1
|
Xu Z, Wang K, Hu H, Hu Y, Huang J, Luo Z. Sinensetin attenuates LPS-induced acute pulmonary inflammation in mice and RAW264.7 cells by modulating NF-κB p65-mediated immune resistance and STAT3-mediated tissue resilience. Int Immunopharmacol 2025; 148:114101. [PMID: 39827664 DOI: 10.1016/j.intimp.2025.114101] [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: 12/14/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
Acute pulmonary inflammation is a severe lower respiratory tract infection. Sinensetin (SIN), a polymethoxyflavone with strong anti-inflammatory properties, is known to ameliorate LPS-induced acute inflammatory lung injury, but its molecular mechanisms are not fully understood. This study aimed to provide insight into the pharmacological mechanisms of SIN in attenuating acute pulmonary inflammation. In LPS-induced inflammation assays in vivo and in vitro, SIN significantly reduced the mRNA levels of inflammatory genes including MCP-1, ICAM1, Ccl3, Ccl4, Ccl5, Ccl7, Cxcl9, Cxcl10, IL1α, IL1β, IL6, IL11, IL18, IL27, TNF-α, IFN-γ, TLR4, MyD88, F4/80, COX2, iNOS, NLRP3, ASC, JAK2, STAT3, STAT4, and Bcl2l1, as well as increased the mRNA levels of anti-inflammatory genes such as IL4, IL10, and IL12α. Besides, SIN markedly decreased the expression of CD68, TLR4, MyD88, phospho-IκBα (S32/S36), phospho-NF-κB p65 (S536), MCP-1, ICAM1, phospho-JAK2 (Tyr1008), phospho-STAT1 (S727), phospho-STAT3 (Y705), and phospho-STAT4 (Y693), inhibited NF-κB p65 translocation into the nucleus, thereby blocking in combination with STAT transcription factors to induce target gene expression. Further GC-MS/MS and LC-MS/MS metabolomic analysis revealed that SIN significantly increased the abundance of anti-inflammatory metabolites, such as L-alanine, L-carnitine, L-glutamic acid, Glycine, and L-cysteine. In conclusion, the results indicated that SIN attenuated LPS-induced acute pulmonary inflammation by modulating NF-κB p65-mediated immune resistance and STAT3-mediated tissue resilience. All these favorable findings presented critical insights into the remarkable abilities and health benefits of SIN in ameliorating inflammatory lung disease.
Collapse
Affiliation(s)
- Zaibin Xu
- Hainan Pharmaceutical Research and Development Science Park, Hainan Medical University, Haikou 571157 China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405 China
| | - Kongyan Wang
- Hainan Pharmaceutical Research and Development Science Park, Hainan Medical University, Haikou 571157 China
| | - Huiyu Hu
- Hainan Pharmaceutical Research and Development Science Park, Hainan Medical University, Haikou 571157 China
| | - Yingjie Hu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405 China.
| | - Jiawen Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405 China.
| | - Zhuohui Luo
- Hainan Pharmaceutical Research and Development Science Park, Hainan Medical University, Haikou 571157 China; Research Center for Drug Safety Evaluation of Hainan Province, Hainan Medical University, Haikou 571199 China.
| |
Collapse
|
2
|
Wang JY, Gui TT, Jiao B, Liu X, Ma XL, Wang C, Qiao J, Liu WY, Peng LJ, Ren JY, Zhu YM, Weng XQ, Wang C, Zhang QQ, Song GX, Dai YT, Wang ZY, Lv G, Gao CX, Qiao N, Zhang M, Tan Y, Liu YF, Wang SY, Hou J, Jing DH, Lyu AK, Mi JQ, Chen Z, Chen WL, Yin T, Fang H, Wang J, Chen SJ. Metabolomic insights into pathogenesis and therapeutic potential in adult acute lymphoblastic leukemia. Proc Natl Acad Sci U S A 2025; 122:e2423169122. [PMID: 39946534 DOI: 10.1073/pnas.2423169122] [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: 11/10/2024] [Accepted: 01/03/2025] [Indexed: 02/26/2025] Open
Abstract
Acute lymphoblastic leukemia (ALL) poses challenges in adult patients, considering its heterogeneous nature and often suboptimal treatment outcomes. Here, we performed a study on 201 newly diagnosed adult ALL cases (age ≥ 15 y) to generate intracellular and dynamic serum metabolomic profiles. Our findings revealed a predominant increase in bile acid (BA) metabolites in serum, alongside metabolic rewiring that supported highly proliferative states and actively metabolic signaling, such as enriched nucleotide metabolism in leukemic blasts. By integrating intracellular metabolomics and transcriptomics, we constructed the Comprehensive Metabolic Information Dataset (CMID), which facilitated the development of a clustering system to supplement current risk stratification. Furthermore, we explored potential metabolic interventions targeting the serum BA profile and energy metabolism in blasts. The combined use of simvastatin with vincristine and dexamethasone regimen demonstrated a synergistic therapeutic effect in a murine ALL model, effectively lowering key BA levels in serum and suppressing the infiltration of leukemic blasts in the liver. In light of the enhanced intracellular redox metabolism, combining FK866 (a nicotinamide phosphoribosyltransferase inhibitor) and venetoclax significantly prolonged survival in a patient-derived xenograft ALL model. Our findings, along with the resulting resources (http://www.genetictargets.com/MALL), provide a framework for the metabolism-centered management of ALL.
Collapse
Affiliation(s)
- Jun-Yu Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Tuan-Tuan Gui
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Bo Jiao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xuan Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiao-Lin Ma
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Cheng Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jing Qiao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wei-Yang Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Li-Jun Peng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jia-Yi Ren
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yong-Mei Zhu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiang-Qin Weng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chao Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qian-Qian Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Gao-Xian Song
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yu-Ting Dai
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhen-Yi Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Gang Lv
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chen-Xu Gao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Niu Qiao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ming Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yun Tan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yuan-Fang Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Sheng-Yue Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jian Hou
- Department of Hematology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Duo-Hui Jing
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - An-Kang Lyu
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jian-Qing Mi
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhu Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wen-Lian Chen
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Tong Yin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hai Fang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jin Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Sai-Juan Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Research Unit of Hematologic Malignancies Genomics and Translational Research of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
3
|
Kyei-Baffour VO, Vijaya AK, Burokas A, Daliri EBM. Psychobiotics and the gut-brain axis: advances in metabolite quantification and their implications for mental health. Crit Rev Food Sci Nutr 2025:1-20. [PMID: 39907087 DOI: 10.1080/10408398.2025.2459341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Psychobiotics are live microorganisms that, when administered in adequate amounts, confer mental health benefits to the host. Several clinical studies have demonstrated significant mental health benefits from psychobiotic administration, making them an emerging topic in food science. Certain strains of Lactobacillus, Bifidobacterium, Streptococcus, Escherichia, and Enterococcus species are known for their ability to modulate the gut-brain axis and provide mental health benefits. Proposed action mechanisms include the production of neuroactive compounds or their precursors, which may cross the blood-brain barrier, or transported by their extracellular vesicles. However, there is a lack of in vivo evidence directly confirming these mechanisms, although indirect evidence from recent studies suggest potential pathways for further investigation. To advance our understanding, it is crucial to study these mechanisms within the host, with accurate quantification of neuroactive compounds and/or their precursors being key in such studies. Current quantification methods, however, face challenges, such as low sensitivity for detecting trace metabolites and limited specificity due to interference from other compounds, impacting the reliability of measurements. This review discusses the emerging field of psychobiotics, their potential action mechanisms, neuroactive compound estimation techniques, and perspectives for improvement in quantifying neuroactive compounds and/or precursors within the host.
Collapse
Affiliation(s)
- Vincent Owusu Kyei-Baffour
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Akshay Kumar Vijaya
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Aurelijus Burokas
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Eric Banan-Mwine Daliri
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
4
|
Wang N, Ockerman FP, Zhou LY, Grove ML, Alkis T, Barnard J, Bowler RP, Clish CB, Chung S, Drzymalla E, Evans AM, Franceschini N, Gerszten RE, Gillman MG, Hutton SR, Kelly RS, Kooperberg C, Larson MG, Lasky-Su J, Meyers DA, Woodruff PG, Reiner AP, Rich SS, Rotter JI, Silverman EK, Ramachandran VS, Weiss ST, Wong KE, Wood AC, Wu L, Yarden R, Blackwell TW, Smith AV, Chen H, Raffield LM, Yu B. Genetic Architecture and Analysis Practices of Circulating Metabolites in the NHLBI Trans-Omics for Precision Medicine (TOPMed) Program. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.23.604849. [PMID: 39211135 PMCID: PMC11361093 DOI: 10.1101/2024.07.23.604849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Circulating metabolite levels partly reflect the state of human health and diseases and can be impacted by genetic determinants. Hundreds of loci associated with circulating metabolites have been identified; however, most findings focus on predominantly European ancestry or single-study analyses. Leveraging the rich metabolomics resources generated by the NHLBI Trans-Omics for Precision Medicine (TOPMed) Program, we harmonized and accessibly cataloged 1,729 circulating metabolites among 25,058 ancestrally diverse samples. We provided a set of reasonable strategies for outlier and imputation handling to process metabolite data. Following the practical analysis framework, we further performed a genome-wide association analysis on 1,135 selected metabolites using whole genome sequencing data from 16,359 individuals passing the quality control filters, and discovered 1,778 independent loci associated with 667 metabolites. Among 108 novel locus-metabolite pairs, we detected not only novel loci within previously implicated metabolite associated genes but also novel genes (such as GAB3 and VSIG4 located in the X chromosome) that have putative roles in metabolic regulation. In the sex-stratified analysis, we revealed 85 independent locus-metabolite pairs with evidence of sexual dimorphism, including well-known metabolic genes such as FADS2 , D2HGDH , SUGP1 , UTG2B17 , strongly supporting the importance of exploring sex difference in the human metabolome. Taken together, our study depicted the genetic contribution to circulating metabolite levels, providing additional insight into the understanding of human health.
