H.H.); (X.L.); (M.W.); (Z.B.)
| |
Collapse
|
19
|
Zhang L, Liang M, Li C, Li F, Xin J, Lang J, Li S, Zhang D. Tailoring MXene-Heterometal-Doped MOF Composites via One-Step Room-Temperature Synthesis for Enhanced Electrochemical Detection of L-Tryptophan. Anal Chem 2025; 97:7203-7211. [PMID: 40136286 DOI: 10.1021/acs.analchem.4c06642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
L-Tryptophan (l-Trp) is an essential amino acid that is critical to human health and biosynthetic processes. Hence, developing a simple and sensitive l-Trp sensor is of great importance. In this study, a Ce-doped MOF-199/Ti3C2TX MXene nanocomposite was developed to detect l-Trp. Density functional theory (DFT) calculation suggested that the doping of Ce can effectively enhance the electronic structure of MOF-199 and improve its adsorption capacity for l-Trp. Additionally, the integration of Ce-doped MOF-199 with Ti3C2TX MXene substantially improves the conductivity of MOFs, resulting in enhanced electrocatalytic activity of the nanocomposite material. The sensor exhibits a wide linear range (0.5-156.5 μM), low limit of detection (0.18 μM), and excellent selectivity. Further, the sensor possesses excellent reproducibility, repeatability, and long-term stability. It has been successfully used to detect l-Trp in milk with excellent recoveries (99.66-104.40%).
Collapse
Affiliation(s)
- Li Zhang
- Key Laboratory of Polymeric Composite Materials of Heilongjiang Province, College of Materials Science and Engineering, Qiqihar University, Qiqihar 161006, China
| | - Minjie Liang
- Key Laboratory of Polymeric Composite Materials of Heilongjiang Province, College of Materials Science and Engineering, Qiqihar University, Qiqihar 161006, China
| | - Chao Li
- Key Laboratory of Polymeric Composite Materials of Heilongjiang Province, College of Materials Science and Engineering, Qiqihar University, Qiqihar 161006, China
| | - Fengbo Li
- College of Chemistry and Chemical Engineering, Qiqihar University, Qiqihar 161006, China
| | - Jianjiao Xin
- College of Chemistry and Chemical Engineering, Qiqihar University, Qiqihar 161006, China
| | - Jiaxin Lang
- Key Laboratory of Polymeric Composite Materials of Heilongjiang Province, College of Materials Science and Engineering, Qiqihar University, Qiqihar 161006, China
| | - Shaobin Li
- Key Laboratory of Polymeric Composite Materials of Heilongjiang Province, College of Materials Science and Engineering, Qiqihar University, Qiqihar 161006, China
| | - Deqing Zhang
- Key Laboratory of Polymeric Composite Materials of Heilongjiang Province, College of Materials Science and Engineering, Qiqihar University, Qiqihar 161006, China
| |
Collapse
|
20
|
He Y, Hu H, Liang X, Liang J, Li F, Zhou X. Gut microbes-muscle axis in muscle function and meat quality. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2885-4. [PMID: 40220074 DOI: 10.1007/s11427-024-2885-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 02/12/2025] [Indexed: 04/14/2025]
Abstract
The concept of the gut microbes-muscle axis underscores the impact of intestinal microbiota on the muscular system, an area that is increasingly coming to light. However, current interpretations and applications of this concept remain underdeveloped. In this review, we concluded and discussed factors, such as short-chain fatty acids, amino acids, vitamins, bile acids, antibiotics, cytokines, hormones, and extracellular vesicles that mediate gut microbes-muscle crosstalk and influence the gut microbes-muscle axis. Additionally, we examined how the gut microbes-muscle axis affects muscle mass, muscle strength, muscle metabolism, as well as muscle oxidative and immune status. Furthermore, we reviewed the influence of the microbes-muscle axis on muscle fiber type transition, muscle fat deposition, and meat quality. These insights illuminate the potential mechanisms by which the gut microbes-muscle axis operates in humans and animals. Thus, this review provides a theoretical foundation for future research and offers practical guidance for its application in biomedical and livestock industries.
Collapse
Affiliation(s)
- Yiwen He
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Hong Hu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China
| | - Xuqing Liang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410128, China
| | - Jing Liang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fengna Li
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Xihong Zhou
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
- Hunan Provincial Key Laboratory of the Traditional Chinese Medicine Agricultural Biogenomics, Changsha Medical University, Changsha, 410219, China.
| |
Collapse
|
21
|
Chen D, Biney CN, Wang Q, Cai M, Cheng S, Chen W, Zhang J, Zhao J, Zhang Y, Zhang W. Purine Metabolism Pathway Influence on Running Capacity in Rats. Metabolites 2025; 15:241. [PMID: 40278370 PMCID: PMC12029474 DOI: 10.3390/metabo15040241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 03/28/2025] [Accepted: 03/29/2025] [Indexed: 04/26/2025] Open
Abstract
Background: The natural differences in running capacities among rats remain poorly understood, and the mechanisms driving these differences need further investigation. Methods: Twenty male Sprague-Dawley (SD) rats were selected. High and low running capacity rats were identified using Treadmill Exhaustion Tests. Peripheral blood was collected for serum isolation, followed by a metabolomics analysis using LC-MS/MS. Data were preprocessed, and a principal component analysis (PCA) and a partial least squares-discriminant analysis (PLS-DA) were applied to identify metabolic profile differences. Significant metabolites were screened, and a pathway enrichment analysis was conducted using the KEGG database to determine key metabolic pathways. Forty SD rats (equal male and female) were randomly divided into an inosine triphosphate (ITP) group (24.29 mg/kg.bw daily) and a control group. Running capacity was assessed after one week of continuous treatment. Results: Three independent measurements showed consistent differences in running capacity. A total of 519 differential metabolites were identified, with 255 up-regulated and 264 down-regulated. The KEGG pathway analysis revealed a significant enrichment of the Purine Metabolism pathway (ITP-ATP) in the high running capacity group (p < 0.05). The ITP-treated group exhibited a significantly higher running capacity than the controls (p < 0.05), confirming the efficacy of dietary ITP supplementation. Conclusions: The running capacity of rats is influenced by the ITP-ATP pathway, and exogenous ITP administration through dietary intervention significantly improves running ability.
Collapse
Affiliation(s)
- Dengbo Chen
- School of Public Health and Preventive Medicine, Wannan Medical College, Wuhu 241002, China; (D.C.); (C.N.B.); (M.C.); (S.C.); (W.C.); (J.Z.); (J.Z.); (Y.Z.)
| | - Christian Noble Biney
- School of Public Health and Preventive Medicine, Wannan Medical College, Wuhu 241002, China; (D.C.); (C.N.B.); (M.C.); (S.C.); (W.C.); (J.Z.); (J.Z.); (Y.Z.)
| | - Qian Wang
- School of Public Health, Shandong Second Medical University, Jinan 252422, China;
| | - Mingzheng Cai
- School of Public Health and Preventive Medicine, Wannan Medical College, Wuhu 241002, China; (D.C.); (C.N.B.); (M.C.); (S.C.); (W.C.); (J.Z.); (J.Z.); (Y.Z.)
| | - Shi Cheng
- School of Public Health and Preventive Medicine, Wannan Medical College, Wuhu 241002, China; (D.C.); (C.N.B.); (M.C.); (S.C.); (W.C.); (J.Z.); (J.Z.); (Y.Z.)
| | - Wentao Chen
- School of Public Health and Preventive Medicine, Wannan Medical College, Wuhu 241002, China; (D.C.); (C.N.B.); (M.C.); (S.C.); (W.C.); (J.Z.); (J.Z.); (Y.Z.)
| | - Jinrui Zhang
- School of Public Health and Preventive Medicine, Wannan Medical College, Wuhu 241002, China; (D.C.); (C.N.B.); (M.C.); (S.C.); (W.C.); (J.Z.); (J.Z.); (Y.Z.)
| | - Junran Zhao
- School of Public Health and Preventive Medicine, Wannan Medical College, Wuhu 241002, China; (D.C.); (C.N.B.); (M.C.); (S.C.); (W.C.); (J.Z.); (J.Z.); (Y.Z.)
| | - Yuhan Zhang
- School of Public Health and Preventive Medicine, Wannan Medical College, Wuhu 241002, China; (D.C.); (C.N.B.); (M.C.); (S.C.); (W.C.); (J.Z.); (J.Z.); (Y.Z.)
| | - Wenzhong Zhang
- School of Public Health and Preventive Medicine, Wannan Medical College, Wuhu 241002, China; (D.C.); (C.N.B.); (M.C.); (S.C.); (W.C.); (J.Z.); (J.Z.); (Y.Z.)
