1
|
Sobolev PD, Burnakova NA, Revelsky AI, Zakharchenko VE, Beloborodova NV, Pautova AK. A sensitive method for the profiling of phenyl- and indole-containing metabolites in blood serum and cerebrospinal fluid samples of patients with severe brain damage using ultra-high-pressure liquid chromatography-tandem mass spectrometry. J Pharm Biomed Anal 2025; 260:116803. [PMID: 40086051 DOI: 10.1016/j.jpba.2025.116803] [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: 12/06/2024] [Revised: 02/19/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
The metabolic profiling of biological fluids is important in understanding the various biochemical processes in the human body. The content of aromatic metabolites, including microbial ones, in the blood and cerebrospinal fluid, can provide essential information reflecting infectious processes, both systemic and in the central nervous system. A sensitive method with protein precipitation and sample concentration using ultra-high-pressure liquid chromatography-tandem mass spectrometry was proposed and subsequently validated to determine the number of aromatic metabolites of phenylalanine, tyrosine, and tryptophan in the blood serum and cerebrospinal fluid at 2.0-3.7 × 103 nmol/L. Reference values of 4-hydroxybenzoic, 3-(4-hydroxyphenyl)propionic, and indole-3-carboxylic acids were measured in the blood serum of healthy donors (n = 48). Profile of eleven phenyl- and indole-containing acids was revealed in the serum samples (n = 29) of the patients with long-term sequelae of severe brain damage and in the cerebrospinal fluid samples (n = 29) of the post-neurosurgical patients using different sample preparation methods to measure analytes in a wide (nmol/L and μmol/L) concentration range. Statistically significant differences in concentrations of most analytes were detected in serum samples of patients compared to healthy donors (p ≤ 0.03) and in concentrations of 3-phenyllactic, 3-(4-hydroxyphenyl)lactic, indole-3-lactic, and indole-3-carboxylic acids in cerebrospinal fluid samples of patients with signs of secondary bacterial meningitis compared to those without signs of secondary bacterial meningitis (p ≤ 0.027).
Collapse
Affiliation(s)
- Pavel D Sobolev
- Exacte Labs Bioanalytical Laboratory, 20-2 Nauchny proezd, Moscow 117246, Russia
| | - Natalia A Burnakova
- Exacte Labs Bioanalytical Laboratory, 20-2 Nauchny proezd, Moscow 117246, Russia
| | - Alexander I Revelsky
- Lomonosov Moscow State University, GSP-1, Leninskie gory, 1-3, Moscow 119991, Russia
| | - Vladislav E Zakharchenko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Petrovka str., 25-2, Moscow 107031, Russia
| | - Natalia V Beloborodova
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Petrovka str., 25-2, Moscow 107031, Russia
| | - Alisa K Pautova
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Petrovka str., 25-2, Moscow 107031, Russia.
| |
Collapse
|
2
|
Kim HJ, Kim HJ, Hong H, Choi M, Ismail A, Mun D, Kim Y, Kim GD, Jo C. Utilizing drip metabolites and predictive modeling for non-destructive freshness assessment in pork loin. NPJ Sci Food 2025; 9:55. [PMID: 40268971 PMCID: PMC12019311 DOI: 10.1038/s41538-025-00421-y] [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: 11/06/2024] [Accepted: 04/10/2025] [Indexed: 04/25/2025] Open
Abstract
This study validated the use of pork drip metabolites for non-destructive freshness prediction. The pork loin was vacuum-packaged and stored for 27 days at 4 °C. The pH, drip loss, total aerobic bacterial counts (TAB), microbial composition and drip metabolites were examined. LASSO and Random Forest (RF) were selected and used for variable selection, while Ridge regression and Support Vector Regression were utilized to develop predictive models. Validation was performed using leave-one-out cross-validation. LASSO and RF selected 13 and 10 metabolites, respectively. The metabolites selected by each method were trained using Ridge regression and SVR. Each of the four trained models achieved R2 values of over 0.9. In the validation step, the model trained by Ridge regression using drip metabolites selected through LASSO showed the lowest RMSE value of 0.283 log CFU/g. Therefore, selected drip metabolites can be used to predict TAB and microbial composition of pork loin through mathematical modeling.
Collapse
Affiliation(s)
- Hyun-Jun Kim
- Institutes of Green Bio Science and Technology, Seoul National University, Pyeongchang, 25354, Republic of Korea
| | - Hye-Jin Kim
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Heesang Hong
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Minwoo Choi
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Azfar Ismail
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Daye Mun
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Younghoon Kim
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Gap-Don Kim
- Institutes of Green Bio Science and Technology, Seoul National University, Pyeongchang, 25354, Republic of Korea
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, 25354, Republic of Korea
| | - Cheorun Jo
- Institutes of Green Bio Science and Technology, Seoul National University, Pyeongchang, 25354, Republic of Korea.
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
3
|
Wang T, Su Z, Zhong M, Wu X, Li L, Gu H, Sun Y, Ji J, Peng X. A Metabolic Signature Specific to the Patients with Type 2 Diabetes and its Association with the Pathogenesis of Diabetic-Foot Syndrome. J Cardiovasc Transl Res 2025:10.1007/s12265-025-10622-1. [PMID: 40237961 DOI: 10.1007/s12265-025-10622-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/10/2025] [Indexed: 04/18/2025]
Abstract
Oxidative stress and protein nonenzymatic glycation are key factors in diabetic-foot syndrome pathogenesis. Type 2 diabetes (T2DM) progression involves excessive gluconolactone (GDL) production, linked to endothelial injury and diabetic arteriosclerosis. This study explored GDL's role in diabetic-foot syndrome using high-performance liquid chromatography-tandem mass spectrometry to analyze sera from 75 T2DM patients (including 32 with diabetic-foot) and 36 healthy controls. GDL levels were significantly higher in T2DM patients and correlated with increased hemoglobin A1c glycation and reactive oxygen species production in endothelial cells, suggesting GDL's role in accelerating macrovascular arteriosclerosis and diabetic-foot syndrome. These findings highlight GDL's potential as a diagnostic biomarker and therapeutic target for diabetic macrovascular complications.
Collapse
Affiliation(s)
- Tao Wang
- Department of Cardiovascular Surgery, Shenzhen Guangming District People's Hospital, Songbai Road 4253, Guangming District, Shenzhen, 518027, P. R. China
| | - Ziyu Su
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shanxi, China
- Bio-Manufacturing Engineering Laboratory, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518000, Guangdong, China
| | - Ming Zhong
- Department of Cardiovascular Surgery, Shenzhen Guangming District People's Hospital, Songbai Road 4253, Guangming District, Shenzhen, 518027, P. R. China
| | - Xuanqin Wu
- Department of Cardiovascular Surgery, Shenzhen Guangming District People's Hospital, Songbai Road 4253, Guangming District, Shenzhen, 518027, P. R. China
| | - Liang Li
- Department of Cardiovascular Surgery, Shenzhen Guangming District People's Hospital, Songbai Road 4253, Guangming District, Shenzhen, 518027, P. R. China
| | - Hong Gu
- Department of Cardiovascular Surgery, Shenzhen Guangming District People's Hospital, Songbai Road 4253, Guangming District, Shenzhen, 518027, P. R. China
| | - Yunhan Sun
- The High School Affiliated to Renmin University of China, Shenzhen, 518000, China
| | - Jun Ji
- Department of Cardiovascular Surgery, Shenzhen Guangming District People's Hospital, Songbai Road 4253, Guangming District, Shenzhen, 518027, P. R. China.
| | - Xingchun Peng
- Department of Pathology, Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Lanjinsi Road 15, Pingshan District, Shenzhen, 518118, P. R. China.