Collapse
|
5
|
Huang J, Qin TS, Bo Y, Li YJ, Liu RS, Yu Y, Li XD, He JC, Ma AX, Tao DP, Ren WJ, Peng J. The Role of the Intestinal Flora and Its Derivatives in Neurocognitive Disorders: A Narrative Review from Surgical Perspective. Mol Neurobiol 2025; 62:1404-1414. [PMID: 38985257 PMCID: PMC11772545 DOI: 10.1007/s12035-024-04322-1] [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: 04/20/2024] [Accepted: 06/19/2024] [Indexed: 07/11/2024]
Abstract
Perioperative neurocognitive dysfunction is a significant concern for population health, impacting postoperative recovery and increasing the financial burden on patients. With an increasing number of surgical procedures being performed, the prevention and management of perioperative neurocognitive dysfunction have garnered significant attention. While factors such as age, lifestyle, genetics, and education are known to influence the development of cognitive dysfunction, recent research has highlighted the role of the gut microbiota in neurological health. An increased abundance of pro-inflammatory gut microbiota can trigger and worsen neuroinflammation, neuronal cell damage, and impaired cellular autophagy. Moreover, the inflammation-promoting gut microbiota can disrupt immune function, impair neuroautophagy, and affect the production and circulation of extracellular vesicles and neurotransmitters. These factors collectively play a role in the onset and advancement of cognitive impairment. This narrative review delves into the molecular mechanisms through which gut microbiota and their derivatives contribute to cognitive impairment, focusing on the impact of anesthesia surgery, changes in gut microbial populations, and perioperative cognitive impairment associations. The study suggests that alterations in the abundance of various bacterial species and their metabolites pre- and post-surgery may be linked to postoperative cognitive impairment. Furthermore, the potential of probiotics or prebiotics in addressing cognitive impairment is discussed, offering a promising avenue for investigating the treatment of perioperative neurocognitive disorders.
Collapse
Affiliation(s)
- Jian Huang
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, People's Republic of China
| | - Tian-Shou Qin
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, People's Republic of China
| | - Yun Bo
- Department of Anesthesiology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| | - Yu-Jin Li
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| | - Rong-Sheng Liu
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| | - Yang Yu
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| | - Xiao-Dong Li
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, People's Republic of China
| | - Jin-Can He
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, People's Republic of China
| | - Ai-Xin Ma
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, People's Republic of China
| | - Da-Peng Tao
- School of Information Science and Engineering, Yunnan University, Kunming, 650504, China
| | - Wen-Jun Ren
- Department of Cardiovascular Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, Yunnan, China.
| | - Jun Peng
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| |
Collapse
|
6
|
Puvvula J, Song LC, Zalewska KJ, Alexander A, Manz KE, Braun JM, Pennell KD, DeFranco EA, Ho SM, Leung YK, Huang S, Vuong AM, Kim SS, Percy Z, Bhashyam P, Lee R, Jones DP, Tran V, Kim DV, Calafat AM, Botelho JC, Chen A. Global metabolomic alterations associated with endocrine-disrupting chemicals among pregnant individuals and newborns. Metabolomics 2025; 21:20. [PMID: 39863779 PMCID: PMC11762426 DOI: 10.1007/s11306-024-02219-7] [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: 10/04/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Gestational exposure to non-persistent endocrine-disrupting chemicals (EDCs) may be associated with adverse pregnancy outcomes. While many EDCs affect the endocrine system, their effects on endocrine-related metabolic pathways remain unclear. This study aims to explore the global metabolome changes associated with EDC biomarkers at delivery. METHODS This study included 75 pregnant individuals who delivered at the University of Cincinnati Hospital from 2014 to 2017. We measured maternal urinary biomarkers of paraben/phenol (12), phthalate (13), and phthalate replacements (4) from the samples collected during the delivery visit. Global serum metabolome profiles were analyzed from maternal blood (n = 72) and newborn (n = 63) cord blood samples collected at delivery. Fifteen of the 29 urinary biomarkers were excluded due to low detection frequency or potential exposures during hospital stay. We assessed metabolome-wide associations between 14 maternal urinary biomarkers and maternal/newborn metabolome profiles. Additionally, performed enrichment analysis to identify potential alterations in metabolic pathways. RESULTS We observed metabolome-wide associations between maternal urinary concentrations of phthalate metabolites (mono-isobutyl phthalate), phthalate replacements (mono-2-ethyl-5-carboxypentyl terephthalate, mono-2-ethyl-5-hydroxyhexyl terephthalate) and phenols (bisphenol-A, bisphenol-S) and maternal serum metabolome, using q-value < 0.2 as a threshold. Additionally, associations of phthalate metabolites (mono-n-butyl phthalate, monobenzyl phthalate) and phenols (2,5-dichlorophenol, BPA) with the newborn metabolome were noted. Enrichment analyses revealed associations (p-gamma < 0.05) with amino acid, carbohydrate, lipid, glycan, vitamin, and other cofactor metabolism pathways. CONCLUSION Maternal paraben, phenol, phthalate, and phthalate replacement biomarker concentrations at delivery were associated with maternal and newborn serum global metabolome.
Collapse
Affiliation(s)
- Jagadeesh Puvvula
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Lucie C Song
- College of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Kathrine E Manz
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Joseph M Braun
- Department of Epidemiology, Brown University, Providence, RI, USA
| | - Kurt D Pennell
- School of Engineering, Brown University, Providence, RI, USA
| | - Emily A DeFranco
- Department of Obstetrics and Gynecology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Shuk-Mei Ho
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Yuet-Kin Leung
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Shouxiong Huang
- Pathogen-Host Interaction Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Ann M Vuong
- Department of Epidemiology and Biostatistics, School of Public Health, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Stephani S Kim
- Health Research, Battelle Memorial Institute, Columbus, OH, USA
| | - Zana Percy
- Department of Environmental & Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Priyanka Bhashyam
- College of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Raymund Lee
- College of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Dean P Jones
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, USA
| | - Vilinh Tran
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, USA
| | - Dasom V Kim
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Antonia M Calafat
- National Center for Environmental Health, U.S. Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Julianne C Botelho
- National Center for Environmental Health, U.S. Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Aimin Chen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
7
|
Li H, Ke X, Feng B, Tian H, Cai Z, Zhang A, Man Q. Research progress on the mechanism and markers of metabolic disorders in the occurrence and development of cognitive dysfunction after ischemic stroke. Front Endocrinol (Lausanne) 2025; 16:1500650. [PMID: 39911922 PMCID: PMC11794095 DOI: 10.3389/fendo.2025.1500650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 01/03/2025] [Indexed: 02/07/2025] Open
Abstract
Post-stroke cognitive impairment (PSCI) is a common complication following a stroke that significantly affects patients' quality of life and rehabilitation outcomes. It also imposes a heavy economic burden. There is an urgent need to better understand the pathophysiology and pathogenesis of PSCI, as well as to identify markers that can predict PSCI early in the clinical stage, facilitating early prevention, monitoring, and treatment. Although the mechanisms underlying PSCI are complex and multifaceted, involving factors such as atherosclerosis and neuroinflammation, metabolic disorders also play a critical role. This article primarily reviews the relationship between metabolic disorders of the three major nutrients-sugar, fat, and protein-and the development of cognitive dysfunction following ischemic stroke (IS). It aims to elucidate how these metabolic disturbances contribute to cognitive dysfunction post-stroke and to explore potential metabolic biomarkers for PSCI. We believe that this review will offer new insights into the early identification, treatment, and prognostic assessment of PSCI.
Collapse
Affiliation(s)
- Huaqiang Li
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaohua Ke
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bianying Feng
- Department of Clinical Laboratory, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huan Tian
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhenzhen Cai
- Department of Clinical Laboratory, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Anren Zhang
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qiuhong Man
- Department of Clinical Laboratory, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
8
|
Lu J, Liu R, Ren H, Wang S, Hu C, Shi Z, Li M, Liu W, Wan Q, Su Q, Li Q, Zheng H, Qu S, Yang F, Ji H, Lin H, Qi H, Wu X, Wu K, Chen Y, Xu Y, Xu M, Wang T, Zheng J, Ning G, Zheng R, Bi Y, Zhong H, Wang W. Impact of omega-3 fatty acids on hypertriglyceridemia, lipidomics, and gut microbiome in patients with type 2 diabetes. MED 2025; 6:100496. [PMID: 39163858 DOI: 10.1016/j.medj.2024.07.024] [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: 12/05/2023] [Revised: 05/14/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024]
Abstract
BACKGROUND Fish oil (FO), a mixture of omega-3 fatty acids mainly comprising docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), has been recommended for patients with type 2 diabetes (T2D) and hypertriglyceridemia. However, its effects on lipidomic profiles and gut microbiota and the factors influencing triglyceride (TG) reduction remain unclear. METHODS We conducted a 12-week, randomized, double-blind, placebo-controlled trial in 309 Chinese patients with T2D with hypertriglyceridemia (ClinicalTrials.gov: NCT03120299). Participants were randomly assigned (1:1) to receive either 4 g FO or corn oil for 12 weeks. The primary outcome was changes in serum TGs and the lipidomic profile, and the secondary outcome included changes in the gut microbiome and other metabolic variables. FINDINGS The FO group had significantly better TG reduction (mean [95% confidence interval (CI)]: -1.51 [-2.01, -1.01] mmol/L) compared to the corn oil group (-0.66 [-1.15, -0.16] mmol/L, p = 0.02). FO significantly altered the serum lipid profile by reducing low-unsaturated TG species and increasing those containing DHA or EPA. FO had minor effects on gut microbiota, while baseline microbial features predicted the TG response to FO better than phenotypic or lipidomic features, potentially mediated by specific lipid metabolites. A total of 9 lipid metabolites significantly mediated the link between 4 baseline microbial variables and the TG response to FO supplementation. CONCLUSIONS Our findings demonstrate differential impacts of omega-3 fatty acids on lipidomic and microbial profiles in T2D and highlight the importance of baseline gut microbiota characteristics in predicting the TG-lowering efficacy of FO. FUNDING This study was funded by the National Nature Science Foundation.