| |
Collapse
|
22
|
Milosavljevic S, Piroli MV, Sandago EJ, Piroli GG, Smith HH, Beggiato S, Frizzell N, Pocivavsek A. Parental kynurenine 3-monooxygenase genotype in mice directs sex-specific behavioral outcomes in offspring. Biol Sex Differ 2025; 16:22. [PMID: 40176166 PMCID: PMC11967062 DOI: 10.1186/s13293-025-00703-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/13/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Disruptions in brain development can impact behavioral traits and increase the risk of neurodevelopmental conditions such as autism spectrum disorder, attention-deficit/hyperactivity disorder (ADHD), schizophrenia, and bipolar disorder, often in sex-specific ways. Dysregulation of the kynurenine pathway (KP) of tryptophan metabolism has been implicated in cognitive and neurodevelopmental disorders. Increased brain kynurenic acid (KYNA), a neuroactive metabolite implicated in cognition and sleep homeostasis, and variations in the Kmo gene, which encodes kynurenine 3-monooxygenase (KMO), have been identified in these patients. We hypothesize that parental Kmo genetics influence KP biochemistry, sleep behavior and brain energy demands, contributing to impairments in cognition and sleep in offspring through the influence of parental genotype and genetic nurture mechanisms. METHODS A mouse model of partial Kmo deficiency, Kmo heterozygous (HET-Kmo+/-), was used to examine brain KMO activity, KYNA levels, and sleep behavior in HET-Kmo+/- compared to wild-type control (WT-Control) mice. Brain mitochondrial respiration was assessed, and KP metabolites and corticosterone levels were measured in breast milk. Behavioral assessments were conducted on wild-type offspring from two parental groups: (i) WT-Control from WT-Control parents, (ii) wild-type Kmo (WT-Kmo+/+) from Kmo heterozygous parents (HET-Kmo+/-). All mice were C57Bl/6J background strain. Adult female and male offspring underwent behavioral testing for learning, memory, anxiety-like behavior and sleep-wake patterns. RESULTS HET-Kmo+/- mice exhibited reduced brain KMO activity, increased KYNA levels, and disrupted sleep architecture and electroencephalogram (EEG) power spectra. Mitochondrial respiration (Complex I and Complex II activity) and electron transport chain protein levels were impaired in the hippocampus of HET-Kmo+/- females. Breast milk from HET-Kmo+/- mothers increased kynurenine exposure during lactation but corticosterone levels were unchanged. Compared to WT-Control offspring, WT-Kmo+/+ females showed impaired spatial learning, heightened anxiety, reduced sleep and abnormal EEG spectral power. WT-Kmo+/+ males had deficits in reversal learning but no sleep disturbances or anxiety-like behaviors. CONCLUSIONS These findings suggest that Kmo deficiency impacts KP biochemistry, sleep behavior, and brain mitochondrial function. Even though WT-Kmo+/+ inherit identical genetic material as WT-Control, their development might be shaped by the parent's physiology, behavior, or metabolic state influenced by their Kmo genotype, leading to phenotypic sex-specific differences in offspring.
Collapse
Affiliation(s)
- Snezana Milosavljevic
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Building 1, D26, 6311 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Maria V Piroli
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Building 1, D26, 6311 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Emma J Sandago
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Building 1, D26, 6311 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Gerardo G Piroli
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Building 1, D26, 6311 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Holland H Smith
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Building 1, D26, 6311 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Sarah Beggiato
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Norma Frizzell
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Building 1, D26, 6311 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Ana Pocivavsek
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Building 1, D26, 6311 Garners Ferry Rd, Columbia, SC, 29209, USA.
| |
Collapse
|
23
|
Chen W, Yang L, Lee VHF, Xu L, Ma L, Ye Z, Xu W, Zhao C, Zheng D, Kiang KMY, Sun S, Qu Y, Zha J, Pang D, Zhang Y, Liang Z, Lin W, Zhang J, Zhang J, Luo M, Xu Z, Li D, Liang X, Leung GKK, Helali AE, Che C, Feng-Ming (Spring) Kong. Indoleamine 2,3-dioxygenase 1-mediated immune suppressive status is positively associated with brain metastasis in patients with non-small cell lung cancer. JOURNAL OF THE NATIONAL CANCER CENTER 2025; 5:179-192. [PMID: 40265090 PMCID: PMC12010389 DOI: 10.1016/j.jncc.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 04/24/2025] Open
Abstract
Background Indoleamine 2,3-dioxygenase (IDO1) activity, measured by kynurenine/tryptophan (K:T) ratio, is known for its association with distant metastasis and overall survival (OS) in patients with non-small cell lung cancer (NSCLC). Here, we aimed to examine whether IDO1 activity is correlated with OS in NSCLC patients with brain metastasis (Bramet) and has negative effect on modulating the anti-tumor functions of immune cells. Methods This study was a part of a prospective clinical trial in circulating biomarkers. Blood or tissues from eligible participants were collected for measurement of kynurenine, tryptophan, immune cell subtype, scRNA-seq analysis, and untargeted metabolomics analysis. Results A total of 195 patients were enrolled. The median kynurenine to tryptophan (K:T) ratio was 0.18, with consistent values observed among patients with NSCLC Bramet and those without (0.18 and 0.11, respectively). Notably, student's t-test analysis revealed significantly higher kynurenine concentrations in stage IV patients compared to those in stage I (2.3 vs 1.7 µM, P < 0.001). In patients with Bramet, both kynurenine concentrations and K:T ratios were significantly elevated in comparison with those of extra-cerebral metastasis (2.7 vs 1.9 µM, P < 0.001; 0.12 vs 0.095, P = 0.028; respectively). Single-cell analysis further validated a high level of IDO1 expression in stage IV tumors or Bramet lesions, particularly in macrophages, regulated by chemokines such as CXCL11. Additionally, K:T ratios exhibited significant associations with Treg cell percentages and OS in patients with Bramet (P = 0.039). Treatment with kynurenine led to the upregulation of immune-suppressive molecules, including PD-1, in T cells. Finally, untargeted metabolomics analysis further identified that, apart from the IDO1 metabolic pathway, other metabolites, such as those involved in phospholipid pathways, were also implicated in Bramet. Conclusion IDO1 metabolites may play immune-suppressive roles in NSCLC patients with Bramet.
Collapse
Affiliation(s)
- Weiwei Chen
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Li Yang
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Victor Ho-fun Lee
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Liangliang Xu
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Lingyu Ma
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Zhenghao Ye
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Wanli Xu
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Caining Zhao
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Danyang Zheng
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Karrie Mei-Yee Kiang
- Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Stella Sun
- Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Yuan Qu
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Jiandong Zha
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Dazhi Pang
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yan Zhang
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Zhibing Liang
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Wenchu Lin
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Jinliang Zhang
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Jitian Zhang
- Department of Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Min Luo
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Zhiyuan Xu
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Ding Li
- Department of Pathology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Xiaoling Liang
- Department of Pathology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Gilberto Ka-Kit Leung
- Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Aya El Helali
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Chiming Che
- Department of Chemistry, State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong, China
| | - Feng-Ming (Spring) Kong
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
24
|
Babakhani K, Kucinskas AL, Ye X, Giles ED, Sun Y. Aging immunity: unraveling the complex nexus of diet, gut microbiome, and immune function. IMMUNOMETABOLISM (COBHAM, SURREY) 2025; 7:e00061. [PMID: 40352822 PMCID: PMC12063687 DOI: 10.1097/in9.0000000000000061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/28/2025] [Indexed: 05/14/2025]
Abstract
Aging is associated with immune senescence and gut dysbiosis, both of which are heavily influenced by the diet. In this review, we summarize current knowledge regarding the impact of diets high in fiber, protein, or fat, as well as different dietary components (tryptophan, omega-3 fatty acids, and galacto-oligosaccharides) on the immune system and the gut microbiome in aging. Additionally, this review discusses how aging alters tryptophan metabolism, contributing to changes in immune function and the gut microbiome. Understanding the relationship between diet, the gut microbiome, and immune function in the context of aging is critical to formulate sound dietary recommendations for older individuals, and these personalized nutritional practices will ultimately improve the health and longevity of the elderly.
Collapse
Affiliation(s)
| | - Amanda L. Kucinskas
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Xiangcang Ye
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Erin D. Giles
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| |
Collapse
|
25
|
Zhu H, Huang C, Luo Z, Wu L, Cheng X, Wu H. Porphyromonas gingivalis Induces Disturbance of Kynurenine Metabolism Through the Gut-Brain Axis: Implications for Alzheimer's Disease. J Dent Res 2025; 104:439-448. [PMID: 39905278 DOI: 10.1177/00220345241303141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025] Open
Abstract
Porphyromonas gingivalis is one of the major pathogens of chronic periodontitis. P. gingivalis can cause systemic inflammation, amyloid β protein deposition, and hyperphosphorylation of tau protein, leading to Alzheimer's disease (AD)-like lesions. P. gingivalis oral infection causes gut microbiota alteration, gut barrier dysfunction, and intestinal immune response and inflammation. The microbiota-gut-brain axis has a potential role in the pathogenesis of AD. Whether P. gingivalis affects AD-like lesions via the gut-brain axis needs more study. In this study, orally administered P. gingivalis induced alveolar resorption, intestinal barrier impairment, and AD-like lesions. Oral infection with P. gingivalis induced oral and gut microflora dysbiosis, imbalance of the tryptophan metabolism pathway of gut microbiota, and elevated levels of 3-hydroxykynurenine in the sera and hippocampi. The key metabolite, 3-hydroxykynurenine, suppressed Bcl2 gene expression, leading to neuronal apoptosis and promoting AD-like lesions in vivo and in vitro. These findings suggest that P. gingivalis can induce AD pathogenesis through the gut-brain axis, providing new ideas for the prevention and treatment of AD.