| |
Collapse
|
4
|
Feng A, Zhao H, Qiu C, Luo D, Wu H, Meng X, Li L, Zou H. Gut microbiota metabolites impact immunologic responses to antiretroviral therapy in HIV-infected men who have sex with men. Infect Dis Poverty 2025; 14:21. [PMID: 40098016 PMCID: PMC11917012 DOI: 10.1186/s40249-025-01291-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] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND The association between gut microbial metabolites and immunologic non-response among people living with HIV (PLHIV) receiving antiretroviral therapy (ART) has not been well established. We aimed to characterize gut microbial metabolites among HIV-infected men who have sex with men (MSM) with different immunologic responses. METHODS We recruited HIV-infected MSM from Guangzhou Eighth People's Hospital and HIV-uninfected MSM (healthy controls, HC) from a local MSM community-based organization in Guangzhou between June and October 2021. HIV-infected MSM were grouped into good immunological responders (GIR) (CD4 + T cell count ≥ 350 cells/μl) and poor immunological responders (PIR) (CD4 + T cell count < 350 cells/μl) after 24 months of ART treatment. Online questionnaires and stool samples were collected. Microbial metabolites in stool were obtained through ultra-performance liquid chromatography coupled to a tandem mass spectrometry (UPLC-MS/MS) system. Differential metabolites were identified and analyzed using the Kruskal-Wallis test, followed by pairwise comparisons with the Wilcoxon rank-sum test. The least absolute selection and shrinkage operator was used to select potential metabolites biomarkers. RESULTS A total of 51 HC, 56 GIR, and 42 PIR were included. No statistically significant differences were observed in the median time since HIV diagnosis and ART duration between GIR and PIR. Among the 174 quantified metabolites, 81 significantly differed among HC, GIR, and PIR (P < 0.05). Among differential metabolites, indole-3-propionic acid significantly decreased from HC (11.39 nmol/g) and GIR (8.16 nmol/g) to PIR (6.50 nmol/g). The pathway analysis showed that tryptophan metabolism differed significantly between GIR and PIR (P < 0.05). Four potential metabolites biomarkers (dimethylglycine, cinnamic acid, 3-hydroxyisovaleric acid, and propionic acid) that distinguish GIR and PIR were identified, and the corresponding area under the curve based on potential biomarkers was 0.773 (95% CI: 0.675-0.871). CONCLUSIONS This study identified significant differences in gut microbial metabolites among HIV-infected MSM with different immunologic responses. These results indicate the potential of gut microbial metabolites as novel disease progression markers and therapeutic targets.
Collapse
Affiliation(s)
- Anping Feng
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
| | - Heping Zhao
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
- Infectious Disease Center, Guangzhou Eighth People's Hospital, Guangzhou Medical University, No 8 Huaying Road, Guangzhou, 510060, Guangdong, China
| | - Chunting Qiu
- Department of Infectious Diseases, Tianjin Second People's Hospital, Tianjin, 300192, China
| | - Dan Luo
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
| | - Hao Wu
- Guangzhou Center for Disease Control and Prevention, Guangzhou, China
| | - Xiaojun Meng
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi, 214023, Jiangsu, China.
| | - Linghua Li
- Infectious Disease Center, Guangzhou Eighth People's Hospital, Guangzhou Medical University, No 8 Huaying Road, Guangzhou, 510060, Guangdong, China.
| | - Huachun Zou
- School of Public Health, Fudan University, Room 435, Bld #8, 130 Dongan Road, Xuhui District, Shanghai, 200032, China.
- School of Public Health, Southwest Medical University, Luzhou, China.
- Kirby Institute, University of New South Wales, Sydney, Australia.
| |
Collapse
|
5
|
Xuan X, Huang Z, Kong Z, Li R, Li J, Huang H. GENETIC INSIGHTS INTO SEPSIS: MENDELIAN RANDOMIZATION ANALYSIS OF CEREBROSPINAL FLUID METABOLITES. Shock 2025; 63:379-384. [PMID: 39454631 DOI: 10.1097/shk.0000000000002494] [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: 10/28/2024]
Abstract
ABSTRACT Background: Sepsis, a life-threatening response to infection leading to systemic inflammation and organ dysfunction, has been hypothesized to be influenced by metabolic alterations in cerebrospinal fluid (CSF). Despite extensive research, the specific metabolic pathways contributing to sepsis remain unclear. This study aims to elucidate the causal relationships between CSF metabolites and sepsis risk using Mendelian randomization (MR), offering insights that could lead to novel therapeutic strategies. Methods: We conducted a two-sample MR analysis using genetic variants as instrumental variables (IVs) to investigate 338 CSF metabolites identified through a genome-wide association study. Data on sepsis-related outcomes were extracted from the genome-wide association study catalog encompassing 486,484 individuals of European descent. IVs were rigorously selected based on stringent genetic association and linkage disequilibrium criteria. Statistical analyses, including inverse variance weighting (IVW) and weighted median methods, were performed using the "TwoSampleMR" package in R software, supplemented by comprehensive sensitivity analyses to ensure the robustness of our findings. Results: Our analysis identified 19 CSF metabolites causally associated with sepsis risk. Notably, metabolites such as 1-palmitoyl-2-stearoyl-gpc (16:0/18:0) and 2-hydroxyglutarate showed significant negative correlations with sepsis risk. The reverse MR analysis further revealed that sepsis could negatively impact certain CSF metabolite levels, particularly ribonate, suggesting a bidirectional relationship. These relationships were substantiated by rigorous statistical testing and sensitivity analyses confirming the absence of horizontal pleiotropy and the stability of our results across various MR methods. Conclusions: This study demonstrates significant causal associations between specific CSF metabolites and the risk of developing sepsis, highlighting the potential for these metabolites to serve as biomarkers or therapeutic targets. The bidirectional nature of these findings also suggests that sepsis itself may alter metabolic profiles, offering further avenues for intervention.
Collapse
Affiliation(s)
- Xin Xuan
- Department of Emergency Medicine, Dongguan Hospital of Guangzhou University of Traditional Chinese Medicine, Dongguan, Guangdong, China
| | - Zhihao Huang
- Department of Otorhinolaryngology, Dongguan Hospital of Guangzhou University of Traditional Chinese Medicine, Dongguan, Guangdong, China
| | - Zhiqian Kong
- Department of Emergency Medicine, Dongguan Hospital of Guangzhou University of Traditional Chinese Medicine, Dongguan, Guangdong, China
| | - Ruoyu Li
- Department of Intensive Care Unit, Dongguan Hospital of Guangzhou University of Traditional Chinese Medicine, Dongguan, Guangdong, China
| | - Jianfeng Li
- Department of Emergency Medicine, Dongguan Hospital of Guangzhou University of Traditional Chinese Medicine, Dongguan, Guangdong, China
| | - Haiyan Huang
- Department of Internal Medicine of Chinese Medicine, Dongguan Hospital of Guangzhou University of Traditional Chinese Medicine, Dongguan, Guangdong, China
| |
Collapse
|
6
|
Zubkowski A, Sferruzzi‐Perri AN, Wishart DS. Mechanisms of Homoarginine: Looking Beyond Clinical Outcomes. Acta Physiol (Oxf) 2025; 241:e14273. [PMID: 39817883 PMCID: PMC11737358 DOI: 10.1111/apha.14273] [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: 04/02/2024] [Revised: 10/31/2024] [Accepted: 01/01/2025] [Indexed: 01/18/2025]
Abstract
PURPOSE Homoarginine (hArg) is an arginine metabolite that has been known for years, but its physiological role in the body remains poorly understood. For instance, it is well known that high hArg concentrations in the blood are protective against several disease states, yet the mechanisms behind these health benefits are unclear. This review compiles what is known about hArg, namely its synthetic pathways, its role in different diseases and conditions, and its proposed mechanisms of action in humans and experimental animals. FINDINGS Previous work has identified multiple pathways that control hArg synthesis and degradation in the body. Furthermore, endogenous hArg can modulate the cardiovascular system, with decreased hArg being associated with cardiovascular complications and increased mortality. Studies also suggest that hArg could serve as a diagnostic biomarker for a variety of immune, pancreatic, renal, and hepatic dysfunctions. Finally, in women, hArg concentrations rapidly increase throughout pregnancy and there are suggestions that alterations in hArg could indicate pregnancy complications like pre-eclampsia. SUMMARY Homoarginine is an under-appreciated amino acid with potential wide-ranging roles in systemic health, pregnancy, and pathophysiology. Although recent research has focused on its health or disease associations, there is a need for more investigations into understanding the mechanistic pathways by which hArg may operate. This could be aided using metabolomics, which provides a comprehensive approach to correlating multiple metabolites and metabolic pathways with physiological effects. Increasing our knowledge of hArg's roles in the body could pave the way for its routine use as both a diagnostic and therapeutic molecule.