Collapse
Affiliation(s)
- Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huahui Ren
- BGI Research, Shenzhen, China; Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen, China
| | - Shuangyuan Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunyan Hu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhun Shi
- BGI Research, Shenzhen, China; Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen, China
| | - Mian Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Liu
- Department of Endocrinology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qin Wan
- Department of Endocrine and Metabolic Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Qing Su
- Department of Endocrinology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qifu Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongting Zheng
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Shen Qu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tenth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Fangming Yang
- BGI Research, Shenzhen, China; Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen, China
| | | | - Hong Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongyan Qi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueyan Wu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kui Wu
- BGI Research, Shenzhen, China; Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen, China
| | - Yuhong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiange Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruizhi Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huanzi Zhong
- BGI Research, Shenzhen, China; Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen, China.
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
9
|
Huang H, Chen Y, Xu W, Cao L, Qian K, Bischof E, Kennedy BK, Pu J. Decoding aging clocks: New insights from metabolomics. Cell Metab 2025; 37:34-58. [PMID: 39657675 DOI: 10.1016/j.cmet.2024.11.007] [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: 02/25/2024] [Revised: 09/23/2024] [Accepted: 11/10/2024] [Indexed: 12/12/2024]
Abstract
Chronological age is a crucial risk factor for diseases and disabilities among older adults. However, individuals of the same chronological age often exhibit divergent biological aging states, resulting in distinct individual risk profiles. Chronological age estimators based on omics data and machine learning techniques, known as aging clocks, provide a valuable framework for interpreting molecular-level biological aging. Metabolomics is an intriguing and rapidly growing field of study, involving the comprehensive profiling of small molecules within the body and providing the ultimate genome-environment interaction readout. Consequently, leveraging metabolomics to characterize biological aging holds immense potential. The aim of this review was to provide an overview of metabolomics approaches, highlighting the establishment and interpretation of metabolomic aging clocks while emphasizing their strengths, limitations, and applications, and to discuss their underlying biological significance, which has the potential to drive innovation in longevity research and development.
Collapse
Affiliation(s)
- Honghao Huang
- Division of Cardiology, State Key Laboratory for Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yifan Chen
- Division of Cardiology, State Key Laboratory for Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Xu
- Division of Cardiology, State Key Laboratory for Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Linlin Cao
- Division of Cardiology, State Key Laboratory for Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kun Qian
- Division of Cardiology, State Key Laboratory for Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Evelyne Bischof
- University Hospital of Basel, Division of Internal Medicine, University of Basel, Basel, Switzerland; Shanghai University of Medicine and Health Sciences, College of Clinical Medicine, Shanghai, China
| | - Brian K Kennedy
- Health Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Centre for Healthy Longevity, National University Health System, Singapore, Singapore; Departments of Biochemistry and Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Jun Pu
- Division of Cardiology, State Key Laboratory for Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Aging Biomarker Consortium, China.
| |
Collapse
|
10
|
Hafner A, DeLeo V, Deng CH, Elsik CG, S Fleming D, Harrison PW, Kalbfleisch TS, Petry B, Pucker B, Quezada-Rodríguez EH, Tuggle CK, Koltes JE. Data reuse in agricultural genomics research: challenges and recommendations. Gigascience 2025; 14:giae106. [PMID: 39804724 PMCID: PMC11727710 DOI: 10.1093/gigascience/giae106] [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: 06/17/2024] [Revised: 09/17/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025] Open
Abstract
The scientific community has long benefited from the opportunities provided by data reuse. Recognizing the need to identify the challenges and bottlenecks to reuse in the agricultural research community and propose solutions for them, the data reuse working group was started within the AgBioData consortium framework. Here, we identify the limitations of data standards, metadata deficiencies, data interoperability, data ownership, data availability, user skill level, resource availability, and equity issues, with a specific focus on agricultural genomics research. We propose possible solutions stakeholders could implement to mitigate and overcome these challenges and provide an optimistic perspective on the future of genomics and transcriptomics data reuse.
Collapse
Affiliation(s)
- Alenka Hafner
- Department of Biology, Frear North, Pennsylvania State University, University Park, PA, 16802, US
- Intercollege Graduate Degree Program in Plant Biology, Pennsylvania State University, University Park, PA, 16802, US
| | | | - Cecilia H Deng
- New Cultivar Innovation, The New Zealand Institute for Plant and Food Research Limited, Auckland, 1025, New Zealand
| | - Christine G Elsik
- Division of Animal Sciences and Division of Plant Science & Technology, University of Missouri, MO, 65211, US
- Institute for Data Science & Informatics, University of Missouri, MO, 65211, US
| | - Damarius S Fleming
- Animal Parasitic Diseases Laboratory, United States Department of Agriculture Agricultural Research Service, Beltsville, MD, 20705, US
| | - Peter W Harrison
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, Cambridgeshire, CB10 1SD, UK
| | - Theodore S Kalbfleisch
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food, and Environment, University of Kentucky, Lexington, KY, 40202, US
| | - Bruna Petry
- Department of Animal Science, Iowa State University, Ames, IA, 50011, US
| | - Boas Pucker
- Institute of Plant Biology & BRICS, TU Braunschweig, Braunschweig, 38106, Germany
| | - Elsa H Quezada-Rodríguez
- Departamento de Producción Agrícola y Animal, Universidad Autónoma Metropolitana-Xochimilco, Ciudad de México, 04510, México
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Ciudad de México, 04510, México
| | | | - James E Koltes
- Department of Animal Science, Iowa State University, Ames, IA, 50011, US
| |
Collapse
|
11
|
Zhang Z, Liang Y, Mo S, Zhao M, Li Y, Zhang C, Shan X, Liu S, Liao J, Luo X, Zhu J, Wang C, Jiang Q, Hou C, Hong W, Lai N, Chen Y, Xu L, Lu W, Wang J, Wang Z, Yang K. Oral administration of pioglitazone inhibits pulmonary hypertension by regulating the gut microbiome and plasma metabolome in male rats. Physiol Rep 2025; 13:e70174. [PMID: 39739369 DOI: 10.14814/phy2.70174] [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: 10/07/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025] Open
Abstract
The oral administrated thiazolidinediones (TZDs) have been widely reported to alleviate experimental pulmonary hypertension (PH). However, previous studies mainly focused on their beneficial effects on the cardiopulmonary vascular system but failed to determine their potential roles on gut microenvironment. This study aims to investigate the effects of pioglitazone, an oral TZD drug, on gut microbiome in classic PH rat models induced by hypoxia (HPH) or SU5416/hypoxia (SuHx-PH) and evaluate the therapeutic potential of supplementation of selective probiotics for experimental PH. Pioglitazone remarkably inhibited the PH pathogenesis in both models and reshaped the gut microbiome and plasma metabolome. Correlation analyses represented strong and unique association between the protective metabolites and bacteria genera (Roseburia, Lactobacillus, and Streptococcus) that were positively stimulated by pioglitazone. Supplementation of selective probiotics Roseburia intestinalis (R. intestinalis) partially attenuated SuHx-PH and rebuilt a novel gut microbiome and host metabolome. This study reports for the first time that oral administration of pioglitazone protects PH by regulating the gut microbiome and host metabolome, providing novel insights for the TZD drugs. The data also supports that modulation of gut microbiota by supplementation of selective probiotics could be a novel effective therapeutic strategy for the treatment of PH.
Collapse
Affiliation(s)
- Zizhou Zhang
- Department of Laboratory Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, China
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yaru Liang
- Department of Laboratory Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Shaocong Mo
- Department of Laboratory Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, China
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Mingming Zhao
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yi Li
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chenting Zhang
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaoqian Shan
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Shiyun Liu
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Pulmonary Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Diseases of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jing Liao
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xiaoyun Luo
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Junqi Zhu
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chen Wang
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qian Jiang
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chi Hou
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wei Hong
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ning Lai
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuqin Chen
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lei Xu
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Zhongfang Wang
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Kai Yang
- Department of Laboratory Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, China
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
12
|
Araújo R, Ramalhete L, Von Rekowski CP, Fonseca TAH, Bento L, R. C. Calado C. Early Mortality Prediction in Intensive Care Unit Patients Based on Serum Metabolomic Fingerprint. Int J Mol Sci 2024; 25:13609. [PMID: 39769370 PMCID: PMC11677344 DOI: 10.3390/ijms252413609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/14/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Predicting mortality in intensive care units (ICUs) is essential for timely interventions and efficient resource use, especially during pandemics like COVID-19, where high mortality persisted even after the state of emergency ended. Current mortality prediction methods remain limited, especially for critically ill ICU patients, due to their dynamic metabolic changes and heterogeneous pathophysiological processes. This study evaluated how the serum metabolomic fingerprint, acquired through Fourier-Transform Infrared (FTIR) spectroscopy, could support mortality prediction models in COVID-19 ICU patients. A preliminary univariate analysis of serum FTIR spectra revealed significant spectral differences between 21 discharged and 23 deceased patients; however, the most significant spectral bands did not yield high-performing predictive models. By applying a Fast-Correlation-Based Filter (FCBF) for feature selection of the spectra, a set of spectral bands spanning a broader range of molecular functional groups was identified, which enabled Naïve Bayes models with AUCs of 0.79, 0.97, and 0.98 for the first 48 h of ICU admission, seven days prior, and the day of the outcome, respectively, which are, in turn, defined as either death or discharge from the ICU. These findings suggest FTIR spectroscopy as a rapid, economical, and minimally invasive diagnostic tool, but further validation is needed in larger, more diverse cohorts.
Collapse
Affiliation(s)
- Rúben Araújo
- NMS—NOVA Medical School, FCM—Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria 130, 1169-056 Lisbon, Portugal; (R.A.)
- CHRC—Comprehensive Health Research Centre, Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal
- ISEL—Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, Rua Conselheiro Emídio Navarro 1, 1959-007 Lisbon, Portugal
| | - Luís Ramalhete
- NMS—NOVA Medical School, FCM—Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria 130, 1169-056 Lisbon, Portugal; (R.A.)
- IPST—Instituto Português do Sangue e da Transplantação, Alameda das Linhas de Torres—nr.117, 1769-001 Lisbon, Portugal
- iNOVA4Health—Advancing Precision Medicine, RG11, Reno-Vascular Diseases Group, NMS—NOVA Medical School, FCM—Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal
| | - Cristiana P. Von Rekowski
- NMS—NOVA Medical School, FCM—Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria 130, 1169-056 Lisbon, Portugal; (R.A.)