Collapse
Affiliation(s)
- H Zhu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Geriatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - C Huang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Geriatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Z Luo
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Geriatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - L Wu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - X Cheng
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Geriatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - H Wu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Geriatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
26
|
Gao S, Leng Y, Qiu Z, Li K, Li J, Peng J, Xie W, Lei S, Xia Z. Burn-Induced Gut Microbiota Dysbiosis Aggravates Skeletal Muscle Atrophy by Tryptophan-Kynurenine Mediated AHR Pathway Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409296. [PMID: 39950940 PMCID: PMC11984878 DOI: 10.1002/advs.202409296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 01/06/2025] [Indexed: 04/12/2025]
Abstract
The hypermetabolic response associated with burns is characterized by skeletal muscle atrophy and an increased incidence of disability and death. Significant remodeling of the gut microbiota occurs after severe burn trauma. However, the specific mechanisms by which gut microbiota contribute to burn-induced muscle atrophy remain unexplored. The results showed that the disruption of the gut microbiota exacerbated skeletal muscle atrophy. Fecal metabolite analysis revealed perturbations, primarily within the tryptophan (Trp) metabolic pathway. Animal models further demonstrated that gut microbiota disorder enhanced the expression of indoleamine 2,3-dioxygenase 1 (IDO-1) in the colon, ultimately resulting in Trp depletion and increased kynurenine (Kyn) levels in the serum and skeletal muscle. Excessive colonic Kyn is released into circulation, transported into skeletal muscle cells, and binds to the aryl hydrocarbon receptor (AHR), consequently triggering AHR nuclear translocation and initiating the transcription of skeletal muscle atrophy-related genes. Notably, serum samples from patients with burns exhibited Trp depletion, and Trp supplementation alleviated skeletal muscle atrophy in rats with burns. This study, for the first time, demonstrates that gut microbiota dysbiosis upregulates colonic IDO-1, promotes Trp-Kyn metabolism, and exacerbates burn-induced skeletal muscle atrophy, suggesting that Trp supplementation may be a potential therapeutic strategy.
Collapse
Affiliation(s)
- Shan Gao
- Department of AnaesthesiologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Department of AnaesthesiologyTongren Hospital of Wuhan University WuhanWuhanHubei430060China
| | - Yan Leng
- Department of AnaesthesiologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Zhen Qiu
- Department of AnaesthesiologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Kai Li
- Department of PainTongren Hospital of Wuhan UniversityWuhanHubei430060China
| | - Jun Li
- Department of AnaesthesiologyTongren Hospital of Wuhan University WuhanWuhanHubei430060China
| | - Jian Peng
- Department of AnaesthesiologyTongren Hospital of Wuhan University WuhanWuhanHubei430060China
| | - Weiguo Xie
- Department of BurnsTongren Hospital of Wuhan UniversityWuhanHubei430060China
| | - Shaoqing Lei
- Department of AnaesthesiologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Zhongyuan Xia
- Department of AnaesthesiologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| |
Collapse
|
27
|
He T, Chen Q, Li H, Mao J, Luo J, Ma D, Yang Z. The potential mechanism of MicroRNA involvement in the regulation of muscle development in weaned piglets by tryptophan and its metabolites. BMC Genomics 2025; 26:330. [PMID: 40169975 PMCID: PMC11963679 DOI: 10.1186/s12864-025-11424-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Muscle development is a key factor influencing the growth performance of piglets. Optimizing this developmental process is crucial for enhancing breeding efficiency and economic profitability. Tryptophan (Trp) is considered one of the key limiting amino acids for weaned piglets, plays an essential role in regulating feed intake, growth, and muscle development. However, the regulatory mechanisms by which Trp and its derivatives influence muscle development in weaned piglets remain unclear. METHODS The aim of this study was to investigate the regulatory pathways and potential mechanisms of Trp and its metabolites on muscle development in weaned piglets. In this study, 10 healthy castrated male piglets, 28 days old and weaned, were selected and randomly assigned to a control group (CON, 0.14% Trp) and a high tryptophan group (HT, 0.35% Trp), with 5 in each group. After a 7-day pre-feeding period, the formal feeding began, and after 28 days, the pigs were slaughtered and the longissimus dorsi muscles was collected for transcriptome sequencing. RESULTS The results indicated that different dietary Trp levels led to the identification of sixteen differentially expressed microRNAs (DE miRNAs) in the longissimus dorsi muscle of the weaned piglets. Target gene functional enrichment analysis showed that these DE miRNAs are involved in muscle cell proliferation, differentiation, protein deposition, and muscle development through multiple biological pathways. Furthermore, we constructed a protein-protein interaction (PPI) network for the target genes, with the enriched core gene cluster functions associated with cellular proliferation, signaling pathways, hormone release, and muscle development. Finally, qRT-PCR validated the reliability and accuracy of the RNA-seq results, revealing a correlation coefficient of 0.97 between the two methods. CONCLUSIONS This study uncovers the potential mechanisms by which miRNAs participate in the regulation of muscle development in weaned piglets mediated by Trp and its metabolites, providing a theoretical basis and practical guidance for optimizing piglet management and health improvement.
Collapse
Affiliation(s)
- Tianle He
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China
| | - Qingyun Chen
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Huifeng Li
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Jiani Mao
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Ju Luo
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Dengjun Ma
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Zhenguo Yang
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
28
|
Li G, Dong S, Liu C, Yang J, Rensen PCN, Wang Y. Serotonin signaling to regulate energy metabolism: a gut microbiota perspective. LIFE METABOLISM 2025; 4:loae039. [PMID: 39926388 PMCID: PMC11803461 DOI: 10.1093/lifemeta/loae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/11/2024] [Accepted: 11/21/2024] [Indexed: 02/11/2025]
Abstract
Serotonin is one of the most potent gastrointestinal, peripheral, and neuronal signaling molecules and plays a key role in regulating energy metabolism. Accumulating evidence has shown the complex interplay between gut microbiota and host energy metabolism. In this review, we summarize recent findings on the role of gut microbiota in serotonin metabolism and discuss the complicated mechanisms by which serotonin, working in conjunction with the gut microbiota, affects total body energy metabolism in the host. Gut microbiota affects serotonin synthesis, storage, release, transport, and catabolism. In addition, serotonin plays an indispensable role in regulating host energy homeostasis through organ crosstalk and microbe-host communication, particularly with a wide array of serotonergic effects mediated by diverse serotonin receptors with unique tissue specificity. This fresh perspective will help broaden the understanding of serotonergic signaling in modulating energy metabolism, thus shedding light on the design of innovative serotonin-targeting strategies to treat metabolic diseases.
Collapse
Affiliation(s)
- Guoli Li
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Sijing Dong
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Department of Endocrinology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Chunhao Liu
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Jing Yang
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Department of Endocrinology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Patrick C N Rensen
- Department of Endocrinology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Yanan Wang
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Department of Endocrinology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
29
|
Kaur M, Porel P, Patel R, Aran KR. Kynurenine Pathway in Epilepsy: Unraveling Its Role in Glutamate Excitotoxicity, GABAergic Dysregulation, Neuroinflammation, and Mitochondrial Dysfunction. Neurotox Res 2025; 43:18. [PMID: 40153181 DOI: 10.1007/s12640-025-00738-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 03/09/2025] [Accepted: 03/15/2025] [Indexed: 03/30/2025]
Abstract
Epilepsy is a chronic noncommunicable neurological disorder characterized by recurrent seizures and ranks as the seventh most prevalent neurological disease globally. According to the Global Burden of Disease report, 3.40 billion people were affected by epilepsy in 2021. The pathophysiology of epilepsy states that a disturbed balance between excitatory and inhibitory signaling at the synaptic level, which can cause seizure activity, is similar across epilepsies and includes mitochondrial dysfunction, neuroinflammation, and kynurenine metabolites such as kynurenic acid and quinolinic acid. The kynurenine pathway (KP) is the major metabolic pathway in which tryptophan (TRP) is the key precursor which is further converted into a variety of neuroactive substances that can have both neurotoxic metabolites (Quinolinic acid) and neuroprotective metabolites such as kynurenic acid, and picolinic acid. KP plays a significant role in the brain such as the metabolism of TRP, the production of metabolites, and its impact on aging. However, higher concentrations of kynurenine and its metabolites, such as quinolinic acid may increase the frequency and intensity of seizures, and dysregulation of the KP has been linked to the pathophysiology of epilepsy. Concurrently, glutamate and GABA signaling is altered by neuroinflammatory processes linked to epilepsy, which results in excitotoxic neuronal damage. This review aims to provide novel therapeutic strategies that might improve the prognosis of individuals with epilepsy and related disorders by elucidating the mechanisms underlying KP dysregulation in these circumstances. To develop targeted therapies for CNS disorders characterized by inflammation and seizures, it is essential to understand how kynurenine metabolites both promote and prevent excitotoxicity.