Collapse
Affiliation(s)
- Ashley Zubkowski
- Department of Biological SciencesUniversity of AlbertaEdmontonAlbertaCanada
| | - Amanda N. Sferruzzi‐Perri
- Centre for Trophoblast Research, Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - David S. Wishart
- Department of Biological SciencesUniversity of AlbertaEdmontonAlbertaCanada
- Department of Computer SciencesUniversity of AlbertaEdmontonAlbertaCanada
| |
Collapse
|
7
|
Tu X, Chen J, Liu W. Development and internal validation of a metabolism-related model for predicting 30-day mortality in neonatal sepsis. BMC Infect Dis 2025; 25:121. [PMID: 39871138 PMCID: PMC11771113 DOI: 10.1186/s12879-025-10527-z] [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: 12/03/2024] [Accepted: 01/21/2025] [Indexed: 01/29/2025] Open
Abstract
OBJECTIVE Neonatal sepsis, a severe infectious disease associated with high mortality rates, is characterized by metabolic disturbances that play a crucial role in its progression. The aim of this study is to develop a metabolism-related model for assessing 30-day mortality in neonatal sepsis. METHODS The clinical data of neonatal sepsis at Ganzhou Women and Children's Health Care Hospital from January 2019 to December 2022 were retrospectively analyzed. Neonatal sepsis cases were divided into survival and non-survival groups. Multivariate logistic regression analysis was used to identify the independent risk factors for 30-day mortality. A nomogram model was developed based on these risk factors. Internal validation of the model was performed using 10-fold cross-validation. The predictive performance was evaluated through receiver operating characteristic (ROC) curves and calibration curve analyses. Decision curve analysis (DCA) was conducted to evaluate the clinical applicability of the developed model. RESULTS The study included a total of 156 cases of neonatal sepsis. Multivariate logistic regression analysis revealed that alanine(ALA), citrulline(CIT)), octadecanoylcarnitine(C18) and methionine(MET) were identified as independent risk factors for 30-day mortality of neonatal sepsis. The ROC curve showed an area under the curve of AUC = 0.866 (95% CI 0.796-0.936, P < 0.05). The calibration curve and DCA indicated excellent performance of the model. CONCLUSION This study establishes a predictive model for neonatal sepsis-associated 30-day mortality, effectively capturing the perturbations in amino acid metabolism and fatty acid oxidation, thereby demonstrating robust predictive capabilities.
Collapse
Affiliation(s)
- Xiangwen Tu
- Laboratory of Eugenics Genetics, GanZhou Women and Children's Health Care Hospital, GanZhou, Jiangxi, China
| | - Junkun Chen
- Laboratory of Eugenics Genetics, GanZhou Women and Children's Health Care Hospital, GanZhou, Jiangxi, China
| | - Wen Liu
- Neonatal intensive care Unit, GanZhou Women and Children's Health Care Hospital, GanZhou, Jiangxi, China.
| |
Collapse
|
8
|
Li K, Liu P, Han L, Tian J, Zheng Z, Sha M, Ye J, Zhu L. Elucidating ferroptosis mechanisms in heart failure through transcriptomics, single-cell sequencing, and experimental validation. Cell Signal 2024; 124:111416. [PMID: 39293745 DOI: 10.1016/j.cellsig.2024.111416] [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: 07/22/2024] [Revised: 08/30/2024] [Accepted: 09/15/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND The mechanisms underlying ferroptosis in heart failure (HF) remain incompletely understood. METHODS This study analyzed the heart failure dataset from the Gene Expression Omnibus to identify differentially expressed ferroptosis-related genes (DFRGs). Key DFRGs were selected using LASSO regression and SVM-RFE machine learning techniques. Their diagnostic accuracy was evaluated via ROC curve analysis. Single-cell sequencing data, heart failure cell, and mouse models were utilized to validate these key DFRGs. Additionally, potential non-coding RNAs targeting these genes were predicted, and analyses for gene set enrichment, immune cell infiltration, and drug targeting were conducted. RESULTS A total of 127 DFRGs were identified, with 83 downregulated and 44 upregulated compared to controls. Seven key DFRGs (PTGS2, BECN1, SLC39A14, QSOX1, MLST8, TMSB4X, KDM4A) were identified, showing high diagnostic accuracy (AUC 0.988) in the GSE5406 dataset. GO and KEGG analyses linked these genes to ferroptosis, FoxO signaling, and autophagy pathways. A ceRNA network identified 217 miRNAs and 243 lncRNAs potentially targeting these genes, and 64 drugs were predicted as potential targets. Single-cell sequencing and in vitro experiments revealed differential expression of SLC39A14 and QSOX1, which was further confirmed in vivo. CONCLUSION This study provides novel insights into the role of ferroptosis in heart failure by identifying and validating DFRGs that exhibit differential expression across various cell types. The differential expression patterns of these genes, particularly SLC39A14 and QSOX1, indicate their potential involvement in the pathophysiological mechanisms contributing to HF. These findings offer new insights for the development of targeted therapies for HF.
Collapse
Affiliation(s)
- Kaiyuan Li
- Graduate School of Dalian Medical University, Dalian Medical University, Dalian, Liaoning 116000, PR China; Department of Cardiology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu 225300, PR China
| | - Peng Liu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, PR China
| | - Lingyu Han
- Department of Cardiology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu 225300, PR China
| | - Jing Tian
- Department of Cardiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250000, PR China
| | - Zhipeng Zheng
- Graduate School of Dalian Medical University, Dalian Medical University, Dalian, Liaoning 116000, PR China; Department of Cardiology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu 225300, PR China
| | - Min Sha
- Department of Cardiology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu 225300, PR China
| | - Jun Ye
- Graduate School of Dalian Medical University, Dalian Medical University, Dalian, Liaoning 116000, PR China; Department of Cardiology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu 225300, PR China.
| | - Li Zhu
- Graduate School of Dalian Medical University, Dalian Medical University, Dalian, Liaoning 116000, PR China; Department of Cardiology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu 225300, PR China.
| |
Collapse
|
9
|
Tan X, Zhang X, Chai J, Ji W, Ru J, Yang C, Zhou W, Bai J, Xiong Y. Constructing a predictive model for early-onset sepsis in neonatal intensive care unit newborns based on SHapley Additive exPlanations explainable machine learning. Transl Pediatr 2024; 13:1933-1946. [PMID: 39649648 PMCID: PMC11621883 DOI: 10.21037/tp-24-278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/05/2024] [Indexed: 12/11/2024] Open
Abstract
Background The clinical characteristics of neonatal sepsis (NS) are subtle and non-specific, posing a serious threat to the lives of newborn infants. Early-onset sepsis (EOS) is sepsis that occurs within 72 hours after birth, with a high mortality rate. Identifying key factors of NS and conducting early diagnosis are of great practical significance. Thus, we developed a robust machine learning (ML) model for the early prediction of EOS in neonates admitted to the neonatal intensive care unit (NICU), investigated the pivotal risk factors associated with EOS development, and provided interpretable insights into the model's predictions. Methods A retrospective cohort study was conducted. This includes 668 newborns (EOS and non-EOS) admitted to the NICU of Bozhou People's Hospital from January to December 2023, excluding 72 newborns born more than three days ago and 166 newborns with medical record data missing more than 30%. Finally, 430 newborns (EOS and non-EOS) were included in the study. Clinical case data were meticulously analyzed, and the dataset was randomly partitioned, allocating 75% for model training and the remaining 25% for test. Data preprocessing was meticulously performed using R language, and the least absolute shrinkage and selection operator (LASSO) regression was implemented to select salient features, mitigating the risk of overfitting. Six ML models were leveraged to forecast the incidence of EOS in neonates. The predictive performance of these models was rigorously evaluated using the receiver operating characteristic (ROC) curve and precision-recall (PR) curve. Furthermore, the SHapley Additive exPlanations (SHAP) framework was employed to provide intuitive explanations for the predictions made by the Categorical Boosting (CatBoost) model, which emerged as the top performer. Results The ROC area under the curve (ROCAUC) of six ML models, CatBoost, random forest (RF), eXtreme Gradient Boosting (XGBoost), multilayer perceptron (MLP), support vector machine (SVM), logistic regression (LR) all exceeded 0.900 on the test set. Especially the CatBoost model exhibited superior performance, with favorable outcomes in calibration, decision curve analysis (DCA), and learning curves. Notably, the ROCAUC attained 0.975, and the area under the PR curve (PRAUC) reached 0.947, signifying a high degree of predictive accuracy. Utilizing the SHAP method, seven key features were identified and ranked by their importance: respiratory rate (RR), procalcitonin (PCT), nasal congestion (NC), yellow staining (YS), white blood cell count (WBC), fever, and amniotic fluid turbidity (AFT). Conclusions By constructing a precision-oriented ML model and harnessing the SHAP method for interpretability, this study effectively identified crucial risk factors for EOS development in neonates. This approach enables early prediction of EOS risk, thereby facilitating timely and targeted clinical interventions for precise diagnosis and treatment.