- CHRC—Comprehensive Health Research Centre, Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal
- ISEL—Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, Rua Conselheiro Emídio Navarro 1, 1959-007 Lisbon, Portugal
| | - Tiago A. H. Fonseca
- NMS—NOVA Medical School, FCM—Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria 130, 1169-056 Lisbon, Portugal; (R.A.)
- CHRC—Comprehensive Health Research Centre, Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal
- ISEL—Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, Rua Conselheiro Emídio Navarro 1, 1959-007 Lisbon, Portugal
| | - Luís Bento
- NMS—NOVA Medical School, FCM—Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria 130, 1169-056 Lisbon, Portugal; (R.A.)
- CHRC—Comprehensive Health Research Centre, Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal
- Intensive Care Department, ULSSJ—Unidade Local de Saúde São José, Rua José António Serrano, 1150-199 Lisbon, Portugal
- Integrated Pathophysiological Mechanisms, CHRC—Comprehensive Health Research Centre, NMS—NOVA Medical School, FCM—Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisbon, Portugal
| | - Cecília R. C. Calado
- ISEL—Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, Rua Conselheiro Emídio Navarro 1, 1959-007 Lisbon, Portugal
- iBB—Institute for Bioengineering and Biosciences, i4HB—The Associate Laboratory Institute for Health and Bioeconomy, IST—Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| |
Collapse
|
13
|
Tsuchida S, Umemura H, Iizuka K, Yamamoto H, Shimazaki I, Shikata E, Nakayama T. Recent findings on metabolomics and the microbiome of oral bacteria involved in dental caries and periodontal disease. World J Microbiol Biotechnol 2024; 41:11. [PMID: 39690257 DOI: 10.1007/s11274-024-04224-3] [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: 02/07/2024] [Accepted: 12/06/2024] [Indexed: 12/19/2024]
Abstract
Periodontal disease is characterized by bacterial toxins within the oral biofilm surrounding the teeth, leading to gingivitis and the gradual dissolution of the alveolar bone, which supports the teeth. Notably, symptoms in the early stages of the disease are often absent. Similarly, dental caries occurs when oral bacteria metabolize dietary sugars, producing acids that dissolve tooth enamel and dentin. These bacteria are commonly present in the oral cavity of most individuals. Metabolomics, a relatively recent addition to the "omics" research landscape, involves the comprehensive analysis of metabolites in vivo to elucidate pathological mechanisms and accelerate drug discovery. Meanwhile, the term "microbiome" refers to the collection of microorganisms within a specific environmental niche or their collective genomes. The human microbiome plays a critical role in health and disease, influencing a wide array of physiological and pathological processes. Recent advances in microbiome research have identified numerous bacteria implicated in dental caries and periodontal disease. Additionally, studies have uncovered various pathogenic factors associated with these microorganisms. This review focuses on recent findings in metabolomics and the microbiome, specifically targeting oral bacteria linked to dental caries and periodontal disease. We acknowledge the limitation of relying exclusively on the MEDLINE database via PubMed, while excluding other sources such as gray literature, conference proceedings, and clinical practice guidelines.
Collapse
Affiliation(s)
- Sachio Tsuchida
- Divisions of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan
| | - Hiroshi Umemura
- Divisions of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan
| | - Kazuhide Iizuka
- Divisions of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan
| | - Haruka Yamamoto
- Divisions of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan
| | - Isamu Shimazaki
- Divisions of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan
| | - Elisa Shikata
- Divisions of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan
| | - Tomohiro Nakayama
- Divisions of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan.
| |
Collapse
|
14
|
Hua D, Wang L, Li N, Xu X, Yin X. The mediating role of the genetically predicted N6, N6, N6-trimethyllysine levels in the association between HLA DR on CD14- CD16+ monocytes and ankylosing spondylitis. Medicine (Baltimore) 2024; 103:e40892. [PMID: 39686417 DOI: 10.1097/md.0000000000040892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2024] Open
Abstract
This study explores the hidden connection between HLA DR on CD14- CD16+ monocytes and ankylosing spondylitis (AS), with a particular emphasis on investigating and measuring the impact of 1091 blood metabolites as potential mediators. We harnessed the power of summary-level data extracted from a comprehensive genome-wide association study to delve into the intricate relationship between genetically predicted HLA DR on CD14- CD16+ monocytes (3621 cases) and AS (1193 cases and 374,621 controls). Furthermore, we employed a two-step Mendelian randomization (MR) methodology to elucidate the extent to which blood metabolites contribute to the effects observed in CD14- CD16+ monocytes, ultimately influencing the development of AS. This methodological approach provides a comprehensive and rigorous exploration of the interplay between blood metabolites and AS, shedding light on the underlying mechanisms governing this intricate association. Through MR analysis, our investigation revealed an increase in HLA DR on CD14- CD16+ monocytes within plasma, which correspondingly led to a reduction in the incidence of AS. The primary MR analysis yielded an odds ratio of 0.64 with a 95% confidence interval spanning from 0.53 to 0.78, underscoring the protective effect of elevated HLA DR on CD14- CD16+ monocytes against the development of AS. Furthermore, our study found no compelling evidence to suggest that AS exerts any discernible influence on HLA DR on CD14- CD16+ monocytes. Instead, our investigation identified N6, N6, N6-trimethyllysine levels (TML), a blood metabolite, as the sole mediator in the relationship between HLA DR on CD14- CD16+ monocytes and AS. Notably, the genetic prediction of AS mediated by TML accounted for a substantial -2.98% proportion of the observed variance. Our investigation has delineated a causal association between HLA DR on CD14- CD16+ monocytes and AS. Specifically, HLA DR on CD14- CD16+ monocytes exhibited a protective effect against the development of AS. Conversely, AS mediated by TML emerged as a risk factor, though the precise impact of HLA DR on CD14- CD16+ monocytes on AS pathogenesis remains enigmatic. It is imperative to embark on further investigations into potential mediators. In a clinical setting, it is imperative to carefully monitor the patient's HLA DR on CD14- CD16+ monocytes levels.
Collapse
Affiliation(s)
- Danyun Hua
- Fenghua Hospital of Traditional Chinese Medicine, Ningbo, Zhejiang Province, China
| | - Lu Wang
- Fenghua Hospital of Traditional Chinese Medicine, Ningbo, Zhejiang Province, China
| | - Na Li
- Fenghua Hospital of Traditional Chinese Medicine, Ningbo, Zhejiang Province, China
| | - Xiang Xu
- Fenghua Hospital of Traditional Chinese Medicine, Ningbo, Zhejiang Province, China
| | - Xiaohu Yin
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| |
Collapse
|
15
|
Raverdy V, Tavaglione F, Chatelain E, Lassailly G, De Vincentis A, Vespasiani-Gentilucci U, Qadri SF, Caiazzo R, Verkindt H, Saponaro C, Kerr-Conte J, Baud G, Marciniak C, Chetboun M, Oukhouya-Daoud N, Blanck S, Vandel J, Olsson L, Chakaroun R, Gnemmi V, Leteurtre E, Lefebvre P, Haas JT, Yki-Järvinen H, Francque S, Staels B, Le Roux CW, Tremaroli V, Mathurin P, Marot G, Romeo S, Pattou F. Data-driven cluster analysis identifies distinct types of metabolic dysfunction-associated steatotic liver disease. Nat Med 2024; 30:3624-3633. [PMID: 39653777 PMCID: PMC11645276 DOI: 10.1038/s41591-024-03283-1] [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: 01/29/2024] [Accepted: 08/30/2024] [Indexed: 12/15/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) exhibits considerable variability in clinical outcomes. Identifying specific phenotypic profiles within MASLD is essential for developing targeted therapeutic strategies. Here we investigated the heterogeneity of MASLD using partitioning around medoids clustering based on six simple clinical variables in a cohort of 1,389 individuals living with obesity. The identified clusters were applied across three independent MASLD cohorts with liver biopsy (totaling 1,099 participants), and in the UK Biobank to assess the incidence of chronic liver disease, cardiovascular disease and type 2 diabetes. Results unveiled two distinct types of MASLD associated with steatohepatitis on histology and liver imaging. The first cluster, liver-specific, was genetically linked and showed rapid progression of chronic liver disease but limited risk of cardiovascular disease. The second cluster, cardiometabolic, was primarily associated with dysglycemia and high levels of triglycerides, leading to a similar incidence of chronic liver disease but a higher risk of cardiovascular disease and type 2 diabetes. Analyses of samples from 831 individuals with available liver transcriptomics and 1,322 with available plasma metabolomics highlighted that these two types of MASLD exhibited distinct liver transcriptomic profiles and plasma metabolomic signatures, respectively. In conclusion, these data provide preliminary evidence of the existence of two distinct types of clinically relevant MASLD with similar liver phenotypes at baseline, but each with specific underlying biological profiles and different clinical trajectories, suggesting the need for tailored therapeutic strategies.