Collapse
Affiliation(s)
- Manpreet Kaur
- Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Pratyush Porel
- Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Royal Patel
- School of Pharmacy, LNCT University, Bhopal, 462042, India
| | - Khadga Raj Aran
- Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| |
Collapse
|
30
|
Kim K, Thome T, Pass C, Stone L, Vugman N, Palzkill V, Yang Q, O’Malley KA, Anderson EM, Fazzone B, Yue F, Berceli SA, Scali ST, Ryan TE. Multiomic Analysis of Calf Muscle in Peripheral Artery Disease and Chronic Kidney Disease. Circ Res 2025; 136:688-703. [PMID: 39963788 PMCID: PMC11949227 DOI: 10.1161/circresaha.124.325642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND Chronic kidney disease (CKD) has emerged as a significant risk factor that accelerates atherosclerosis, decreases muscle function, and increases the risk of amputation or death in patients with peripheral artery disease (PAD). However, the modulators underlying this exacerbated pathobiology are ill-defined. Recent work has demonstrated that uremic toxins are associated with limb amputation in PAD and have pathological effects in both the limb muscle and vasculature. Herein, we use multiomics to identify novel modulators of disease pathobiology in patients with PAD and CKD. METHODS A cross-sectional study enrolled 4 groups of participants: controls without PAD or CKD (n=28), patients with PAD only (n=46), patients with CKD only (n=31), and patients with both PAD and CKD (n=18). Both targeted (uremic toxins) and nontargeted metabolomics in plasma were performed using mass spectrometry. Calf muscle biopsies were used to measure histopathology, perform bulk and single-nucleus RNA sequencing, and assess mitochondrial function. Differential gene and metabolite analyses, as well as pathway and gene set enrichment analyses, were performed. RESULTS Patients with both PAD and CKD exhibited significantly lower calf muscle strength and smaller muscle fiber areas compared with controls and those with only PAD. Compared with controls, mitochondrial function was impaired in patients with CKD, with or without PAD, but not in PAD patients without CKD. Plasma metabolomics revealed substantial alterations in the metabolome of patients with CKD, with significant correlations observed between uremic toxins (eg, kynurenine and indoxyl sulfate) and both muscle strength and mitochondrial function. RNA sequencing analyses identified downregulation of mitochondrial genes and pathways associated with protein translation in patients with both PAD and CKD. Single-nucleus RNA sequencing further highlighted a mitochondrial deficiency in muscle fibers along with unique remodeling of fibro-adipogenic progenitor cells in patients with both PAD and CKD, with an increase in adipogenic cell populations. CONCLUSIONS CKD significantly exacerbates ischemic muscle pathology in PAD, as evidenced by diminished muscle strength, reduced mitochondrial function, and altered transcriptome profiles. The correlation between uremic toxins and muscle dysfunction suggests that targeting these metabolites may offer therapeutic potential for improving muscle health in PAD patients with CKD.
Collapse
Affiliation(s)
- Kyoungrae Kim
- Department of Applied Physiology and Kinesiology (K.K., T.T., C.P., L.S., N.V., V.P., Q.Y., T.E.R.)
| | - Trace Thome
- Department of Applied Physiology and Kinesiology (K.K., T.T., C.P., L.S., N.V., V.P., Q.Y., T.E.R.)
| | - Caroline Pass
- Department of Applied Physiology and Kinesiology (K.K., T.T., C.P., L.S., N.V., V.P., Q.Y., T.E.R.)
| | - Lauren Stone
- Department of Applied Physiology and Kinesiology (K.K., T.T., C.P., L.S., N.V., V.P., Q.Y., T.E.R.)
| | - Nicholas Vugman
- Department of Applied Physiology and Kinesiology (K.K., T.T., C.P., L.S., N.V., V.P., Q.Y., T.E.R.)
| | - Victoria Palzkill
- Department of Applied Physiology and Kinesiology (K.K., T.T., C.P., L.S., N.V., V.P., Q.Y., T.E.R.)
| | - Qingping Yang
- Department of Applied Physiology and Kinesiology (K.K., T.T., C.P., L.S., N.V., V.P., Q.Y., T.E.R.)
| | | | | | - Brian Fazzone
- Department of Surgery (K.A.O., E.M.A., B.F., S.A.B., S.T.S.)
| | - Feng Yue
- Department of Animal Sciences (F.Y.)
- Myology Institute (F.Y., T.E.R.)
| | | | | | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology (K.K., T.T., C.P., L.S., N.V., V.P., Q.Y., T.E.R.)
- Center for Exercise Science (T.E.R.)
- Myology Institute (F.Y., T.E.R.)
- University of Florida, Gainesville (T.E.R.)
| |
Collapse
|
31
|
Murgiano M, Bartocci B, Puca P, di Vincenzo F, Del Gaudio A, Papa A, Cammarota G, Gasbarrini A, Scaldaferri F, Lopetuso LR. Gut Microbiota Modulation in IBD: From the Old Paradigm to Revolutionary Tools. Int J Mol Sci 2025; 26:3059. [PMID: 40243712 PMCID: PMC11988433 DOI: 10.3390/ijms26073059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 03/18/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Inflammatory bowel diseases (IBDs) are chronic inflammatory disorders primarily comprising two main conditions: ulcerative colitis and Crohn's disease. The gut microbiota's role in driving inflammation in IBD has garnered significant attention, yet the precise mechanisms through which the microbiota influences IBD pathogenesis remain largely unclear. Given the limited therapeutic options for IBD, alternative microbiota-targeted therapies-including prebiotics, probiotics, postbiotics, and symbiotics-have been proposed. While these approaches have shown promising results, microbiota modulation is still mainly considered an adjunct therapy to conventional treatments, with a demonstrated impact on patients' quality of life. Fecal microbiota transplantation (FMT), already approved for treating Clostridioides difficile infection, represents the first in a series of innovative microbiota-based therapies under investigation. Microbial biotherapeutics are emerging as personalized and cutting-edge tools for IBD management, encompassing next-generation probiotics, bacterial consortia, bacteriophages, engineered probiotics, direct metabolic pathway modulation, and nanotherapeutics. This review explores microbial modulation as a therapeutic strategy for IBDs, highlighting current approaches and examining promising tools under development to better understand their potential clinical applications in managing intestinal inflammatory disorders.
Collapse
Affiliation(s)
- Marco Murgiano
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
| | - Bianca Bartocci
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
| | - Pierluigi Puca
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Federica di Vincenzo
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
| | - Angelo Del Gaudio
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
| | - Alfredo Papa
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Giovanni Cammarota
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Franco Scaldaferri
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Loris Riccardo Lopetuso
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Dipartimento di Scienze della Vita, della Salute e delle Professioni Sanitarie, Università degli Studi Link, 00165 Rome, Italy
| |
Collapse
|
32
|
Wang Y, Wu P, Chen Z, Li Z, Wang Y, Yan M, Zhang Y, Wang S, Fang S, Yu B. Prognostic value of tryptophan catabolism-base scores in acute myocardial infarction patients. J Adv Res 2025:S2090-1232(25)00185-7. [PMID: 40147625 DOI: 10.1016/j.jare.2025.03.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/09/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
AIMS Tryptophan catabolism is implicated in the progression of cardiovascular disease. We sought to investigate the prognostic value of tryptophan catabolism-related features in patients with acute myocardial infarction (AMI). METHODS AND RESULTS A prospective cohort of 4071 patients (mean age: 60.7 years; 69.1 % men) with AMI between February 2017 and June 2019 was included and followed up for a median of 5.6 years (IQR 5.1-6.2). There were 666 all-cause deaths, 365 cardiovascular deaths, and 559 HF events. Plasma levels of tryptophan-related metabolites were measured using liquid chromatography tandem mass spectrometry (LC-MS/MS), and were repeatedly determined in 1044 patients after discharge. Tryptophan, kynurenine, indole-3-propionic acid, and indole-3-lactic acid were screened to construct tryptophan metabolites combination (TMC) score using coefficients from predictive models for MACE. Patients were divided into 3 groups by TMC tertiles. Patients with higher TMC score were older, more likely to be male and have hypertension. Compared to those with TMC tertile 1, patients in TMC tertile 3 had significant associations with the risk of all-cause death (HR: 1.90; 95 %CI: 1.54-2.34), cardiovascular death (HR: 2.32; 95 %CI: 1.71-3.15) and incident HF (HR: 1.77; 95 %CI: 1.40-2.24). The incremental prognostic value of TMC score over the Grace score was measured by the likelihood ratio, C-statistic, continuous net reclassification improvement (NRI), and integrated discrimination improvement (IDI) for prediction, discrimination, and reclassification of outcomes. CONCLUSIONS In this hospital-based AMI cohort, the TMC score was significantly associated with all-cause mortality, cardiovascular mortality, and incident HF, and improved risk stratification beyond established clinical risk factors. The TMC score provided a novel tool for assessment of Trp catabolism dysfunction and outcomes risk.
Collapse
Affiliation(s)
- Ye Wang
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, China
| | - Pengyan Wu
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, China
| | - Zhanchao Chen
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, China
| | - Zhaoying Li
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, China
| | - Yini Wang
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China
| | - Miao Yan
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, China
| | - Yiying Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Jiamusi University, Jiamusi, China
| | - Shanjie Wang
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, China.
| | - Shaohong Fang
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, China.
| | - Bo Yu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), China; Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, China.
| |
Collapse
|
33
|
Wang Y, Zhang Y, Wang W, Zhang Y, Dong X, Liu Y. Diverse Physiological Roles of Kynurenine Pathway Metabolites: Updated Implications for Health and Disease. Metabolites 2025; 15:210. [PMID: 40137174 PMCID: PMC11943880 DOI: 10.3390/metabo15030210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
Tryptophan is an essential amino acid critical for human health. It plays a pivotal role in numerous physiological and biochemical processes through its metabolism. The kynurenine (KYN) pathway serves as the principal metabolic route for tryptophan, producing bioactive metabolites, including KYN, quinolinic acid, and 3-hydroxykynurenine. Numerous studies are actively investigating the relationship between tryptophan metabolism and physiological functions. These studies are highlighting the interactions among metabolites that may exert synergistic or antagonistic effects, such as neuroprotective or neurotoxic, and pro-oxidative or antioxidant activities. Minor disruptions in the homeostasis of these metabolites can result in immune dysregulation, contributing to a spectrum of diseases. These diseases include neurological disorders, mental illnesses, cardiovascular conditions, autoimmune diseases, and chronic kidney disease. Therefore, understanding the physiological roles of the KYN pathway metabolites is essential for elucidating the contribution of tryptophan metabolism to health regulation. The present review emphasizes the physiological roles of KYN pathway metabolites and their mechanisms in disease development, aiming to establish a theoretical basis for leveraging dietary nutrients to enhance human health.