Collapse
Affiliation(s)
- Xuefeng Tan
- Department of Laboratory Medicine, The People’s Hospital, Bozhou, China
| | - Xiufang Zhang
- Department of Laboratory Medicine, The People’s Hospital, Bozhou, China
| | - Jie Chai
- Department of Laboratory Medicine, The People’s Hospital, Bozhou, China
| | - Wenjuan Ji
- Department of Laboratory Medicine, The People’s Hospital, Bozhou, China
| | - Jinling Ru
- Department of Laboratory Medicine, The People’s Hospital, Bozhou, China
| | - Cuilin Yang
- Department of Laboratory Medicine, The People’s Hospital, Bozhou, China
| | - Wenjing Zhou
- Department of Laboratory Medicine, The People’s Hospital, Bozhou, China
| | - Jing Bai
- Department of Laboratory Medicine, The People’s Hospital, Bozhou, China
| | - Yueling Xiong
- Translational Medicine Center, The Second Affiliated Hospital, Wannan Medical College, Wuhu, China
| |
Collapse
|
10
|
Wan L, Shi X, Yan H, Liang Y, Liu X, Zhu G, Zhang J, Wang J, Wang M, Yang G. Abnormalities in Clostridioides and related metabolites before ACTH treatment may be associated with its efficacy in patients with infantile epileptic spasm syndrome. CNS Neurosci Ther 2024; 30:e14398. [PMID: 37553527 PMCID: PMC10805391 DOI: 10.1111/cns.14398] [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: 05/08/2023] [Revised: 07/10/2023] [Accepted: 07/27/2023] [Indexed: 08/10/2023] Open
Abstract
OBJECTIVE Adrenocorticotropic hormone (ACTH) is the first-line treatment of infantile epileptic spasm syndrome (IESS). Its reported effectiveness varies, and our current understanding regarding the role of gut microbiota composition in IESS treatment response is limited. This study assessed the microbiome-metabolome association to understand the role and mechanism of gut microbiota composition in IESS treatment outcomes. METHODS Children with IESS undergoing ACTH treatment were enrolled. Pre-treatment stool and serum samples were collected for 16S rRNA gene sequencing and liquid chromatography-tandem mass spectrometry, respectively. The children were divided into "responsive" and "non-responsive" groups, and gut microbiota and serum metabolome differences were analyzed. RESULTS Of the 30 patients with IESS, 14 responded to ACTH and 16 did not. The "non-responsive" group had larger maleficent Clostridioides and Peptoclostridium_phage_p630P populations (linear discriminant analysis >2; false discovery rate q < 0.05). Ten metabolites were upregulated (e.g., xanthurenic acid) and 15 were downregulated (e.g., vanillylmandelic acid) (p < 0.05). Association analysis of the gut microbiome and serum metabolome revealed that Clostridioides and Peptoclostridium_phage_p630P2 were positively correlated with linoleic and xanthurenic acids, while Clostridioides was negatively correlated with vanillylmandelic acid (p < 0.05). A classifier using differential gut bacteria and metabolites achieved an area under the receiver operating characteristic curve of 0.906 to distinguish responders from non-responders. CONCLUSION This study found significant differences in pre-treatment gut microbiota and serum metabolome between children with IESS who responded to ACTH and those who did not. Additional exploration may provide valuable information for treatment selection and potential interventions. Our results suggest that varying ACTH responses in patients with IESS may be associated with increased gut Clostridioides bacteria and kynurenine pathway alteration, but additional experiments are needed to verify this association.
Collapse
Affiliation(s)
- Lin Wan
- Senior Department of PediatricsThe Seventh Medical Center of PLA General HospitalBeijingChina
- Department of PediatricsThe First Medical Centre, Chinese PLA General HospitalBeijingChina
- Medical School of Chinese People's Liberation ArmyBeijingChina
| | - Xiuyu Shi
- Senior Department of PediatricsThe Seventh Medical Center of PLA General HospitalBeijingChina
- Department of PediatricsThe First Medical Centre, Chinese PLA General HospitalBeijingChina
- Medical School of Chinese People's Liberation ArmyBeijingChina
- The Second School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| | - Huimin Yan
- Senior Department of PediatricsThe Seventh Medical Center of PLA General HospitalBeijingChina
- Department of PediatricsThe First Medical Centre, Chinese PLA General HospitalBeijingChina
- Medical School of Chinese People's Liberation ArmyBeijingChina
| | - Yan Liang
- Senior Department of PediatricsThe Seventh Medical Center of PLA General HospitalBeijingChina
- Department of PediatricsThe First Medical Centre, Chinese PLA General HospitalBeijingChina
- Medical School of Chinese People's Liberation ArmyBeijingChina
| | - Xinting Liu
- Senior Department of PediatricsThe Seventh Medical Center of PLA General HospitalBeijingChina
- Department of PediatricsThe First Medical Centre, Chinese PLA General HospitalBeijingChina
- Medical School of Chinese People's Liberation ArmyBeijingChina
| | - Gang Zhu
- Senior Department of PediatricsThe Seventh Medical Center of PLA General HospitalBeijingChina
- Department of PediatricsThe First Medical Centre, Chinese PLA General HospitalBeijingChina
- Medical School of Chinese People's Liberation ArmyBeijingChina
| | - Jing Zhang
- Senior Department of PediatricsThe Seventh Medical Center of PLA General HospitalBeijingChina
- Department of PediatricsThe First Medical Centre, Chinese PLA General HospitalBeijingChina
- Medical School of Chinese People's Liberation ArmyBeijingChina
| | - Jing Wang
- Senior Department of PediatricsThe Seventh Medical Center of PLA General HospitalBeijingChina
- Department of PediatricsThe First Medical Centre, Chinese PLA General HospitalBeijingChina
- Medical School of Chinese People's Liberation ArmyBeijingChina
| | - Mingbang Wang
- Microbiome Therapy Center, South China Hospital, Medical School, Shenzhen UniversityShenzhenChina
- Shanghai Key Laboratory of Birth Defects, Division of NeonatologyChildren's Hospital of Fudan University, National Center for Children's HealthShanghaiChina
- Marshall Laboratory of Biomedical EngineeringMedical School, Shenzhen UniversityShenzhenChina
| | - Guang Yang
- Senior Department of PediatricsThe Seventh Medical Center of PLA General HospitalBeijingChina
- Department of PediatricsThe First Medical Centre, Chinese PLA General HospitalBeijingChina
- Medical School of Chinese People's Liberation ArmyBeijingChina
- The Second School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
11
|
Chen W, Zhang P, Zhang X, Xiao T, Zeng J, Guo K, Qiu H, Cheng G, Wang Z, Zhou W, Zeng S, Wang M. Machine learning-causal inference based on multi-omics data reveals the association of altered gut bacteria and bile acid metabolism with neonatal jaundice. Gut Microbes 2024; 16:2388805. [PMID: 39166704 PMCID: PMC11340767 DOI: 10.1080/19490976.2024.2388805] [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: 03/07/2024] [Revised: 07/09/2024] [Accepted: 07/31/2024] [Indexed: 08/23/2024] Open
Abstract
Early identification of neonatal jaundice (NJ) appears to be essential to avoid bilirubin encephalopathy and neurological sequelae. The interaction between gut microbiota and metabolites plays an important role in early life. It is unclear whether the composition of the gut microbiota and metabolites can be used as an early indicator of NJ or to aid clinical decision-making. This study involved a total of 196 neonates and conducted two rounds of "discovery-validation" research on the gut microbiome-metabolome. It utilized methods of machine learning, causal inference, and clinical prediction model evaluation to assess the significance of gut microbiota and metabolites in classifying neonatal jaundice (NJ), as well as the potential causal relationships between corresponding clinical variables and NJ. In the discovery stage, NJ-associated gut microbiota, network modules, and metabolite composition were identified by gut microbiome-metabolome association analysis. The NJ-associated gut microbiota was closely related to bile acid metabolites. By Lasso machine learning assessment, we found that the gut bacteria were associated with abnormal bile acid metabolism. The machine learning-causal inference approach revealed that gut bacteria affected serum total bilirubin and NJ by influencing bile acid metabolism. NJ-associated gut bile acids are potential biomarkers of NJ, and clinical prediction models constructed based on these biomarkers have some clinical effects and the model may be used for disease risk prediction. In the validation stage, it was found that intestinal metabolites can predict NJ, and the machine learning-causal inference approach revealed that bile acid metabolites affected NJ itself by affecting the total bilirubin content. Intestinal bile acid metabolites are potential biomarkers of NJ. By applying machine learning-causal inference methods to gut microbiome-metabolome association studies, we found NJ-associated intestinal bacteria and their network modules and bile acid metabolite composition. The important role of intestinal bacteria and bile acid metabolites in NJ was determined, which can predict the risk of NJ.