Collapse
Affiliation(s)
- Violeta Raverdy
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France
| | - Federica Tavaglione
- Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Estelle Chatelain
- US 41 - UAR 2014 - PLBS Bilille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, F-59000, Lille, France
| | - Guillaume Lassailly
- Department of Hepato-Gastroenterology CHU Lille, University of Lille, Inserm INFINITE-U1286, Lille, France
| | - Antonio De Vincentis
- Operative Unit of Internal Medicine, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Internal Medicine, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Umberto Vespasiani-Gentilucci
- Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Sami F Qadri
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Robert Caiazzo
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France
| | - Helene Verkindt
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France
| | - Chiara Saponaro
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
| | - Julie Kerr-Conte
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
| | - Gregory Baud
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France
| | - Camille Marciniak
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France
| | - Mikael Chetboun
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France
| | - Naima Oukhouya-Daoud
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France
| | - Samuel Blanck
- ULR 2694 METRICS: Évaluation des technologies de santé et des pratiques médicales, University of Lille, CHU Lille, F-59000, Lille, France
| | - Jimmy Vandel
- US 41 - UAR 2014 - PLBS Bilille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, F-59000, Lille, France
| | - Lisa Olsson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Rima Chakaroun
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Viviane Gnemmi
- Cancer Heterogeneity Plasticity and Resistance to Therapies, CANTHER-UMR9020-U1277 - CNRS, Inserm, CHU Lille, University of Lille, Lille, France
- Department of Pathology, CHU Lille, University of Lille, Lille, France
| | - Emmanuelle Leteurtre
- Cancer Heterogeneity Plasticity and Resistance to Therapies, CANTHER-UMR9020-U1277 - CNRS, Inserm, CHU Lille, University of Lille, Lille, France
- Department of Pathology, CHU Lille, University of Lille, Lille, France
| | - Philippe Lefebvre
- Nuclear Receptors, Metabolic and Cardiovascular Diseases - U1011, University of Lille, Inserm, CHU Lille, Institut Pasteur Lille, Lille, France
| | - Joel T Haas
- Nuclear Receptors, Metabolic and Cardiovascular Diseases - U1011, University of Lille, Inserm, CHU Lille, Institut Pasteur Lille, Lille, France
| | - Hannele Yki-Järvinen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Sven Francque
- Department of Gastroenterology Hepatology, Antwerp University Hospital, Edegem, Belgium
- InflaMed Centre of Excellence, Laboratory for Experimental Medicine and Paediatrics, Translational Sciences in Inflammation and Immunology, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - Bart Staels
- Nuclear Receptors, Metabolic and Cardiovascular Diseases - U1011, University of Lille, Inserm, CHU Lille, Institut Pasteur Lille, Lille, France
| | - Carel W Le Roux
- Diabetes Complications Research Centre, University College Dublin, Dublin, Ireland
| | - Valentina Tremaroli
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Philippe Mathurin
- Department of Hepato-Gastroenterology CHU Lille, University of Lille, Inserm INFINITE-U1286, Lille, France
| | - Guillemette Marot
- ULR 2694 METRICS: Évaluation des technologies de santé et des pratiques médicales, University of Lille, CHU Lille, F-59000, Lille, France
- MODAL: Models for Data Analysis and Learning, Inria, F-59000, Lille, France
| | - Stefano Romeo
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy.
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden.
- Department of Medicine Huddinge (H7), Karolinska Institutet and University Hospital, Stockholm, Sweden.
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Gothenburg, Sweden.
| | - François Pattou
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France.
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France.
| |
Collapse
|
16
|
Bocher O, Singh A, Huang Y, Võsa U, Reimann E, Arruda A, Barysenska A, Kolde A, Rayner NW, Esko T, Mägi R, Zeggini E. Disentangling the consequences of type 2 diabetes on targeted metabolite profiles using causal inference and interaction QTL analyses. PLoS Genet 2024; 20:e1011346. [PMID: 39625957 PMCID: PMC11642953 DOI: 10.1371/journal.pgen.1011346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 12/13/2024] [Accepted: 10/30/2024] [Indexed: 12/14/2024] Open
Abstract
Circulating metabolite levels have been associated with type 2 diabetes (T2D), but the extent to which T2D affects metabolite levels and their genetic regulation remains to be elucidated. In this study, we investigate the interplay between genetics, metabolomics, and T2D risk in the UK Biobank dataset using the Nightingale panel composed of 249 metabolites, 92% of which correspond to lipids (HDL, IDL, LDL, VLDL) and lipoproteins. By integrating these data with large-scale T2D GWAS from the DIAMANTE meta-analysis through Mendelian randomization analyses, we find 79 metabolites with a causal association to T2D, all spanning lipid-related classes except for Glucose and Tyrosine. Twice as many metabolites are causally affected by T2D liability, spanning almost all tested classes, including branched-chain amino acids. Secondly, using an interaction quantitative trait locus (QTL) analysis, we describe four metabolites consistently replicated in an independent dataset from the Estonian Biobank, for which genetic loci in two different genomic regions show attenuated regulation in T2D cases compared to controls. The significant variants from the interaction QTL analysis are significant QTLs for the corresponding metabolites in the general population but are not associated with T2D risk, pointing towards consequences of T2D on the genetic regulation of metabolite levels. Finally, through differential level analyses, we find 165 metabolites associated with microvascular, macrovascular, or both types of T2D complications, with only a few discriminating between complication classes. Of the 165 metabolites, 40 are not causally linked to T2D in either direction, suggesting biological mechanisms specific to the occurrence of complications. Overall, this work provides a map of the consequences of T2D on Nightingale targeted metabolite levels and on their genetic regulation, enabling a better understanding of the T2D trajectory leading to complications.
Collapse
Affiliation(s)
- Ozvan Bocher
- Institute of Translational Genomics, Helmholtz Zentrum München- German Research Center for Environmental Health, Neuherberg, Germany
| | - Archit Singh
- Institute of Translational Genomics, Helmholtz Zentrum München- German Research Center for Environmental Health, Neuherberg, Germany
- Munich School for Data Science (MUDS), Helmholtz Zentrum München- German Research Center for Environmental Health, Neuherberg, Germany
- Technical University of Munich (TUM), TUM School of Medicine and Health, Graduate School of Experimental Medicine, Munich, Germany
| | - Yue Huang
- Institute of Translational Genomics, Helmholtz Zentrum München- German Research Center for Environmental Health, Neuherberg, Germany
- Technical University of Munich (TUM), TUM School of Medicine and Health, Graduate School of Experimental Medicine, Munich, Germany
| | - Urmo Võsa
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Ene Reimann
- Institute of Translational Genomics, Helmholtz Zentrum München- German Research Center for Environmental Health, Neuherberg, Germany
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Ana Arruda
- Institute of Translational Genomics, Helmholtz Zentrum München- German Research Center for Environmental Health, Neuherberg, Germany
- Munich School for Data Science (MUDS), Helmholtz Zentrum München- German Research Center for Environmental Health, Neuherberg, Germany
- Technical University of Munich (TUM), TUM School of Medicine and Health, Graduate School of Experimental Medicine, Munich, Germany
| | - Andrei Barysenska
- Institute of Translational Genomics, Helmholtz Zentrum München- German Research Center for Environmental Health, Neuherberg, Germany
| | - Anastassia Kolde
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Mathematics and Statistics, University of Tartu, Tartu, Estonia
| | - Nigel W. Rayner
- Institute of Translational Genomics, Helmholtz Zentrum München- German Research Center for Environmental Health, Neuherberg, Germany
| | | | - Tõnu Esko
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Reedik Mägi
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München- German Research Center for Environmental Health, Neuherberg, Germany
- TUM school of medicine and health, Technical University Munich and Klinikum Rechts der Isar, Munich, Germany
| |
Collapse
|
17
|
Nathan Mandal R, Ke J, Hasan Kanika N, Hou X, Zhang Z, Zhang P, Chen H, Zeng C, Chen X, Wang J, Wang C. Gut Microbiome-Driven metabolites influence skin pigmentation in TYRP1 mutant Oujiang Color Common Carp. Gene 2024; 928:148811. [PMID: 39094713 DOI: 10.1016/j.gene.2024.148811] [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: 05/04/2024] [Revised: 06/28/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024]
Abstract
The gut microbiome plays a key role in regulating the gut-skin axis, and host genetics partially influence this regulation. The study investigated the role of gut microbiota and host genetics in the gut-skin axis, focusing on the unusual "coffee-like" color phenotype observed in TYRP1 mutant Oujiang Color Common Carp. We employed comparative high-throughput omics data from wild-type and mutant fish to quantify the influence of both genetics and gut microbes on skin transcriptomic expression and blood metabolites. We found 525 differential metabolites (DMs) and 45 distinct gut microbial genera in TYRP1 mutant fish compared to wild type. Interaction and causal mediation analyses revealed a complex interplay. The TYRP1 mutation likely triggers an inflammatory pathway involving Acinetobacter bacteria, Leukotrience-C4 and Spermine. This inflammatory response appears to be counterbalanced by an anti-inflammatory cardiovascular genetic network. The net effect is the upregulation of COMT, PLG, C2, C3, F10, TDO2, MHC1, and SERPINF2, leading to unusual coffee-like coloration. This study highlights the intricate interplay between gut microbiota, host genetics, and metabolic pathways in shaping complex phenotypes.
Collapse
Affiliation(s)
- Roland Nathan Mandal
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China.
| | - Jing Ke
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China.
| | - Nusrat Hasan Kanika
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China.
| | - Xin Hou
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China.
| | - Zhiyi Zhang
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China.
| | - Penghui Zhang
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China.
| | - Huifan Chen
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China.
| | - Chunxiao Zeng
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China.
| | - Xiaowen Chen
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China.
| | - Jun Wang
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China.
| | - Chenghui Wang
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China.
| |
Collapse
|
18
|
Leclercq S, Ahmed H, Amadieu C, Petit G, Koistinen V, Leyrolle Q, Poncin M, Stärkel P, Kok E, Karhunen PJ, de Timary P, Laye S, Neyrinck AM, Kärkkäinen OK, Hanhineva K, Delzenne N. Blood metabolomic profiling reveals new targets in the management of psychological symptoms associated with severe alcohol use disorder. eLife 2024; 13:RP96937. [PMID: 39611656 PMCID: PMC11606602 DOI: 10.7554/elife.96937] [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] [Indexed: 11/30/2024] Open
Abstract
Background Alcohol use disorder (AUD) is a global health problem with limited therapeutic options. The biochemical mechanisms that lead to this disorder are not yet fully understood, and in this respect, metabolomics represents a promising approach to decipher metabolic events related to AUD. The plasma metabolome contains a plethora of bioactive molecules that reflects the functional changes in host metabolism but also the impact of the gut microbiome and nutritional habits. Methods In this study, we investigated the impact of severe AUD (sAUD), and of a 3-week period of alcohol abstinence, on the blood metabolome (non-targeted LC-MS metabolomics analysis) in 96 sAUD patients hospitalized for alcohol withdrawal. Results We found that the plasma levels of different lipids ((lyso)phosphatidylcholines, long-chain fatty acids), short-chain fatty acids (i.e. 3-hydroxyvaleric acid) and bile acids were altered in sAUD patients. In addition, several microbial metabolites, including indole-3-propionic acid, p-cresol sulfate, hippuric acid, pyrocatechol sulfate, and metabolites belonging to xanthine class (paraxanthine, theobromine and theophylline) were sensitive to alcohol exposure and alcohol withdrawal. 3-Hydroxyvaleric acid, caffeine metabolites (theobromine, paraxanthine, and theophylline) and microbial metabolites (hippuric acid and pyrocatechol sulfate) were correlated with anxiety, depression and alcohol craving. Metabolomics analysis in postmortem samples of frontal cortex and cerebrospinal fluid of those consuming a high level of alcohol revealed that those metabolites can be found also in brain tissue. Conclusions Our data allow the identification of neuroactive metabolites, from interactions between food components and microbiota, which may represent new targets arising in the management of neuropsychiatric diseases such as sAUD. Funding Gut2Behave project was initiated from ERA-NET NEURON network (Joint Transnational Call 2019) and was financed by Academy of Finland, French National Research Agency (ANR-19-NEUR-0003-03) and the Fonds de la Recherche Scientifique (FRS-FNRS; PINT-MULTI R.8013.19, Belgium). Metabolomics analysis of the TSDS samples was supported by grant from the Finnish Foundation for Alcohol Studies.