Collapse
Affiliation(s)
| | | | | | | | | | - Yang Liu
- Shandong Food Ferment Industry & Design Institute, QiLu University of Technology (Shandong Academy of Sciences), No. 41, Jiefang Road, Jinan 250013, China
| |
Collapse
|
34
|
Rangel MVDS, Lopes KG, Qin X, Borges JP. Exercise-induced adaptations in the kynurenine pathway: implications for health and disease management. Front Sports Act Living 2025; 7:1535152. [PMID: 40115437 PMCID: PMC11922725 DOI: 10.3389/fspor.2025.1535152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/25/2025] [Indexed: 03/23/2025] Open
Abstract
Background Tryptophan (TRP) metabolism through the kynurenine (KYN) pathway is influenced by inflammatory mediators, generating metabolites that regulate immune and inflammatory responses. Exercise has been proposed as a modulator of this pathway, but its role in health benefits and chronic disease management remains unclear. Objective This systematic review examines exercise-induced adaptations in the KYN pathway and their potential implications for health and disease management. Additionally, we identify key methodological considerations for future research. Methods A structured search of PubMed/Medline, Web of Science, and Scopus was conducted up to October 2024 to identify clinical trials investigating the effects of exercise training on the KYN pathway. Results Of 2,795 articles initially found, 13 clinical trials involving 592 participants met the inclusion criteria. Most studies reported exercise-induced adaptations in the KYN pathway, particularly in cancer survivors. These adaptations appeared to be influenced by exercise intensity and duration. However, several methodological limitations were noted, and no trials included patients with metabolic or cardiovascular diseases. Conclusions Here, we show that exercise training modulates the KYN pathway in both healthy and diseased populations, highlighting its potential for disease prevention and management. However, further randomized-controlled trials are needed to clarify its mechanisms and clinical applications, particularly in metabolic and cardiovascular diseases. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO/view/CRD42022351481, PROSPERO (CRD42022351481).
Collapse
Affiliation(s)
- Marcus Vinicius Dos Santos Rangel
- Laboratory of Physical Activity and Health Promotion, Institute of Physical Education and Sports, University of Rio de Janeiro State, Rio de Janeiro, RJ, Brazil
| | - Karynne Grutter Lopes
- Postgraduate Program in Clinical and Experimental Physiopathology, Faculty of Medical Sciences, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Xuebin Qin
- Department of Microbiology and Immunology, Tulane National Primate Research Center and Tulane University School of Medicine, Covington, LA, United States
| | - Juliana Pereira Borges
- Laboratory of Physical Activity and Health Promotion, Institute of Physical Education and Sports, University of Rio de Janeiro State, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
35
|
Sapienza J, Agostoni G, Repaci F, Spangaro M, Comai S, Bosia M. Metabolic Syndrome and Schizophrenia: Adding a Piece to the Interplay Between the Kynurenine Pathway and Inflammation. Metabolites 2025; 15:176. [PMID: 40137141 PMCID: PMC11944102 DOI: 10.3390/metabo15030176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/29/2025] [Accepted: 02/21/2025] [Indexed: 03/27/2025] Open
Abstract
The biology of schizophrenia is highly complex and multifaceted. Numerous efforts have been made over the years to disentangle the heterogeneity of the disease, gradually leading to a more detailed understanding of its underlying pathogenic mechanisms. Two cardinal elements in the pathophysiology of schizophrenia are neuroinflammation and alterations of neurotransmission. The kynurenine (KYN) pathway (KP) is of particular importance because it is inducted by systemic low-grade inflammation in peripheral tissues, producing metabolites that are neuroactive (i.e., modulating glutamatergic and cholinergic neurotransmission), neuroprotective, or neurotoxic. Consequently, the KP is at the crossroads between two primary systems involved in the pathogenesis of schizophrenia. It bridges the central nervous system (CNS) and the periphery, as KP metabolites can cross the blood-brain barrier and modulate neuronal activity. Metabolic syndrome plays a crucial role in this context, as it frequently co-occurs with schizophrenia, contributing to a sub-inflammatory state able to activate the KP. This narrative review provides valuable insights into these complex interactions, offering a framework for developing targeted therapeutic interventions or precision psychiatry approaches of the disorder.
Collapse
Affiliation(s)
- Jacopo Sapienza
- Schizophrenia Research and Clinical Unit, IRCCS San Raffaele Scientific Institute, 20127 Milan, Italy; (J.S.)
- Department of Humanities and Life Sciences, University School for Advanced Studies IUSS, 27100 Pavia, Italy
| | - Giulia Agostoni
- Schizophrenia Research and Clinical Unit, IRCCS San Raffaele Scientific Institute, 20127 Milan, Italy; (J.S.)
| | - Federica Repaci
- Schizophrenia Research and Clinical Unit, IRCCS San Raffaele Scientific Institute, 20127 Milan, Italy; (J.S.)
| | - Marco Spangaro
- Schizophrenia Research and Clinical Unit, IRCCS San Raffaele Scientific Institute, 20127 Milan, Italy; (J.S.)
| | - Stefano Comai
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35123 Padua, Italy
- Division of Neurosciences, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Department of Psychiatry, McGill University, Montreal, QC H3A 0G4, Canada
- Department of Biomedical Sciences, University of Padua, 35123 Padua, Italy
| | - Marta Bosia
- Schizophrenia Research and Clinical Unit, IRCCS San Raffaele Scientific Institute, 20127 Milan, Italy; (J.S.)
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
| |
Collapse
|
36
|
Ghosh A, Rayanki N, Joshi AR, Kulkarni OP. UPF 648, a Selective KMO Inhibitor, Attenuates Psychomotor and Cognitive Impairment in Chronic Kidney Disease. ACS Chem Neurosci 2025; 16:908-919. [PMID: 39961731 DOI: 10.1021/acschemneuro.4c00844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025] Open
Abstract
Kynurenine-3-monooxygenase (KMO), a key enzyme in the kynurenine pathway (KP) of tryptophan metabolism, converts kynurenine into the neurotoxic intermediate quinolinic acid (QA). QA, an N-methyl-d-aspartate (NMDA) receptor agonist, increases glutamate release and inhibits its reuptake, resulting in excitotoxic cell death in the hippocampus and striatum. Plasma metabolomics study exhibited KP metabolites as the most altered pathway in patients with chronic kidney disease (CKD). Recently, QA was linked to the kidney-brain axis as one of the major neurotoxins responsible for cognitive impairment in advanced CKD stages. Various preclinical models are being tested to explore different intermediates of KP that can be targeted to ameliorate the central nervous system (CNS) complications of CKD. In this study, an adenine-induced CKD model was developed in C57BL/6 mice, where UPF 648, a selective KMO inhibitor, was administered to observe the changes in KP metabolites in the hippocampus. Treatment with UPF 648 did not alter kidney function or morphology in CKD. KMO inhibition led to decreased plasma QA levels and reduced levels of pro-inflammatory cytokine interleukin-1-β (IL-1β). UPF 648 treatment in CKD ameliorated the characteristic symptoms of motor dysfunction, anxiety, depression, and hippocampus-dependent memory. Important markers for neuronal survival and plasticity through the brain-derived neurotrophic factor (BDNF)-tropomyosin receptor kinase B (TRKB)-cAMP-responsive element binding protein 1 (CREB1) pathway were upregulated in the hippocampus after KMO inhibition. In conclusion, KMO inhibition can be an exciting target to attenuate the neuropsychiatric burden of advanced stages in CKD.
Collapse
Affiliation(s)
- Aparajita Ghosh
- Metabolic Disorders and Neuroscience Research Lab, Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Hyderabad Campus, Telangana 500078, India
| | - Neeraja Rayanki
- Metabolic Disorders and Neuroscience Research Lab, Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Hyderabad Campus, Telangana 500078, India
| | - Abhijeet R Joshi
- Metabolic Disorders and Neuroscience Research Lab, Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Hyderabad Campus, Telangana 500078, India
| | - Onkar P Kulkarni
- Metabolic Disorders and Neuroscience Research Lab, Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Hyderabad Campus, Telangana 500078, India
| |
Collapse
|
37
|
Wang M, Liu Y, Zhong L, Wu F, Wang J. Advancements in the investigation of gut microbiota-based strategies for stroke prevention and treatment. Front Immunol 2025; 16:1533343. [PMID: 40103814 PMCID: PMC11914130 DOI: 10.3389/fimmu.2025.1533343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Stroke represents a predominant cause of mortality and disability on a global scale, impacting millions annually and exerting a considerable strain on healthcare systems. The incidence of stroke exhibits regional variability, with ischemic stroke accounting for the majority of occurrences. Post-stroke complications, such as cognitive impairment, motor dysfunction, and recurrent stroke, profoundly affect patients' quality of life. Recent advancements have elucidated the microbiota-gut-brain axis (MGBA), underscoring the complex interplay between gut health and brain function. Dysbiosis, characterized by an imbalance in gut microbiota, is significantly linked to an elevated risk of stroke and unfavorable outcomes. The MGBA plays a crucial role in modulating immune function, neurotransmitter levels, and metabolic byproducts, which may intensify neuroinflammation and impair cerebral health. This review elucidates the role of MGBA in stroke pathophysiology and explores potential gut-targeted therapeutic strategies to reduce stroke risk and promote recovery, including probiotics, prebiotics, pharmacological interventions, and dietary modifications. However, the current prevention and treatment strategies based on intestinal flora still face many problems, such as the large difference of individual intestinal flora, the stability of efficacy, and the long-term safety need to be considered. Further research needs to be strengthened to promote its better application in clinical practice.