Collapse
Affiliation(s)
- Wanling Chen
- Division of Neonatology, Longgang Central Hospital of Shenzhen, Shenzhen, China
- Shenzhen Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Peng Zhang
- Division of Neonatology, Children’s Hospital of Fudan University, Shanghai, China
| | - Xueli Zhang
- Division of Neonatology, Shenzhen Longhua People’s Hospital, Shenzhen, China
| | - Tiantian Xiao
- Department of Neonatology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jianhai Zeng
- Division of Neonatology, Longgang Central Hospital of Shenzhen, Shenzhen, China
| | - Kaiping Guo
- Division of Pediatric, Longgang Central Hospital of Shenzhen, Shenzhen, China
| | - Huixian Qiu
- Division of Neonatology, Longgang Central Hospital of Shenzhen, Shenzhen, China
| | - Guoqiang Cheng
- Division of Neonatology, Children’s Hospital of Fudan University, Shanghai, China
| | - Zhangxing Wang
- Division of Neonatology, Shenzhen Longhua People’s Hospital, Shenzhen, China
| | - Wenhao Zhou
- Division of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Shujuan Zeng
- Division of Neonatology, Longgang Central Hospital of Shenzhen, Shenzhen, China
| | - Mingbang Wang
- Department of Neonatology, Longgang Maternity and Child Institute of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital of Shenzhen City), Shenzhen, China
- Microbiome Therapy Center, Department of Experiment & Research, South China Hospital, Medical School, Shenzhen University, Shenzhen, China
| |
Collapse
|
12
|
Liu J, Zhang J, Zhao X, Pan C, Liu Y, Luo S, Miao X, Wu T, Cheng X. Identification of CXCL16 as a diagnostic biomarker for obesity and intervertebral disc degeneration based on machine learning. Sci Rep 2023; 13:21316. [PMID: 38044363 PMCID: PMC10694141 DOI: 10.1038/s41598-023-48580-w] [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: 08/24/2023] [Accepted: 11/28/2023] [Indexed: 12/05/2023] Open
Abstract
Intervertebral disc degeneration (IDD) is the primary cause of neck and back pain. Obesity has been established as a significant risk factor for IDD. The objective of this study was to explore the molecular mechanisms affecting obesity and IDD by identifying the overlapping crosstalk genes associated with both conditions. The identification of specific diagnostic biomarkers for obesity and IDD would have crucial clinical implications. We obtained gene expression profiles of GSE70362 and GSE152991 from the Gene Expression Omnibus, followed by their analysis using two machine learning algorithms, least absolute shrinkage and selection operator and support vector machine-recursive feature elimination, which enabled the identification of C-X-C motif chemokine ligand 16 (CXCL16) as a shared diagnostic biomarker for obesity and IDD. Additionally, gene set variant analysis was used to explore the potential mechanism of CXCL16 in these diseases, and CXCL16 was found to affect IDD through its effect on fatty acid metabolism. Furthermore, correlation analysis between CXCL16 and immune cells demonstrated that CXCL16 negatively regulated T helper 17 cells to promote IDD. Finally, independent external datasets (GSE124272 and GSE59034) were used to verify the diagnostic efficacy of CXCL16. In conclusion, a common diagnostic biomarker for obesity and IDD, CXCL16, was identified using a machine learning algorithm. This study provides a new perspective for exploring the possible mechanisms by which obesity impacts the development of IDD.
Collapse
Affiliation(s)
- Jiahao Liu
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- Institute of Orthopedics of Jiangxi Province, Nanchang, 330006, Jiangxi, China
| | - Jian Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- Institute of Orthopedics of Jiangxi Province, Nanchang, 330006, Jiangxi, China
| | - Xiaokun Zhao
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- Institute of Orthopedics of Jiangxi Province, Nanchang, 330006, Jiangxi, China
| | - Chongzhi Pan
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- Institute of Orthopedics of Jiangxi Province, Nanchang, 330006, Jiangxi, China
| | - Yuchi Liu
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- Institute of Orthopedics of Jiangxi Province, Nanchang, 330006, Jiangxi, China
| | - Shengzhong Luo
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- Institute of Orthopedics of Jiangxi Province, Nanchang, 330006, Jiangxi, China
| | - Xinxin Miao
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- Institute of Orthopedics of Jiangxi Province, Nanchang, 330006, Jiangxi, China
- Institute of Minimally Invasive Orthopedics, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Tianlong Wu
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- Institute of Orthopedics of Jiangxi Province, Nanchang, 330006, Jiangxi, China
- Institute of Minimally Invasive Orthopedics, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xigao Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
- Institute of Orthopedics of Jiangxi Province, Nanchang, 330006, Jiangxi, China.
- Institute of Minimally Invasive Orthopedics, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
13
|
She H, Du Y, Du Y, Tan L, Yang S, Luo X, Li Q, Xiang X, Lu H, Hu Y, Liu L, Li T. Metabolomics and machine learning approaches for diagnostic and prognostic biomarkers screening in sepsis. BMC Anesthesiol 2023; 23:367. [PMID: 37946144 PMCID: PMC10634148 DOI: 10.1186/s12871-023-02317-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Sepsis is a life-threatening disease with a poor prognosis, and metabolic disorders play a crucial role in its development. This study aims to identify key metabolites that may be associated with the accurate diagnosis and prognosis of sepsis. METHODS Septic patients and healthy individuals were enrolled to investigate metabolic changes using non-targeted liquid chromatography-high-resolution mass spectrometry metabolomics. Machine learning algorithms were subsequently employed to identify key differentially expressed metabolites (DEMs). Prognostic-related DEMs were then identified using univariate and multivariate Cox regression analyses. The septic rat model was established to verify the effect of phenylalanine metabolism-related gene MAOA on survival and mean arterial pressure after sepsis. RESULTS A total of 532 DEMs were identified between healthy control and septic patients using metabolomics. The main pathways affected by these DEMs were amino acid biosynthesis, phenylalanine metabolism, tyrosine metabolism, glycine, serine and threonine metabolism, and arginine and proline metabolism. To identify sepsis diagnosis-related biomarkers, support vector machine (SVM) and random forest (RF) algorithms were employed, leading to the identification of four biomarkers. Additionally, analysis of transcriptome data from sepsis patients in the GEO database revealed a significant up-regulation of the phenylalanine metabolism-related gene MAOA in sepsis. Further investigation showed that inhibition of MAOA using the inhibitor RS-8359 reduced phenylalanine levels and improved mean arterial pressure and survival rate in septic rats. Finally, using univariate and multivariate cox regression analysis, six DEMs were identified as prognostic markers for sepsis. CONCLUSIONS This study employed metabolomics and machine learning algorithms to identify differential metabolites that are associated with the diagnosis and prognosis of sepsis patients. Unraveling the relationship between metabolic characteristics and sepsis provides new insights into the underlying biological mechanisms, which could potentially assist in the diagnosis and treatment of sepsis. TRIAL REGISTRATION This human study was approved by the Ethics Committee of the Research Institute of Surgery (2021-179) and was registered by the Chinese Clinical Trial Registry (Date: 09/12/2021, ChiCTR2200055772).