Collapse
Affiliation(s)
- Sophie Leclercq
- Laboratory of Nutritional Psychiatry, Institute of Neuroscience, UCLouvain, Université catholique de LouvainBrusselsBelgium
| | - Hany Ahmed
- Food Sciences Unit, Department of Life Technologies, University of TurkuTurkuFinland
| | - Camille Amadieu
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de LouvainBrusselsBelgium
| | - Géraldine Petit
- Department of Adult Psychiatry, Cliniques Universitaires Saint-Luc and Institute of Neuroscience, UCLouvain, Université catholique de LouvainBrusselsBelgium
| | - Ville Koistinen
- Food Sciences Unit, Department of Life Technologies, University of TurkuTurkuFinland
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern FinlandKuopioFinland
| | - Quentin Leyrolle
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de LouvainBrusselsBelgium
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286BordeauxFrance
| | - Marie Poncin
- Department of Adult Psychiatry, Cliniques Universitaires Saint-Luc and Institute of Neuroscience, UCLouvain, Université catholique de LouvainBrusselsBelgium
| | - Peter Stärkel
- Department of gastro-enterology, Cliniques Universitaires Saint LucBrusselsBelgium
| | - Eloise Kok
- Department of Pathology, University of HelsinkiHelsinkiFinland
- Faculty of Medicine and Health Technology, Tampere University and Fimlab LaboratoriesTampereFinland
| | - Pekka J Karhunen
- Faculty of Medicine and Health Technology, Tampere University and Fimlab LaboratoriesTampereFinland
| | - Philippe de Timary
- Department of Adult Psychiatry, Cliniques Universitaires Saint-Luc and Institute of Neuroscience, UCLouvain, Université catholique de LouvainBrusselsBelgium
| | - Sophie Laye
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286BordeauxFrance
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de LouvainBrusselsBelgium
| | | | - Kati Hanhineva
- Food Sciences Unit, Department of Life Technologies, University of TurkuTurkuFinland
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern FinlandKuopioFinland
| | - Nathalie Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de LouvainBrusselsBelgium
| |
Collapse
|
19
|
Lu T, Chen Y, Yoshiji S, Ilboudo Y, Forgetta V, Zhou S, Greenwood CMT. Circulating Metabolite Abundances Associated With Risks of Bipolar Disorder, Schizophrenia, and Depression: A Mendelian Randomization Study. Biol Psychiatry 2024; 96:782-791. [PMID: 38705554 DOI: 10.1016/j.biopsych.2024.04.016] [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: 08/30/2023] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND Preventive measures and treatments for psychiatric disorders are limited. Circulating metabolites are potential candidates for biomarker and therapeutic target identification, given their measurability and essential roles in biological processes. METHODS Leveraging large-scale genome-wide association studies, we conducted Mendelian randomization analyses to assess the associations between circulating metabolite abundances and the risks of bipolar disorder, schizophrenia, and depression. Genetic instruments were selected for 94 metabolites measured in the Canadian Longitudinal Study on Aging cohort (N = 8299). We repeated Mendelian randomization analyses based on the UK Biobank, INTERVAL, and EPIC (European Prospective Investigation into Cancer)-Norfolk studies. RESULTS After validating Mendelian randomization assumptions and colocalization evidence, we found that a 1 SD increase in genetically predicted circulating abundances of eicosapentaenoate and docosapentaenoate was associated with odds ratios of 0.72 (95% CI, 0.65-0.79) and 0.63 (95% CI, 0.55-0.72), respectively, for bipolar disorder. Genetically increased Ω-3 unsaturated fatty acids abundance and Ω-3-to-total fatty acids ratio, as well as genetically decreased Ω-6-to-Ω-3 ratio, were negatively associated with the risk of bipolar disorder in the UK Biobank. Genetically increased circulating abundances of 3 N-acetyl-amino acids were associated with an increased risk of schizophrenia with a maximum odds ratio of 1.31 (95% CI, 1.18-1.44) per 1 SD increase. Furthermore, a 1 SD increase in genetically predicted circulating abundance of hypotaurine was associated with an odds ratio of 0.85 (95% CI, 0.78-0.93) for depression. CONCLUSIONS The biological mechanisms that underlie Ω-3 unsaturated fatty acids, NAT8-catalyzed N-acetyl-amino acids, and hypotaurine warrant exploration to identify new biomarkers and potential therapeutic targets.
Collapse
Affiliation(s)
- Tianyuan Lu
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada; Department of Statistical Sciences, University of Toronto, Toronto, Ontario, Canada.
| | - Yiheng Chen
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada; Five Prime Sciences Inc., Montréal, Québec, Canada; Department of Human Genetics, McGill University, Montréal, Québec, Canada
| | - Satoshi Yoshiji
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada; Department of Human Genetics, McGill University, Montréal, Québec, Canada; Kyoto-McGill International Collaborative Program in Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Japan Society for the Promotion of Science, Tokyo, Japan
| | - Yann Ilboudo
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada
| | | | - Sirui Zhou
- Department of Human Genetics, McGill University, Montréal, Québec, Canada; McGill Genome Centre, McGill University, Montréal, Québec, Canada
| | - Celia M T Greenwood
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada; Department of Human Genetics, McGill University, Montréal, Québec, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, Québec, Canada; Gerald Bronfman Department of Oncology, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
20
|
Tanigawa Y, Kellis M. Hypometric genetics: Improved power in genetic discovery by incorporating quality control flags. Am J Hum Genet 2024; 111:2478-2493. [PMID: 39442521 PMCID: PMC11568753 DOI: 10.1016/j.ajhg.2024.09.008] [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: 05/02/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
Balancing the tradeoff between quantity and quality of phenotypic data is critical in omics studies. Measurements below the limit of quantification (BLQ) are often tagged in quality control fields, but these flags are currently underutilized in human genetics studies. Extreme phenotype sampling is advantageous for mapping rare variant effects. We hypothesize that genetic drivers, along with environmental and technical factors, contribute to the presence of BLQ flags. Here, we introduce "hypometric genetics" (hMG) analysis and uncover a genetic basis for BLQ flags, indicating an additional source of genetic signal for genetic discovery, especially from phenotypic extremes. Applying our hMG approach to n = 227,469 UK Biobank individuals with metabolomic profiles, we reveal more than 5% heritability for BLQ flags and report biologically relevant associations, for example, at APOC3, APOA5, and PDE3B loci. For common variants, polygenic scores trained only for BLQ flags predict the corresponding quantitative traits with 91% accuracy, validating the genetic basis. For rare coding variant associations, we find an asymmetric 65.4% higher enrichment of metabolite-lowering associations for BLQ flags, highlighting the impact of putative loss-of-function variants with large effects on phenotypic extremes. Joint analysis of binarized BLQ flags and the corresponding quantitative metabolite measurements improves power in Bayesian rare variant aggregation tests, resulting in an average of 181% more prioritized genes. Our approach is broadly applicable to omics profiling. Overall, our results underscore the benefit of integrating quality control flags and quantitative measurements and highlight the advantage of joint analysis of population-based samples and phenotypic extremes in human genetics studies.
Collapse
Affiliation(s)
- Yosuke Tanigawa
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Manolis Kellis
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
21
|
Rein M, Elkan M, Godneva A, Dolev NC, Segal E. Sex-specific dietary habits and their association with weight change in healthy adults. BMC Med 2024; 22:512. [PMID: 39501340 PMCID: PMC11539530 DOI: 10.1186/s12916-024-03730-3] [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: 11/22/2023] [Accepted: 10/26/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Dietary intake plays a pivotal role in the prevalence and management of obesity. While women and men exhibit differences in dietary habits and food-related behaviors, sex-based weight loss recommendations are lacking. This study aims to examine the impact of specific foods and food categories on weight reduction in men and women over a two-year period. METHODS A total of 8,548 participants from the 10K cohort, from 2019 to 2023, were included in the analysis (53.1% women, mean age 51.7 years). Anthropometric measurements and laboratory results were collected at baseline and at the two-year follow-up visit. Dietary assessment was based on daily food intake digitally logged through an application for at least 3 consecutive days at both timepoints. We compared intake of macronutrients, micronutrients, food groups and daily energy consumption between sex and body mass index (BMI) categories at baseline and weight change categories at follow-up. Using linear regression, we assessed the associations between food categories or specific foods and BMI at baseline as well as weight change percentage at follow-up. RESULTS Dietary habits varied by BMI and sex. Women and men living with obesity (BMI > 30 kg/m2) reported a greater intake of animal-based protein and lower intake of plant-based proteins and fats at baseline, as compared to participants with normal weight. In linear regression models predicting two-year weight change, including age, income, and baseline weight, the explained variance was 5.6% for men and 5.8% for women. Adding food categories and specific foods increased the explained variance to 20.6% for men and 17.5% for women. Weight reduction in men was linked to daily consumption of an egg (1.2% decrease) and beef (1.5% decrease), while in women, the most pronounced reductions were associated with an apple (1.2% decrease) and cashew nuts (3.4% decrease). Notably, total energy intake changes significantly impacted weight outcomes only in women. CONCLUSIONS Sex-specific dietary habits significantly influence weight change over time. In men, weight loss was primarily associated with the addition of animal-based protein, while in women, it was linked to caloric deficit and plant-based fat, suggesting that sex-based nutritional interventions may demonstrate greater efficacy. TRIAL REGISTRATION NCT05817734 (retrospectively registered January 31, 2023).