Collapse
Affiliation(s)
| | | | | | | | - Jinjin Wang
- Department of Gastroenterology, The First People’s Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| |
Collapse
|
38
|
Yang S, Liu H, Liu Y. Advances in intestinal epithelium and gut microbiota interaction. Front Microbiol 2025; 16:1499202. [PMID: 40104591 PMCID: PMC11914147 DOI: 10.3389/fmicb.2025.1499202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/17/2025] [Indexed: 03/20/2025] Open
Abstract
The intestinal epithelium represents a critical interface between the host and external environment, serving as the second largest surface area in the human body after the lungs. This dynamic barrier is sustained by specialized epithelial cell types and their complex interactions with the gut microbiota. This review comprehensively examines the recent advances in understanding the bidirectional communication between intestinal epithelial cells and the microbiome. We briefly highlight the role of various intestinal epithelial cell types, such as Paneth cells, goblet cells, and enteroendocrine cells, in maintaining intestinal homeostasis and barrier function. Gut microbiota-derived metabolites, particularly short-chain fatty acids and bile acids, influence epithelial cell function and intestinal barrier integrity. Additionally, we highlight emerging evidence of the sophisticated cooperation between different epithelial cell types, with special emphasis on the interaction between tuft cells and Paneth cells in maintaining microbial balance. Understanding these complex interactions has important implications for developing targeted therapeutic strategies for various gastrointestinal disorders, including inflammatory bowel disease, metabolic disorders, and colorectal cancer.
Collapse
Affiliation(s)
- Sen Yang
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Pediatrics, The Fifth Peoples Hospital of Chengdu, Chengdu, China
| | - Hanmin Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yang Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
Li J, Liu T, Xian M, Zhou K, Wei J. The Power of Exercise: Unlocking the Biological Mysteries of Peripheral-Central Crosstalk in Parkinson's Disease. J Adv Res 2025:S2090-1232(25)00143-2. [PMID: 40049515 DOI: 10.1016/j.jare.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 01/06/2025] [Accepted: 03/01/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Exercise is a widely recognized non-pharmacological treatment for Parkinson's Disease (PD). The bidirectional regulation between the brain and peripheral organs has emerged as a promising area of research, with the mechanisms by which exercise impacts PD closely linked to the interplay between peripheral signals and the central nervous system. AIM OF REVIEW This review aims to summarize the mechanisms by which exercise influences peripheral-central crosstalk to improve PD, discuss the molecular processes mediating these interactions, elucidate the pathways through which exercise may modulate PD pathophysiology, and identify directions for future research. KEY SCIENTIFIC CONCEPTS OF REVIEW This review examines how exercise-induced cytokine release promotes neuroprotection in PD. It discusses how exercise can stimulate cytokine secretion through various pathways, including the gut-brain, muscle-brain, liver-brain, adipose-brain, and bone-brain axes, thereby alleviating PD symptoms. Additionally, the potential contributions of the heart-brain, lung-brain, and spleen-brain axes, as well as multi-axis crosstalk-such as the brain-gut-muscle and brain-gut-bone axes-are explored in the context of exercise therapy. The study highlights the need for further research into peripheral-central crosstalk and outlines future directions to address challenges in clinical PD therapy.
Collapse
Affiliation(s)
- Jingwen Li
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, Henan, 475004, China
| | - Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Meiyan Xian
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Ke Zhou
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, Henan, 475004, China.
| | - Jianshe Wei
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, Henan, 475004, China; Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China.
| |
Collapse
|
40
|
Sun P, Wang M, Chai X, Liu YX, Li L, Zheng W, Chen S, Zhu X, Zhao S. Disruption of tryptophan metabolism by high-fat diet-triggered maternal immune activation promotes social behavioral deficits in male mice. Nat Commun 2025; 16:2105. [PMID: 40025041 PMCID: PMC11873046 DOI: 10.1038/s41467-025-57414-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 02/20/2025] [Indexed: 03/04/2025] Open
Abstract
Diet-related maternal obesity has been implicated in neurodevelopmental disorders in progeny. Although the precise mechanisms and effective interventions remain uncertain, our research elucidates some of these complexities. We established that a prenatal high-fat diet triggered maternal immune activation (MIA), marked by elevated serum lipopolysaccharide levels and inflammatory-cytokine overproduction, which dysregulated the maternal tryptophan metabolism promoting the accumulation of neurotoxic kynurenine metabolites in the embryonic brain. Interventions aimed at mitigating MIA or blocking the kynurenine pathway effectively rescued the male mice social performance. Furthermore, excessive kynurenine metabolites initiated oxidative stress response causing neuronal migration deficits in the fetal neocortex, an effect that was mitigated by administering the glutathione synthesis precursor N-Acetylcysteine, underscoring the central role of maternal immune-metabolic homeostasis in male mice behavioral outcomes. Collectively, our study accentuated the profound influence of maternal diet-induced immuno-metabolic dysregulation on fetal brain development and provided the preventive strategies for addressing neurodevelopmental disorders.
Collapse
Affiliation(s)
- Penghao Sun
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Mengli Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xuejun Chai
- College of Basic Medicine, Xi'an Medical University, Xi'an, Shaanxi, China.
| | - Yong-Xin Liu
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, Guangdong, China
| | - Luqi Li
- Life Science Research Core Services, Northwest A&F University, Yangling, Shaanxi, China
| | - Wei Zheng
- College of Resources and Environment Sciences, Northwest A&F University, Yangling, Shaanxi, China
| | - Shulin Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiaoyan Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| | - Shanting Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
41
|
Dos Santos Y, Emond P, Schwartz IVD, Lefèvre A, Dupuy C, Chicheri G, Blasco H, Maillot F. Multimodal Metabolomic Analysis Reveals Novel Metabolic Disturbances in Adults With Early Treated Phenylketonuria. JIMD Rep 2025; 66:e70010. [PMID: 40135139 PMCID: PMC11932803 DOI: 10.1002/jmd2.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
Phenylketonuria (PKU) is an inborn error of metabolism responsible for an accumulation of phenylalanine, which leads to cognitive and developmental disorders if left untreated. Most studies of adult PKU focus on neuropsychiatric complications, but new questions have been raised about systemic manifestations of PKU in adulthood. Fifteen adults with classic PKU with poor metabolic control and 15 matched healthy controls were recruited to compare their blood metabolomes by an untargeted multimodal approach (polar, apolar, and lipids) by LC/MS and a targeted approach to the tryptophan pathway. Targeted analysis revealed systemic serotonin hypometabolism and aberrant kynurenine metabolism, as well as potential implication of microbiota by differences in some indole compounds compared to controls. Untargeted analysis confirms previous findings regarding the TCA cycle, alanine aspartate glutamate metabolism, arginine and proline metabolism, and revealed some new metabolic perturbations such as arginine biosynthesis or glyoxylate and dicarboxylate metabolism. Future studies involving larger numbers of patients with varying degrees of metabolic control are needed to confirm these findings.
Collapse
Affiliation(s)
- Yann Dos Santos
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253ToursFrance
| | - Patrick Emond
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253ToursFrance
- In Vitro Nuclear Medicine DepartmentUniversity Hospital of ToursToursFrance
| | - Ida Vanessa Doederlein Schwartz
- Medical Genetics ServiceHospital de Clinicas de Porto AlegrePorto AlegreBrazil
- Departamento de GeneticaUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Antoine Lefèvre
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253ToursFrance
| | - Camille Dupuy
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253ToursFrance
| | - Gabrielle Chicheri
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253ToursFrance
| | - Hélène Blasco
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253ToursFrance
- Department of Biochemistry and Molecular BiologyUniversity Hospital of ToursToursFrance
| | - François Maillot
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253ToursFrance
- Department of Internal MedicineUniversity Hospital of ToursToursFrance
| |
Collapse
|
42
|
Wang Y, Leung E, Tomek P. N-formylkynurenine but not kynurenine enters a nucleophile-scavenging branch of the immune-regulatory kynurenine pathway. Bioorg Chem 2025; 156:108219. [PMID: 39891998 DOI: 10.1016/j.bioorg.2025.108219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/18/2025] [Accepted: 01/26/2025] [Indexed: 02/03/2025]
Abstract
Tryptophan catabolism along the kynurenine pathway (KP) mediates key physiological functions ranging from immune tolerance to lens UV protection, but the contributory roles and chemical fates of individual KP metabolites are incompletely understood. This particularly concerns the first KP metabolite, N-formylkynurenine (NFK), canonically viewed as a transient precursor to the downstream kynurenine (KYN). Here, we challenge that canon and show that hydrolytic enzymes act as a rheostat switching NFK's fate between the canonical KP and a novel non-enzymatic branch of tryptophan catabolism. In the physiological environment (37 °C, pH 7.4), NFK deaminated into electrophilic NFK-carboxyketoalkene (NFK-CKA), which rapidly (<2 min) formed adducts with nucleophiles such as cysteine and glutathione, the key intracellular antioxidants. Serum hydrolases suppressed NFK deamination as they hydrolysed NFK to KYN ∼3 times faster than NFK deaminates. Whilst KYN did not deaminate, its deaminated product (KYN-CKA) rapidly reacted with cysteine but not glutathione. The new NFK transformations of a yet to be discovered function highlight NFK's significance beyond hydrolysis to KYN and suggests the dominance of its chemical transformations over those of KYN. Enzyme compartmentalisation and abundance offer insights into the regulation of non-enzymatic KP metabolite transformations such as KYN involved in immune regulation, protein modification, lens aging or neuropathology.