Collapse
Affiliation(s)
- Han She
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yuanlin Du
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yunxia Du
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Lei Tan
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Shunxin Yang
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Xi Luo
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Qinghui Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Xinming Xiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Haibin Lu
- Department of Intensive Care Unit, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yi Hu
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Liangming Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Tao Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
14
|
Hu J, Yang X, Ren J, Zhong S, Fan Q, Li W. Identification and verification of characteristic differentially expressed ferroptosis-related genes in osteosarcoma using bioinformatics analysis. Toxicol Mech Methods 2023; 33:781-795. [PMID: 37488941 DOI: 10.1080/15376516.2023.2240879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 07/26/2023]
Abstract
BACKGROUND This study identified and verified the characteristic differentially expressed ferroptosis-related genes (CDEFRGs) in osteosarcoma (OS). METHODS We extracted ferroptosis-related genes (FRGs), identified differentially expressed FRGs (DEFRGs) in OS, and conducted correlation analysis between DEFRGs. Next, we conducted GO and KEGG analyses to explore the biological functions and pathways of DEFRGs. Furthermore, we used LASSO and SVM-RFE algorithms to screen CDEFRGs, and evaluated its accuracy in diagnosing OS through ROC curves. Then, we demonstrated the molecular function and pathway enrichment of CDEFRGs through GSEA analysis. In addition, we evaluated the differences in immune cell infiltration between OS and NC groups, as well as the correlation between CDEFRGs expressions and immune cell infiltrations. Finally, the expression of CDEFRGs was verified through qRT-PCR, western blotting, and immunohistochemistry experiments. RESULTS We identified 51 DEFRGs and the expression relationship between them. GO and KEGG analysis revealed their key functions and important pathways. Based on four CDEFRGs (PEX3, CPEB1, NOX1, and ALOX5), we built the OS diagnostic model, and verified its accuracy. GSEA analysis further revealed the important functions and pathways of CDEFRGs. In addition, there were differences in immune cell infiltration between OS group and NC group, and CDEFRGs showed significant correlation with certain infiltrating immune cells. Finally, we validated the differential expression levels of four CDEFRGs through external experiments. CONCLUSIONS This study has shed light on the molecular pathological mechanism of OS and has offered novel perspectives for the early diagnosis and immune-targeted therapy of OS patients.
Collapse
Affiliation(s)
- Jianhua Hu
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, P. R. China
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, P. R. China
| | - Xi Yang
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, P. R. China
- Yunnan Key Laboratory of Digital Orthopaedics, Kunming, P. R. China
| | - Jing Ren
- Department of Spinal Surgery, Qujing No. 1 Hospital, Affiliated Qujing Hospital of Kunming Medical University, Qujing, P. R. China
| | - Shixiao Zhong
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, P. R. China
- Yunnan Key Laboratory of Digital Orthopaedics, Kunming, P. R. China
| | - Qianbo Fan
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, P. R. China
- Yunnan Key Laboratory of Digital Orthopaedics, Kunming, P. R. China
| | - Weichao Li
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, P. R. China
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, P. R. China
- Yunnan Key Laboratory of Digital Orthopaedics, Kunming, P. R. China
| |
Collapse
|
15
|
Wang T, Wang M, Liu L, Xie F, Wu X, Li L, Ji J, Wu D. Lower serum branched-chain amino acid catabolic intermediates are predictive signatures specific to patients with diabetic foot. Nutr Res 2023; 119:33-42. [PMID: 37716292 DOI: 10.1016/j.nutres.2023.08.009] [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: 04/25/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/18/2023]
Abstract
Diabetic foot (DF) is one of the serious chronic complications of diabetes. Accurate prediction of the risk of DF may take timely intervention measures to prevent its occurrence. The understanding of metabolomic changes in the progression of diabetes to DF may reveal new targets for interventions. We hypothesized that changes in metabolic pathways during DF would lead to changes in the metabolic profile, which could be predictive signature specific to it. In the present study, 43 participants with type 2 diabetes mellitus (T2DM), 32 T2DM participants with DF (T2DM-F), and 36 healthy subjects were enrolled and their serum samples were used for targeted and nonpolar metabolic analysis with liquid chromatography-tandem mass spectrometry. Differential metabolites related to T2DM-F were discovered in metabolomic analysis. Lasso machine learning regression model, random forest algorithm, causal mediation analysis, disease risk assessment, and clinical decision model were carried out. T2DM and T2DM-F groups could be distinguished with the healthy control group. The differential metabolites were all enriched in alpha-linolenic acid and linoleic acid metabolic pathways including arachidonic acid, docosapentaenoic-acid 22N-6, and docosahexaenoic-acid, which were significantly lower in the T2DM and T2DM-F groups compared with the healthy control group. The differential metabolites in T2DM-F vs T2DM groups were enriched to branched-chain amino acid (BCAA) catabolic pathways involving in methylmalonic acid, succinic acid, 3-methyl-2-oxovaleric acid, and ketoleucine, which were the BCAA catabolic intermediates and significantly lower in the T2DM-F compared with the T2DM group except for succinic acid. We reveal a new set of predictive signatures and associate the lower BCAA catabolic intermediates with the progression from T2DM to T2DM-F.
Collapse
Affiliation(s)
- Tao Wang
- Department of Cardiovascular Surgery, University of Chinese Academy of Science Shenzhen Hospital, Shenzhen, 518027, China
| | - Mingbang Wang
- Microbiome Therapy Center, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, China; Shanghai Key Laboratory of Birth Defects, Division of Neonatology, Children's Hospital of Fudan University, National Center for Children's Health, Shanghai, 201102, China
| | - Liming Liu
- Pathology Department, Shenzhen People's Hospital, Shenzhen, 518027, China
| | - Fang Xie
- Department of Endocrinology, University of Chinese Academy of Science Shenzhen Hospital, Shenzhen, 518027, China
| | - Xuanqin Wu
- Department of Cardiovascular Surgery, University of Chinese Academy of Science Shenzhen Hospital, Shenzhen, 518027, China
| | - Liang Li
- Department of Cardiovascular Surgery, University of Chinese Academy of Science Shenzhen Hospital, Shenzhen, 518027, China
| | - Jun Ji
- Department of Cardiovascular Surgery, University of Chinese Academy of Science Shenzhen Hospital, Shenzhen, 518027, China.
| | - Dafang Wu
- Department of Endocrinology, Affiliated Xi'an No.1 Hospital of Northwest University, Xi'an, 710000, Shanxi, China.
| |
Collapse
|
16
|
O'Sullivan C, Tsai DHT, Wu ICY, Boselli E, Hughes C, Padmanabhan D, Hsia Y. Machine learning applications on neonatal sepsis treatment: a scoping review. BMC Infect Dis 2023; 23:441. [PMID: 37386442 PMCID: PMC10308703 DOI: 10.1186/s12879-023-08409-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/20/2023] [Indexed: 07/01/2023] Open
Abstract
INTRODUCTION Neonatal sepsis is a major cause of health loss and mortality worldwide. Without proper treatment, neonatal sepsis can quickly develop into multisystem organ failure. However, the signs of neonatal sepsis are non-specific, and treatment is labour-intensive and expensive. Moreover, antimicrobial resistance is a significant threat globally, and it has been reported that over 70% of neonatal bloodstream infections are resistant to first-line antibiotic treatment. Machine learning is a potential tool to aid clinicians in diagnosing infections and in determining the most appropriate empiric antibiotic treatment, as has been demonstrated for adult populations. This review aimed to present the application of machine learning on neonatal sepsis treatment. METHODS PubMed, Embase, and Scopus were searched for studies published in English focusing on neonatal sepsis, antibiotics, and machine learning. RESULTS There were 18 studies included in this scoping review. Three studies focused on using machine learning in antibiotic treatment for bloodstream infections, one focused on predicting in-hospital mortality associated with neonatal sepsis, and the remaining studies focused on developing machine learning prediction models to diagnose possible sepsis cases. Gestational age, C-reactive protein levels, and white blood cell count were important predictors to diagnose neonatal sepsis. Age, weight, and days from hospital admission to blood sample taken were important to predict antibiotic-resistant infections. The best-performing machine learning models were random forest and neural networks. CONCLUSION Despite the threat antimicrobial resistance poses, there was a lack of studies focusing on the use of machine learning for aiding empirical antibiotic treatment for neonatal sepsis.