Collapse
Affiliation(s)
- Michal Rein
- Department of Computer Science and Applied Mathematics, The Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Matan Elkan
- Department of Internal Medicine A, Shamir Medical Center (Assaf Harofeh), Zerifin, Israel
| | - Anastasia Godneva
- Department of Computer Science and Applied Mathematics, The Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Noa Cohen Dolev
- Department of Computer Science and Applied Mathematics, The Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Eran Segal
- Department of Computer Science and Applied Mathematics, The Weizmann Institute of Science, Rehovot, Israel.
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
22
|
Debik J, Mrowiec K, Kurczyk A, Widłak P, Jelonek K, Bathen TF, Giskeødegård GF. Sources of variation in the serum metabolome of female participants of the HUNT2 study. Commun Biol 2024; 7:1450. [PMID: 39506131 PMCID: PMC11541904 DOI: 10.1038/s42003-024-07137-x] [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: 03/18/2024] [Accepted: 10/24/2024] [Indexed: 11/08/2024] Open
Abstract
The aim of this study was to explore the intricate relationship between serum metabolomics and lifestyle factors, shedding light on their impact on health in the context of breast cancer risk. Detailed metabolic profiles of 2283 female participants in the Trøndelag Health Study (HUNT study) were obtained through nuclear magnetic resonance (NMR) spectroscopy and mass spectrometry (MS).We show that lifestyle-related variables can explain up to 30% of the variance in individual metabolites. Age and obesity were the primary factors affecting the serum metabolic profile, both associated with increased levels of triglyceride-rich very low-density lipoproteins (VLDL) and intermediate-density lipoproteins (IDL), amino acids and glycolysis-related metabolites, and decreased levels of high-density lipoproteins (HDL). Moreover, factors like hormonal changes associated with menstruation and contraceptive use or education level influence the metabolite levels.Participants were clustered into three distinct clusters based on lifestyle-related factors, revealing metabolic similarities between obese and older individuals, despite diverse lifestyle factors, suggesting accelerated metabolic aging with obesity. Our results show that metabolic associations to cancer risk may partly be explained by modifiable lifestyle factors.
Collapse
Affiliation(s)
- Julia Debik
- Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Katarzyna Mrowiec
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Agata Kurczyk
- Department of Biostatistics and Bioinformatics, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Piotr Widłak
- 2nd Radiology Department, Medical University of Gdańsk, Gdańsk, Poland
| | - Karol Jelonek
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Tone F Bathen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Radiology and Nuclear Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Guro F Giskeødegård
- Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway.
- Clinic of Surgery, St. Olav's University Hospital, Trondheim, Norway.
| |
Collapse
|
23
|
Hu FF, Pan SY, Chu JY, Liu JJ, Duan TT, Luo Y, Zhou W, Wang ZM, Liu W, Zeng Y. Xanthohumol Protects Against Neuronal Excitotoxicity and Mitochondrial Dysfunction in APP/PS1 Mice: An Omics-Based Study. Nutrients 2024; 16:3754. [PMID: 39519590 PMCID: PMC11548031 DOI: 10.3390/nu16213754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Neuronal excitotoxicity and metabolic decline, which begin in the early stages of Alzheimer's disease (AD), pose challenges for effective amelioration. Our previous work suggested that the natural compound xanthohumol, the most abundant prenylated flavonoid in hops, prevents memory deficits in APP/PS1 mice; however, the underlying mechanisms remain unclear. Methods: This study utilized APP/PS1 mice and cutting-edge omics techniques to investigate the effects of xanthohumol on hippocampal proteome, serum metabolome, and microbiome. Results: Our findings revealed that xanthohumol reduces the postsynaptic overexpression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, N-methyl-D-aspartate, and metabotropic glutamate receptors, but enhances ATP synthesis and mitophagy in the young AD hippocampus. Further mechanistic analyses suggested systemic regulatory effects, particularly on the decreasing glutamate synthesis in the blood and intestines of AD mice following xanthohumol administration. Conclusions: These results underscore the potential of xanthohumol in mitigating AD pathology through multifaceted mechanisms, sparking interest and curiosity in its preventive and therapeutic potential in AD.
Collapse
Affiliation(s)
- Fei-Fei Hu
- Hubei Provincial Clinical Research Center for Alzheimer’s Disease, Wuhan University of Science and Technology, Wuhan 430065, China; (F.-F.H.); (S.-Y.P.); (J.-Y.C.); (T.-T.D.); (Y.L.); (W.Z.); (Z.-M.W.)
- Brain Science and Advanced Technology Institute, Medical School, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Shi-Yao Pan
- Hubei Provincial Clinical Research Center for Alzheimer’s Disease, Wuhan University of Science and Technology, Wuhan 430065, China; (F.-F.H.); (S.-Y.P.); (J.-Y.C.); (T.-T.D.); (Y.L.); (W.Z.); (Z.-M.W.)
- Brain Science and Advanced Technology Institute, Medical School, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Jin-Yu Chu
- Hubei Provincial Clinical Research Center for Alzheimer’s Disease, Wuhan University of Science and Technology, Wuhan 430065, China; (F.-F.H.); (S.-Y.P.); (J.-Y.C.); (T.-T.D.); (Y.L.); (W.Z.); (Z.-M.W.)
- Brain Science and Advanced Technology Institute, Medical School, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Jian-Jun Liu
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China;
| | - Ting-Ting Duan
- Hubei Provincial Clinical Research Center for Alzheimer’s Disease, Wuhan University of Science and Technology, Wuhan 430065, China; (F.-F.H.); (S.-Y.P.); (J.-Y.C.); (T.-T.D.); (Y.L.); (W.Z.); (Z.-M.W.)
- Brain Science and Advanced Technology Institute, Medical School, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yu Luo
- Hubei Provincial Clinical Research Center for Alzheimer’s Disease, Wuhan University of Science and Technology, Wuhan 430065, China; (F.-F.H.); (S.-Y.P.); (J.-Y.C.); (T.-T.D.); (Y.L.); (W.Z.); (Z.-M.W.)
- Brain Science and Advanced Technology Institute, Medical School, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Wen Zhou
- Hubei Provincial Clinical Research Center for Alzheimer’s Disease, Wuhan University of Science and Technology, Wuhan 430065, China; (F.-F.H.); (S.-Y.P.); (J.-Y.C.); (T.-T.D.); (Y.L.); (W.Z.); (Z.-M.W.)
- Brain Science and Advanced Technology Institute, Medical School, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Zhi-Ming Wang
- Hubei Provincial Clinical Research Center for Alzheimer’s Disease, Wuhan University of Science and Technology, Wuhan 430065, China; (F.-F.H.); (S.-Y.P.); (J.-Y.C.); (T.-T.D.); (Y.L.); (W.Z.); (Z.-M.W.)
- Brain Science and Advanced Technology Institute, Medical School, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Wei Liu
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China;
| | - Yan Zeng
- Hubei Provincial Clinical Research Center for Alzheimer’s Disease, Wuhan University of Science and Technology, Wuhan 430065, China; (F.-F.H.); (S.-Y.P.); (J.-Y.C.); (T.-T.D.); (Y.L.); (W.Z.); (Z.-M.W.)
- Brain Science and Advanced Technology Institute, Medical School, Wuhan University of Science and Technology, Wuhan 430065, China
| |
Collapse
|
24
|
Morais LH, Boktor JC, MahmoudianDehkordi S, Kaddurah-Daouk R, Mazmanian SK. α-synuclein overexpression and the microbiome shape the gut and brain metabolome in mice. NPJ Parkinsons Dis 2024; 10:208. [PMID: 39477976 PMCID: PMC11525669 DOI: 10.1038/s41531-024-00816-w] [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: 06/07/2024] [Accepted: 10/10/2024] [Indexed: 11/02/2024] Open
Abstract
Pathological forms of α-synuclein contribute to synucleinopathies, including Parkinson's disease (PD). Most cases of PD arise from gene-environment interactions. Microbiome composition is altered in PD, and gut bacteria are causal to symptoms in animal models. We quantitatively profiled nearly 630 metabolites in the gut, plasma, and brain of α-synuclein-overexpressing (ASO) mice, compared to wild-type (WT) animals, and comparing germ-free (GF) to specific pathogen-free (SPF) animals (n = 5 WT-SPF; n = 6 ASO-SPF; n = 6 WT-GF; n = 6 ASO-GF). Many differentially expressed metabolites in ASO mice are also dysregulated in human PD patients, including amine oxides, bile acids and indoles. The microbial metabolite trimethylamine N-oxide (TMAO) strongly correlates from the gut to the plasma to the brain in mice, notable since TMAO is elevated in the blood and cerebrospinal fluid of PD patients. These findings uncover broad metabolomic changes that are influenced by the intersection of host genetics and microbiome in a mouse model of PD.
Collapse
Affiliation(s)
- Livia H Morais
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Joseph C Boktor
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | | | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA.
- Duke Institute of Brain Sciences, Duke University, Durham, NC, USA.
- Department of Medicine, Duke University, Durham, NC, USA.
| | - Sarkis K Mazmanian
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
| |
Collapse
|
25
|
Ren D, Li Y, Zhang G, Li T, Liu Z. Lipid metabolic profiling and diagnostic model development for hyperlipidemic acute pancreatitis. Front Physiol 2024; 15:1457349. [PMID: 39512473 PMCID: PMC11540618 DOI: 10.3389/fphys.2024.1457349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 09/23/2024] [Indexed: 11/15/2024] Open
Abstract
Introduction Hyperlipidemic acute pancreatitis (HLAP) is a form of pancreatitis induced by hyperlipidemia, posing significant diagnostic challenges due to its complex lipid metabolism disturbances. Methods This study compared the serum lipid profiles of HLAP patients with those of a healthy cohort using ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). Orthogonal partial least squares discriminant analysis (OPLS-DA) was applied to identify distinct lipid metabolites. Logistic regression and LASSO regression were used to develop a diagnostic model based on the lipid molecules identified. Results A total of 393 distinct lipid metabolites were detected, impacting critical pathways such as fatty acid, sphingolipid, and glycerophospholipid metabolism. Five specific lipid molecules were selected to construct a diagnostic model, which achieved an area under the curve (AUC) of 1 in the receiver operating characteristic (ROC) analysis, indicating outstanding diagnostic accuracy. Discussion These findings highlight the importance of lipid metabolism disturbances in HLAP. The identified lipid molecules could serve as valuable biomarkers for HLAP diagnosis, offering potential for more accurate and early detection.