Collapse
Affiliation(s)
- Yongxin Wang
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland 1023 New Zealand
| | - Euphemia Leung
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland 1023 New Zealand
| | - Petr Tomek
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland 1023 New Zealand.
| |
Collapse
|
43
|
Erboz A, Kesekler E, Gentili PL, Uversky VN, Coskuner-Weber O. Electromagnetic radiation and biophoton emission in neuronal communication and neurodegenerative diseases. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 195:87-99. [PMID: 39732343 DOI: 10.1016/j.pbiomolbio.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/08/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
The intersection of electromagnetic radiation and neuronal communication, focusing on the potential role of biophoton emission in brain function and neurodegenerative diseases is an emerging research area. Traditionally, it is believed that neurons encode and communicate information via electrochemical impulses, generating electromagnetic fields detectable by EEG and MEG. Recent discoveries indicate that neurons may also emit biophotons, suggesting an additional communication channel alongside the regular synaptic interactions. This dual signaling system is analyzed for its potential in synchronizing neuronal activity and improving information transfer, with implications for brain-like computing systems. The clinical relevance is explored through the lens of neurodegenerative diseases and intrinsically disordered proteins, where oxidative stress may alter biophoton emission, offering clues for pathological conditions, such as Alzheimer's and Parkinson's diseases. The potential therapeutic use of Low-Level Laser Therapy (LLLT) is also examined for its ability to modulate biophoton activity and mitigate oxidative stress, presenting new opportunities for treatment. Here, we invite further exploration into the intricate roles the electromagnetic phenomena play in brain function, potentially leading to breakthroughs in computational neuroscience and medical therapies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Aysin Erboz
- Molecular Biotechnology, Turkish-German University, Sahinkaya Caddesi No. 106, Beykoz, Istanbul, 34820, Turkey
| | - Elif Kesekler
- Molecular Biotechnology, Turkish-German University, Sahinkaya Caddesi No. 106, Beykoz, Istanbul, 34820, Turkey
| | - Pier Luigi Gentili
- Department of Chemistry, Biology, and Biotechnology, Università degli Studi di Perugia, 06123, Perugia, Italy.
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Institute, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd., MDC07, Tampa, FL 33612, USA.
| | - Orkid Coskuner-Weber
- Molecular Biotechnology, Turkish-German University, Sahinkaya Caddesi No. 106, Beykoz, Istanbul, 34820, Turkey.
| |
Collapse
|
44
|
Li S, Zhou L, Ren J, Zhang Q, Xiao X. Maternal exercise programs placental miR-495-5p-mediated Snx7 expression and kynurenic acid metabolic pathway induced by prenatal high-fat diet: Based on miRNA-seq, transcriptomics, and metabolomics. J Nutr Biochem 2025; 137:109830. [PMID: 39647668 DOI: 10.1016/j.jnutbio.2024.109830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/21/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024]
Abstract
Poor intrauterine environments increase the prevalence of chronic metabolic diseases in offspring, whereas maternal exercise is an effective measure to break this vicious intergenerational cycle. Placenta is increasingly being studied to explore its role in maternal-fetal metabolic cross-talk. The association between placental miRNA and offspring development trajectories has been established, yet the specific role and mechanism thereof in maternal exercise-induced metabolic protection remain elusive. Here, C57BL/6 female mice were subjected to either a normal control or a high-fat diet (HFD), half of the HFD-fed dams were housed with voluntary wheel running for 3 weeks before and during gestation. At embryonic day 18.5, we sacrificed parturient mice and then conducted miRNA-seq, transcriptomic, and metabolomic profiling of the placenta. Our data revealed that maternal HFD resulted in significant alterations in both miRNA and gene expressions, as well as metabolic pathways of the placenta, whereas prenatal exercise negated these perturbations. The common differentially expressed transcripts among three groups were enriched in multiple critical pathways involving energy expenditure, signal transduction, and fetal development. Through integrated analysis of multiomics data, we speculated that maternal exercise reversed the suppression of miR-495-5p induced by HFD, thereby inhibiting miR-495-5p-targeted Snx7 and modulating kynurenic acid production. These datasets provided novel mechanistic insight into how maternal exercise positively affects the metabolic homeostasis of offspring. The discovered important miRNAs, mRNAs, and metabolites could be promising predictive and therapeutic targets for protecting offspring metabolic health.
Collapse
Affiliation(s)
- Shunhua Li
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Liyuan Zhou
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jing Ren
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Qian Zhang
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xinhua Xiao
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
45
|
Dong J, Xu L, Qu A, Hao C, Sun M, Xu C, Hu S, Kuang H. Chiral Inorganic Nanomaterial-Based Diagnosis and Treatments for Neurodegenerative Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2418723. [PMID: 39924754 DOI: 10.1002/adma.202418723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/27/2025] [Indexed: 02/11/2025]
Abstract
Chiral nanomaterials are widely investigated over recent decades due to their biocompatibility and unique chiral effects. These key properties have significantly promoted the rapid development of chiral nanomaterials in bioengineering and medicine. In this review, the basic principles of constructing chiral nanomaterials along with the latest progress in research are comprehensively summarized. Then, the application of chiral nanomaterials for the diagnosis of neurodegenerative diseases (NDDs) is systematically described. In addition, the significant potential and broad prospects of chiral nanomaterials in the treatment of NDDs are highlighted from several aspects, including the disaggregation of neurofibrils, the scavenging of reactive oxygen species, regulation of the microbial-gut-brain axis, the elimination of senescent cells, and the promotion of directed differentiation in neural stem cells. Finally, a perspective of the challenges and future development of chiral nanomaterials for the treatment of NDDs is provided.
Collapse
Affiliation(s)
- Jingqi Dong
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Liguang Xu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Aihua Qu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Changlong Hao
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Maozhong Sun
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Chuanlai Xu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Shudong Hu
- Department of Radiology, Affiliated Hospital, Jiangnan University, No. 1000, Hefeng Road, Wuxi, Jiangsu, 214122, China
| | - Hua Kuang
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
46
|
Xiao T, Chen Y, Jiang B, Huang M, Liang Y, Xu Y, Zheng X, Wang W, Chen X, Cai G. Ultrasound-guided renal subcapsular transplantation of mesenchymal stem cells for treatment of acute kidney injury in a minipig model: safety and efficacy evaluation. Stem Cell Res Ther 2025; 16:102. [PMID: 40022148 PMCID: PMC11871648 DOI: 10.1186/s13287-025-04137-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 01/10/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is a major global public health concern with limited treatment options. While preclinical studies have suggested the potential of mesenchymal stem cells (MSCs) to repair and protect injured kidneys in AKI, clinical trials using transarterial MSCs transplantation have yielded disappointing results. This study aimed to investigate the feasibility and safety of minimally invasive renal subcapsular transplantation of MSCs for treating AKI in a minipig model, ultimately aiming to facilitate the clinical translation of this approach. METHODS A novel AKI minipig model was established by combining cisplatin with hydration to evaluate the effectiveness of potential therapies. Renal subcapsular catheterization was successfully achieved under ultrasound guidance. Subsequently, the efficacy of renal subcapsular MSCs transplantation was assessed, and the biological role of the tryptophan metabolite kynurenine (Kyn) in AKI was elucidated through both in vivo and in vitro experiments. RESULTS The method of pre-hydration at 4% of body weight, followed by post-cisplatin (3.8 mg/kg) hydration at 2% of body weight, successfully established a cisplatin-induced AKI minipig model with a survival time exceeding 28 days, closely mimicking the clinical characteristics of typical AKI patients. Additionally, we discovered that multiple MSCs transplantations promoted renal function recovery more effectively than single transplantation via the renal subcapsular catheter. Furthermore, elevated levels of Kyn were observed in kidney during AKI, which activated the aryl hydrocarbon receptor (AhR)-mediated NF-κB/NLRP3/IL-1β signaling pathway in tubular epithelial cells, thereby exacerbating inflammatory injury. CONCLUSIONS Ultrasound-guided renal subcapsular transplantation of mesenchymal stem cells is a safe and effective therapeutic approach for AKI, with the potential to bring about significant clinical advancements in the future.
Collapse
Affiliation(s)
- Tuo Xiao
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing, 100853, China
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuhao Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing, 100853, China
| | - Bo Jiang
- Department of Ultrasound, First Medical Centre of Chinese PLA General Hospital, Beijing, 100853, China
| | - Mengjie Huang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing, 100853, China
| | - Yanjun Liang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing, 100853, China
| | - Yue Xu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing, 100853, China
| | - Xumin Zheng
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing, 100853, China
| | - Wenjuan Wang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing, 100853, China
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing, 100853, China.
| | - Guangyan Cai
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing, 100853, China.
| |
Collapse
|
47
|
Bibi A, Zhang F, Shen J, Din AU, Xu Y. Behavioral alterations in antibiotic-treated mice associated with gut microbiota dysbiosis: insights from 16S rRNA and metabolomics. Front Neurosci 2025; 19:1478304. [PMID: 40092066 PMCID: PMC11906700 DOI: 10.3389/fnins.2025.1478304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
The gut and brain interact through various metabolic and signaling pathways, each of which influences mental health. Gut dysbiosis caused by antibiotics is a well-known phenomenon that has serious implications for gut microbiota-brain interactions. Although antibiotics disrupt the gut microbiota's fundamental structure, the mechanisms that modulate the response and their impact on brain function are still unclear. It is imperative to comprehend and investigate crucial regulators and factors that play important roles. We aimed to study the effect of long-term antibiotic-induced disruption of gut microbiota, host metabolomes, and brain function and, particularly, to determine the basic interactions between them by treating the C57BL/6 mice with two different, most commonly used antibiotics, ciprofloxacin and amoxicillin. Anxiety-like behavior was confirmed by the elevated plus-maze test and open field test. Gut microbes and their metabolite profiles in fecal, serum, and brain samples were determined by 16S rRNA sequencing and untargeted metabolomics. In our study, long-term antibiotic treatment exerted anxiety-like effects. The fecal microbiota and metabolite status revealed that the top five genera found were Lactobacillus, Bacteroides, Akkermansia, Ruminococcus_gnavus_group, and unclassified norank_f_Muribaculaceae. The concentration of serotonin, L-Tyrosine, 5-Hydroxy-L-tryptophan, L-Glutamic acid, L-Glutamate, 5-Hydroxyindole acetic acid, and dopaminergic synapsis was comparatively low, while adenosine was high in antibiotic-treated mice. The KEGG enrichment analysis of serum and brain samples showed that amino acid metabolism pathways, such as tryptophan metabolism, threonine metabolism, serotonergic synapsis, methionine metabolism, and neuroactive ligand-receptor interaction, were significantly decreased in antibiotic-treated mice. Our study demonstrates that long-term antibiotic use induces gut dysbiosis and alters metabolic responses, leading to the dysregulation of brain signaling molecules and anxiety-like behavior. These findings highlight the complex interactions between gut microbiota and metabolic functions, providing new insights into the influence of microbial communities on gut-brain communication.