Collapse
Affiliation(s)
| | - Daniel Hsiang-Te Tsai
- Centre for Neonatal and Paediatric Infection, St. George's, University of London, London, UK
- School of Pharmacy, Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ian Chang-Yen Wu
- Centre for Neonatal and Paediatric Infection, St. George's, University of London, London, UK
- School of Pharmacy, Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Pharmacy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Emanuela Boselli
- Department of Pediatrics, V. Buzzi Children's Hospital, University of Milan, Milan, Italy
| | - Carmel Hughes
- School of Pharmacy, Queen's University Belfast, Belfast, UK
| | - Deepak Padmanabhan
- School of Electronics, Electrical Engineering and Computer Science, Queen's University Belfast, Belfast, UK
| | - Yingfen Hsia
- School of Pharmacy, Queen's University Belfast, Belfast, UK
- Centre for Neonatal and Paediatric Infection, St. George's, University of London, London, UK
| |
Collapse
|
17
|
Starikova EA, Rubinstein AA, Mammedova JT, Isakov DV, Kudryavtsev IV. Regulated Arginine Metabolism in Immunopathogenesis of a Wide Range of Diseases: Is There a Way to Pass between Scylla and Charybdis? Curr Issues Mol Biol 2023; 45:3525-3551. [PMID: 37185755 PMCID: PMC10137093 DOI: 10.3390/cimb45040231] [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: 03/29/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023] Open
Abstract
More than a century has passed since arginine was discovered, but the metabolism of the amino acid never ceases to amaze researchers. Being a conditionally essential amino acid, arginine performs many important homeostatic functions in the body; it is involved in the regulation of the cardiovascular system and regeneration processes. In recent years, more and more facts have been accumulating that demonstrate a close relationship between arginine metabolic pathways and immune responses. This opens new opportunities for the development of original ways to treat diseases associated with suppressed or increased activity of the immune system. In this review, we analyze the literature describing the role of arginine metabolism in the immunopathogenesis of a wide range of diseases, and discuss arginine-dependent processes as a possible target for therapeutic approaches.
Collapse
Affiliation(s)
- Eleonora A Starikova
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L'va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
| | - Artem A Rubinstein
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia
| | - Jennet T Mammedova
- Laboratory of General Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia
| | - Dmitry V Isakov
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L'va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
| | - Igor V Kudryavtsev
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia
- School of Biomedicine, Far Eastern Federal University, FEFU Campus, 10 Ajax Bay, Russky Island, 690922 Vladivostok, Russia
| |
Collapse
|
18
|
Surianarayanan C, Lawrence JJ, Chelliah PR, Prakash E, Hewage C. Convergence of Artificial Intelligence and Neuroscience towards the Diagnosis of Neurological Disorders-A Scoping Review. SENSORS (BASEL, SWITZERLAND) 2023; 23:3062. [PMID: 36991773 PMCID: PMC10053494 DOI: 10.3390/s23063062] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 06/19/2023]
Abstract
Artificial intelligence (AI) is a field of computer science that deals with the simulation of human intelligence using machines so that such machines gain problem-solving and decision-making capabilities similar to that of the human brain. Neuroscience is the scientific study of the struczture and cognitive functions of the brain. Neuroscience and AI are mutually interrelated. These two fields help each other in their advancements. The theory of neuroscience has brought many distinct improvisations into the AI field. The biological neural network has led to the realization of complex deep neural network architectures that are used to develop versatile applications, such as text processing, speech recognition, object detection, etc. Additionally, neuroscience helps to validate the existing AI-based models. Reinforcement learning in humans and animals has inspired computer scientists to develop algorithms for reinforcement learning in artificial systems, which enables those systems to learn complex strategies without explicit instruction. Such learning helps in building complex applications, like robot-based surgery, autonomous vehicles, gaming applications, etc. In turn, with its ability to intelligently analyze complex data and extract hidden patterns, AI fits as a perfect choice for analyzing neuroscience data that are very complex. Large-scale AI-based simulations help neuroscientists test their hypotheses. Through an interface with the brain, an AI-based system can extract the brain signals and commands that are generated according to the signals. These commands are fed into devices, such as a robotic arm, which helps in the movement of paralyzed muscles or other human parts. AI has several use cases in analyzing neuroimaging data and reducing the workload of radiologists. The study of neuroscience helps in the early detection and diagnosis of neurological disorders. In the same way, AI can effectively be applied to the prediction and detection of neurological disorders. Thus, in this paper, a scoping review has been carried out on the mutual relationship between AI and neuroscience, emphasizing the convergence between AI and neuroscience in order to detect and predict various neurological disorders.
Collapse
Affiliation(s)
| | | | | | - Edmond Prakash
- Research Center for Creative Arts, University for the Creative Arts (UCA), Farnham GU9 7DS, UK
| | - Chaminda Hewage
- Cardiff School of Technologies, Cardiff Metropolitan University, Cardiff CF5 2YB, UK
| |
Collapse
|
19
|
Investigation into the anti-inflammatory mechanism of coffee leaf extract in LPS-induced Caco-2/U937 co-culture model through cytokines and NMR-based untargeted metabolomics analyses. Food Chem 2023; 404:134592. [DOI: 10.1016/j.foodchem.2022.134592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 08/15/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022]
|
20
|
Dalal S, Onyema EM, Malik A. Hybrid XGBoost model with hyperparameter tuning for prediction of liver disease with better accuracy. World J Gastroenterol 2022; 28:6551-6563. [PMID: 36569269 PMCID: PMC9782838 DOI: 10.3748/wjg.v28.i46.6551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/27/2022] [Accepted: 11/21/2022] [Indexed: 12/08/2022] Open
Abstract
BACKGROUND Liver disease indicates any pathology that can harm or destroy the liver or prevent it from normal functioning. The global community has recently witnessed an increase in the mortality rate due to liver disease. This could be attributed to many factors, among which are human habits, awareness issues, poor healthcare, and late detection. To curb the growing threats from liver disease, early detection is critical to help reduce the risks and improve treatment outcome. Emerging technologies such as machine learning, as shown in this study, could be deployed to assist in enhancing its prediction and treatment.
AIM To present a more efficient system for timely prediction of liver disease using a hybrid eXtreme Gradient Boosting model with hyperparameter tuning with a view to assist in early detection, diagnosis, and reduction of risks and mortality associated with the disease.
METHODS The dataset used in this study consisted of 416 people with liver problems and 167 with no such history. The data were collected from the state of Andhra Pradesh, India, through https://www.kaggle.com/datasets/uciml/indian-liver-patient-records. The population was divided into two sets depending on the disease state of the patient. This binary information was recorded in the attribute "is_patient".
RESULTS The results indicated that the chi-square automated interaction detection and classification and regression trees models achieved an accuracy level of 71.36% and 73.24%, respectively, which was much better than the conventional method. The proposed solution would assist patients and physicians in tackling the problem of liver disease and ensuring that cases are detected early to prevent it from developing into cirrhosis (scarring) and to enhance the survival of patients. The study showed the potential of machine learning in health care, especially as it concerns disease prediction and monitoring.
CONCLUSION This study contributed to the knowledge of machine learning application to health and to the efforts toward combating the problem of liver disease. However, relevant authorities have to invest more into machine learning research and other health technologies to maximize their potential.
Collapse
Affiliation(s)
- Surjeet Dalal
- Department of CSE, Amity University, Gurugram 122413, Haryana, India
| | - Edeh Michael Onyema
- Department of Mathematics and Computer Science, Coal City University, Enugu 400102, Nigeria
| | - Amit Malik
- Department of CSE, SRM University, Delhi-NCR, Sonipat 131001, Haryana, India
| |
Collapse
|
21
|
Liu A, Cai C, Wang Z, Wang B, He J, Xie Y, Deng H, Liu S, Zeng S, Yin Z, Wang M. Inductively coupled plasma mass spectrometry based urine metallome to construct clinical decision models for autism spectrum disorder. METALLOMICS : INTEGRATED BIOMETAL SCIENCE 2022; 14:6849992. [PMID: 36442146 DOI: 10.1093/mtomcs/mfac091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND The global prevalence of autism spectrum disorder (ASD) is on the rise, and high levels of exposure to toxic heavy metals may be associated with this increase. Urine analysis is a noninvasive method for investigating the accumulation and excretion of heavy metals. The aim of this study was to identify ASD-associated urinary metal markers. METHODS Overall, 70 children with ASD and 71 children with typical development (TD) were enrolled in this retrospective case-control study. In this metallomics investigation, inductively coupled plasma mass spectrometry was performed to obtain the urine profile of 27 metals. RESULTS Children with ASD could be distinguished from children with TD based on the urine metal profile, with ASD children showing an increased urine metal Shannon diversity. A metallome-wide association analysis was used to identify seven ASD-related metals in urine, with cobalt, aluminum, selenium, and lithium significantly higher, and manganese, mercury, and titanium significantly lower in the urine of children with ASD than in children with TD. The least absolute shrinkage and selection operator (LASSO) machine learning method was used to rank the seven urine metals in terms of their effect on ASD. On the basis of these seven urine metals, we constructed a LASSO regression model for ASD classification and found an area under the receiver operating characteristic curve of 0.913. We also constructed a clinical prediction model for ASD based on the seven metals that were different in the urine of children with ASD and found that the model would be useful for the clinical prediction of ASD risk. CONCLUSIONS The study findings suggest that altered urine metal concentrations may be an important risk factor for ASD, and we recommend further exploration of the mechanisms and clinical treatment measures for such alterations.
Collapse
Affiliation(s)
- Aiping Liu
- T he department of Laboratory, Baoan Public Health Service Center of Shenzhen, Baoan District, Shenzhen, 518108, China
| | - Chunquan Cai
- Tianjin Pediatric Research Institute, Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Tianjin 300134, China
| | - Zhangxing Wang
- Division of Neonatology, Shenzhen Longhua People's Hospital, Guangdong 518109, China
| | - Bin Wang
- The department of Dermatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Juntao He
- Shenzhen Prevention and Treatment Center for Occupational Diseases (Physical Testing & Chemical Analysis Department), Shenzhen 518020, China
| | - Yanhong Xie
- T he department of Laboratory, Baoan Public Health Service Center of Shenzhen, Baoan District, Shenzhen, 518108, China
| | - Honglian Deng
- T he department of Laboratory, Baoan Public Health Service Center of Shenzhen, Baoan District, Shenzhen, 518108, China
| | - Shaozhi Liu
- T he department of Laboratory, Baoan Public Health Service Center of Shenzhen, Baoan District, Shenzhen, 518108, China
| | - Shujuan Zeng
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Guangdong 518116, China
| | - Zhaoqing Yin
- Division of Pediatrics, The People's Hospital of Dehong Autonomous Prefecture, Dehong Hospital of Kunming Medical University, Mangshi, Yunnan 678400, China
| | - Mingbang Wang
- Microbiome Therapy Center, South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, China.,Shanghai Key Laboratory of Birth Defects, Division of Neonatology, Children's Hospital of Fudan University, National Center for Children's Health, Shanghai 201102, China
| |
Collapse
|
22
|
Ju Z, Guo P, Xiang J, Lei R, Ren G, Zhou M, Yang X, Zhou P, Huang R. Low-dose radiation exaggerates HFD-induced metabolic dysfunction by gut microbiota through PA-PYCR1 axis. Commun Biol 2022; 5:945. [PMID: 36088469 PMCID: PMC9464247 DOI: 10.1038/s42003-022-03929-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 08/31/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractCo-exposure of High-fat-diet (HFD) behavior and environmental low-dose radiation (LDR) is common among majority occupational workers, but the synergism of this co-exposure in metabolic health is poorly understood. This study aimed to investigate the impact of gut microbiota and its metabolites on the regulation of HFD accompanied by LDR-associated with metabolic dysfunction and insulin resistance. Here, we reported that Parasutterella was markedly elevated in the gut microbiota of mice in co-exposure of HFD and LDR, accompanied by increased pyrrolidinecarboxylic acid (PA) level in both intestine and plasma. Transplantation of fecal microbiota from mice with co-exposure HFD and LDR with metabolic dysfunction resulted in increased disruption of metabolic dysfunction, insulin resistance and increased PYCR1 (Pyrroline-5-carboxylate reductase 1) expression. Mechanistically, intestinal barrier was damaged more serious in mice with co-exposure of HFD and LDR, leading high PA level in plasma, activating PYCR1 expression to inhibit insulin Akt/mTOR (AKT kinase-transforming protein/Serine threonine-protein kinase) signaling pathway to aggravate HFD-induced metabolic impairments. This study suggests a new avenue for interventions against western diet companied with low dose radiation exposure-driven metabolic impairments.
Collapse
|
23
|
Zeng S, Wang Z, Zhang P, Yin Z, Huang X, Tang X, Shi L, Guo K, Liu T, Wang M, Qiu H. Machine learning approach identifies meconium metabolites as potential biomarkers of neonatal hyperbilirubinemia. Comput Struct Biotechnol J 2022; 20:1778-1784. [PMID: 35495115 PMCID: PMC9027383 DOI: 10.1016/j.csbj.2022.03.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 12/12/2022] Open
Abstract
Background The gut microbiota plays an important role in the early stages of human life. Our previous study showed that the abundance of intestinal flora involved in galactose metabolism was altered and correlated with increased serum bilirubin levels in children with jaundice. We conducted the present study to systematically evaluate alterations in the meconium metabolome of neonates with jaundice and search for metabolic markers associated with neonatal jaundice. Methods We included 68 neonates with neonatal hyperbilirubinemia, also known as neonatal jaundice (NJ) and 68 matched healthy controls (HC), collected meconium samples from them at birth, and performed metabolomic analysis via liquid chromatography-mass spectrometry. Results Gut metabolites enabled clearly distinguishing the neonatal jaundice (NJ) and healthy control (HC) groups. We also identified the compositions of the gut metabolites that differed significantly between the NJ and HC groups; these differentially significant metabolites were enriched in aminyl tRNA biosynthesis; pantothenic acid and coenzyme biosynthesis; and the valine, leucine and isoleucine biosynthesis pathways. Gut branched-chain amino acid (BCAA) levels were positively correlated with serum bilirubin levels, and the area under the receiver operating characteristic curve of the random forest classifier model based on BCAAs, proline, methionine, phenylalanine and total bilirubin reached 96.9%, showing good potential for diagnostic applications. Machine learning-based causal inference analysis revealed the causal effect of BCAAs on serum total bilirubin and NJ. Conclusions Altered gut metabolites in neonates with jaundice showed that increased BCAAs and total serum bilirubin were positively correlated. BCAAs proline, methionine, phenylalanine are potential biomarkers of NJ.
Collapse
Key Words
- AUROC, the area under the ROC
- BCAA, branched-chain amino acid
- Gut microbiota
- HC, healthy controls
- KEGG, Kyoto Encyclopedia of Genes and Genomes
- LC-MS, liquid chromatography-mass spectrometry
- MSUD, maple syrup urine disease
- Machine learning
- NJ, neonatal jaundice
- OPLS-DA, orthogonal partial least squares-discriminant analysis
- PCA, the principal component analysis
- PLS, partial least-squares regression
- ROC, receiver operating characteristic
- branched-chain amino acid
- causal inference
- metabolome
- neonatal hyperbilirubinemia
Collapse
Affiliation(s)
- Shujuan Zeng
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Guangdong 518116, China
| | - Zhangxing Wang
- Division of Neonatology, Shenzhen Longhua People’s Hospital, Guangdong 518109, China
| | - Peng Zhang
- Division of Neonatology, Shenzhen Longhua People’s Hospital, Guangdong 518109, China
| | - Zhaoqing Yin
- Division of Neonatology, The People's Hospital of Dehong Autonomous Prefecture, Mangshi, Yunnan 678400, China
| | - Xunbin Huang
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Guangdong 518116, China
| | - Xisheng Tang
- Oncology Department, Longgang District Central Hospital of Shenzhen, Shenzhen 518116, China
| | - Lindong Shi
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Guangdong 518116, China
| | - Kaiping Guo
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Guangdong 518116, China
| | - Ting Liu
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Guangdong 518116, China
| | - Mingbang Wang
- Shanghai Key Laboratory of Birth Defects, Division of Neonatology, Children’s Hospital of Fudan University, National Center for Children’s Health, Shanghai 201102, China
- Microbiome Therapy Center, South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, China
| | - Huixian Qiu
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Guangdong 518116, China
| |
Collapse
|