Collapse
Affiliation(s)
- Asma Bibi
- The Key Laboratory of Microbiology and Parasitology Anhui, School of Basic Medical Sciences, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Clinical Laboratory Diagnostics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Famin Zhang
- The Key Laboratory of Microbiology and Parasitology Anhui, School of Basic Medical Sciences, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Clinical Laboratory Diagnostics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jilong Shen
- The Key Laboratory of Microbiology and Parasitology Anhui, School of Basic Medical Sciences, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Clinical Laboratory Diagnostics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ahmad Ud Din
- Department of Food, Bioprocessing and Nutrition Sciences, Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, United States
| | - Yuanhong Xu
- The Key Laboratory of Microbiology and Parasitology Anhui, School of Basic Medical Sciences, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Clinical Laboratory Diagnostics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
48
|
Xu S, Liu ZL, Zhang TW, Li B, Cao YC, Wang XN, Jiao W. Self-control study of multi-omics in identification of microenvironment characteristics in calcium oxalate kidney stones. BMC Nephrol 2025; 26:104. [PMID: 40016672 PMCID: PMC11869433 DOI: 10.1186/s12882-025-04026-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Perform proteomic and metabolomic analysis on bilateral renal pelvis urine of patients with unilateral calcium oxalate kidney stones to identify the specific urinary microenvironment associated with stone formation. METHODS Using cystoscopy-guided insertion of ureteral catheters, bilateral renal pelvis urine samples are collected. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) is employed to identify differential proteins and metabolites in the urine microenvironment. Differentially expressed proteins and differential metabolites are further analyzed for their biological functions and potential metabolic pathways through Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, Reactome pathway analysis and Biomolecular Interaction Network Database protein-protein interaction (PPI) network analysis. RESULTS In the urine from the stone-affected side, 36 differential proteins were significantly upregulated, 4 differential proteins were downregulated, and 10 differential metabolites were significantly upregulated. Functional and pathway analyses indicate that the differentially expressed proteins are primarily involved in inflammatory pathways and complement and coagulation cascades, while the differential metabolites are mainly associated with oxidative stress. CONCLUSION The proteomic and metabolomic profiles of the urinary microenvironment in stone-affected kidneys provide a more precise reflection of the pathophysiological mechanisms involved in stone formation and development.
Collapse
Affiliation(s)
- Shang Xu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266000, China
| | - Zhi-Long Liu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266000, China
| | - Tian-Wei Zhang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266000, China
| | - Bin Li
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266000, China
| | - Yuan-Chao Cao
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266000, China
| | - Xin-Ning Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266000, China.
| | - Wei Jiao
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266000, China.
| |
Collapse
|
49
|
Coyle S, Chapman E, Hughes DM, Baker J, Slater R, Davison AS, Norman BP, Roberts I, Nwosu AC, Gallagher JA, Ranganath LR, Boyd MT, Mayland CR, Kell DB, Mason S, Ellershaw J, Probert C. Urinary metabolite model to predict the dying process in lung cancer patients. COMMUNICATIONS MEDICINE 2025; 5:49. [PMID: 40016594 PMCID: PMC11868640 DOI: 10.1038/s43856-025-00764-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 02/07/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Accurately recognizing that a person may be dying is central to improving their experience of care at the end-of-life. However, predicting dying is frequently inaccurate and often occurs only hours or a few days before death. METHODS We performed urinary metabolomics analysis on patients with lung cancer to create a metabolite model to predict dying over the last 30 days of life. RESULTS Here we show a model, using only 7 metabolites, has excellent accuracy in the Training cohort n = 112 (AUC = 0·85, 0·85, 0·88 and 0·86 on days 5, 10, 20 and 30) and Validation cohort n = 49 (AUC = 0·86, 0·83, 0·90, 0·86 on days 5, 10, 20 and 30). These results are more accurate than existing validated prognostic tools, and uniquely give accurate predictions over a range of time points in the last 30 days of life. Additionally, we present changes in 125 metabolites during the final four weeks of life, with the majority exhibiting statistically significant changes within the last week before death. CONCLUSIONS These metabolites identified offer insights into previously undocumented pathways involved in or affected by the dying process. They not only imply cancer's influence on the body but also illustrate the dying process. Given the similar dying trajectory observed in individuals with cancer, our findings likely apply to other cancer types. Prognostic tests, based on the metabolites we identified, could aid clinicians in the early recognition of people who may be dying and thereby influence clinical practice and improve the care of dying patients.
Collapse
Affiliation(s)
- Séamus Coyle
- Liverpool Head and Neck Cancer Centre, University of Liverpool, Liverpool, UK.
- Department of Palliative Medicine, Clatterbridge Cancer Centre, Liverpool, UK.
| | - Elinor Chapman
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - David M Hughes
- Department of Health Data Science, University of Liverpool, Liverpool, UK
| | - James Baker
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Rachael Slater
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Andrew S Davison
- Department of Clinical Biochemistry and Metabolic Medicine, Liverpool Clinical Laboratories, Liverpool University Hospitals Foundation Trust, Liverpool, UK
- Institute of Life Course & Medical Sciences, University of Liverpool, Liverpool, UK
| | - Brendan P Norman
- Institute of Life Course & Medical Sciences, University of Liverpool, Liverpool, UK
| | - Ivayla Roberts
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Centre for Metabolomics Research (CMR), Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Amara C Nwosu
- Lancaster Medical School, Lancaster University, Lancaster, UK
- Marie Curie Hospice, Liverpool, UK
- Palliative Care Unit, Institute of Life Course & Medical Sciences, University of Liverpool, Liverpool, UK
| | - James A Gallagher
- Institute of Life Course & Medical Sciences, University of Liverpool, Liverpool, UK
| | - Lakshminarayan R Ranganath
- Department of Clinical Biochemistry and Metabolic Medicine, Liverpool Clinical Laboratories, Liverpool University Hospitals Foundation Trust, Liverpool, UK
- Centre for Metabolomics Research (CMR), Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Mark T Boyd
- Liverpool Head and Neck Cancer Centre, University of Liverpool, Liverpool, UK
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Catriona R Mayland
- Palliative Care Unit, Institute of Life Course & Medical Sciences, University of Liverpool, Liverpool, UK
- Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
| | - Douglas B Kell
- Centre for Metabolomics Research (CMR), Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Novo Nordisk Foundation Centre for Biosustainability, Danish Technical University of Denmark, Lyngby, Denmark
| | - Stephen Mason
- Palliative Care Unit, Institute of Life Course & Medical Sciences, University of Liverpool, Liverpool, UK
| | - John Ellershaw
- Palliative Care Unit, Institute of Life Course & Medical Sciences, University of Liverpool, Liverpool, UK
- Academic Palliative & End of Life Care Department, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Chris Probert
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| |
Collapse
|
50
|
Bertollo AG, Mingoti MED, Ignácio ZM. Neurobiological mechanisms in the kynurenine pathway and major depressive disorder. Rev Neurosci 2025; 36:169-187. [PMID: 39245854 DOI: 10.1515/revneuro-2024-0065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/20/2024] [Indexed: 09/10/2024]
Abstract
Major depressive disorder (MDD) is a prevalent psychiatric disorder that has damage to people's quality of life. Tryptophan is the precursor to serotonin, a critical neurotransmitter in mood modulation. In mammals, most free tryptophan is degraded by the kynurenine pathway (KP), resulting in a range of metabolites involved in inflammation, immune response, and neurotransmission. The imbalance between quinolinic acid (QA), a toxic metabolite, and kynurenic acid (KynA), a protective metabolite, is a relevant phenomenon involved in the pathophysiology of MDD. Proinflammatory cytokines increase the activity of the enzyme indoleamine 2,3-dioxygenase (IDO), leading to the degradation of tryptophan in the KP and an increase in the release of QA. IDO activates proinflammatory genes, potentiating neuroinflammation and deregulating other physiological mechanisms related to chronic stress and MDD. This review highlights the physiological mechanisms involved with stress and MDD, which are underlying an imbalance of the KP and discuss potential therapeutic targets.
Collapse
Affiliation(s)
- Amanda Gollo Bertollo
- Laboratory of Physiology, Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Maiqueli Eduarda Dama Mingoti
- Laboratory of Physiology, Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Zuleide Maria Ignácio
- Laboratory of Physiology, Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| |
Collapse
|