Collapse
Affiliation(s)
- Dongmei Ren
- Department of Hepatobiliary Surgery II, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yong Li
- Department of Hepatobiliary Surgery II, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Guangnian Zhang
- Department of Hepatobiliary Surgery II, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Tiantian Li
- Department of Hepatobiliary Surgery II, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Zhenglong Liu
- School of Basic Medical Sciences and Forensic Medicine, North Sichuan Medical College, Nanchong, China
| |
Collapse
|
26
|
Jaagura M, Kronberg J, Reigo A, Aasmets O, Nikopensius T, Võsa U, Bomba L, Estrada K, Wuster A, Esko T, Org E. Comorbidities confound metabolomics studies of human disease. Sci Rep 2024; 14:24810. [PMID: 39438584 PMCID: PMC11496539 DOI: 10.1038/s41598-024-75556-1] [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: 05/14/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024] Open
Abstract
The co-occurrence of multiple chronic conditions, termed multimorbidity, presents an expanding global health challenge, demanding effective diagnostics and treatment strategies. Chronic ailments such as obesity, diabetes, and cardiovascular diseases have been linked to metabolites interacting between the host and microbiota. In this study, we investigated the impact of co-existing conditions on risk estimations for 1375 plasma metabolites in 919 individuals from population-based Estonian Biobank cohort using liquid chromatography mass spectrometry (LC-MS) method. We leveraged annually linked national electronic health records (EHRs) data to delineate comorbidities in incident cases and controls for the 14 common chronic conditions. Among the 254 associations observed across 13 chronic conditions, we primarily identified disease-specific risk factors (92%, 217/235), with most predictors (93%, 219/235) found to be related to the gut microbiome upon cross-referencing recent literature data. Accounting for comorbidities led to a reduction of common metabolite predictors across various conditions. In conclusion, our study underscores the potential of utilizing biobank-linked retrospective and prospective EHRs for the disease-specific profiling of diverse multifactorial chronic conditions.
Collapse
Affiliation(s)
- Madis Jaagura
- Institute of Genomics, Estonian Genome Centre, University of Tartu, Tartu, Estonia
| | - Jaanika Kronberg
- Institute of Genomics, Estonian Genome Centre, University of Tartu, Tartu, Estonia
| | - Anu Reigo
- Institute of Genomics, Estonian Genome Centre, University of Tartu, Tartu, Estonia
| | - Oliver Aasmets
- Institute of Genomics, Estonian Genome Centre, University of Tartu, Tartu, Estonia
| | - Tiit Nikopensius
- Institute of Genomics, Estonian Genome Centre, University of Tartu, Tartu, Estonia
| | - Urmo Võsa
- Institute of Genomics, Estonian Genome Centre, University of Tartu, Tartu, Estonia
| | | | | | | | - Tõnu Esko
- Institute of Genomics, Estonian Genome Centre, University of Tartu, Tartu, Estonia
| | - Elin Org
- Institute of Genomics, Estonian Genome Centre, University of Tartu, Tartu, Estonia.
| |
Collapse
|
27
|
Harrison BR, Partida-Aguilar M, Marye A, Djukovic D, Kauffman M, Dunbar MD, Mariner BL, McCoy BM, Algavi YM, Muller E, Baum S, Bamberger T, Raftery D, Creevy KE, Avery A, Borenstein E, Snyder-Mackler N, Promislow DE. Protein catabolites as blood-based biomarkers of aging physiology: Findings from the Dog Aging Project. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.17.618956. [PMID: 39484426 PMCID: PMC11526923 DOI: 10.1101/2024.10.17.618956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Our understanding of age-related physiology and metabolism has grown through the study of systems biology, including transcriptomics, single-cell analysis, proteomics and metabolomics. Studies in lab organisms in controlled environments, while powerful and complex, fall short of capturing the breadth of genetic and environmental variation in nature. Thus, there is now a major effort in geroscience to identify aging biomarkers and to develop aging interventions that might be applied across the diversity of humans and other free-living species. To meet this challenge, the Dog Aging Project (DAP) is designed to identify cross-sectional and longitudinal patterns of aging in complex systems, and how these are shaped by the diversity of genetic and environmental variation among companion dogs. Here we surveyed the plasma metabolome from the first year of sampling of the Precision Cohort of the DAP. By incorporating extensive metadata and whole genome sequencing information, we were able to overcome the limitations inherent in breed-based estimates of genetic and physiological effects, and to probe the physiological and dietary basis of the age-related metabolome. We identified a significant effect of age on approximately 40% of measured metabolites. Among other insights, we discovered a potentially novel biomarker of age in the post-translationally modified amino acids (ptmAAs). The ptmAAs, which can only be generated by protein hydrolysis, covaried both with age and with other biomarkers of amino acid metabolism, and in a way that was robust to diet. Clinical measures of kidney function mediated about half of the higher ptmAA levels in older dogs. This work identifies ptmAAs as robust indicators of age in dogs, and points to kidney function as a physiological mediator of age-associated variation in the plasma metabolome.
Collapse
Affiliation(s)
- Benjamin R. Harrison
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Maria Partida-Aguilar
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Abbey Marye
- University of Utah, Department of Microbiology and Immunology, Salt Lake City, UT, USA
| | - Danijel Djukovic
- Center for Studies in Ecology and Demography, University of Washington, Seattle, WA, USA
| | - Mandy Kauffman
- Center for Studies in Ecology and Demography, University of Washington, Seattle, WA, USA
| | - Matthew D. Dunbar
- Center for Studies in Ecology and Demography, University of Washington, Seattle, WA, USA
| | | | - Brianah M. McCoy
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Yadid M. Algavi
- Department of Clinical Microbiology and Immunology, Tel Aviv University, Tel Aviv, Israel
| | - Efrat Muller
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Shiri Baum
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Tal Bamberger
- Department of Clinical Microbiology and Immunology, Tel Aviv University, Tel Aviv, Israel
| | - Dan Raftery
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
| | - Kate E. Creevy
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | | | - Anne Avery
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, CO, USA
| | - Elhanan Borenstein
- Department of Clinical Microbiology and Immunology, Tel Aviv University, Tel Aviv, Israel
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | | | - Daniel E. Promislow
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| |
Collapse
|
28
|
Liu M, Wen Y. Point-of-care testing for early-stage liver cancer diagnosis and personalized medicine: Biomarkers, current technologies and perspectives. Heliyon 2024; 10:e38444. [PMID: 39397977 PMCID: PMC11470528 DOI: 10.1016/j.heliyon.2024.e38444] [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: 04/09/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024] Open
Abstract
Liver cancer is a highly prevalent and lethal form of cancer worldwide. In the absence of early diagnosis, treatment options for this disease are severely restricted. Recent advancements in genomics and bioinformatics have facilitated the discovery of a multitude of novel biomarkers that accurately depict an individual's disease diagnosis, progression, and treatment response. Leveraging these breakthroughs, personalized medicine employs an individual's biomarker profile to enable early detection of liver cancer and inform decisions regarding treatment selection, dosage determination, and prognosis assessment. The current lack of readily applicable, timely, and economically viable tools for biomarker analysis has hindered the incorporation of personalized medicine into regular clinical procedures. Over the past decade, significant advancements have been achieved in the field of molecular point-of-care testing (POCT) and amplification techniques, leading to substantial improvements in the diagnosis of liver cancer and the implementation of precision medicine. Instrument-free PCR technology or plasma PCR technology can shorten the complex procedure of in vitro detection of nucleic acid-based biomarkers. Also, compared to traditional ELISA, various nanomaterials modified with monoclonal antibodies to target proteins for recognition, capture, and detection have improved the efficiency of protein-based biomarker detection. These advances have reduced the time and cost of clinical detection of early-stage hepatocellular carcinoma and improved the efficiency of timely diagnosis and survival of suspected patients while reducing unnecessary testing costs and procedures. This review aims to provide a comprehensive overview of the current and emerging biomarkers employed in the early detection of liver cancer, as well as the advancements in point-of-care molecular testing technology and platforms. The primary objective is to assess their potential in facilitating the implementation of personalized medicine. This review ultimately revealed that the diagnosis of early-stage hepatocellular carcinoma not only requires sensitive biomarkers, but its various modifications and changes during the progression of cirrhosis to early-stage hepatocellular carcinoma will be a greater focus of our attention in the future. The rapid development of POCT has facilitated the opportunity to readily detect liver cancer in the general population in the future, and the integration of multi-pathway multiplexing and intelligent algorithms has improved the sensitivity and accuracy of early liver cancer biomarker detection. It is expected that the integration of point-of-care technology will be instrumental in the widespread adoption of personalized medicine in the foreseeable future.
Collapse
Affiliation(s)
- Mengxiang Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, China
| | - Yanrong Wen
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| |
Collapse
|
29
|
Hang Y, Chen Z, Ren J, Wang Y, Zhu K, Zhu Q. Causal relationship between metabolites and embolic stroke: based on Mendelian randomization and metabolomics. Front Neurol 2024; 15:1460852. [PMID: 39463788 PMCID: PMC11502368 DOI: 10.3389/fneur.2024.1460852] [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: 07/07/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024] Open
Abstract
Purpose This research employed Mendelian randomization (MR) methods to explore whether metabolites are causally associated with embolic stroke of undetermined source (ESUS). Methods Genome-Wide Association Study (GWAS) data regarding metabolites and ESUS were downloaded from the database. Metabolites were employed as exposure factors, ESUS served as the outcome variable, and single nucleotide polymorphisms (SNPs) exhibiting significant association with ESUS were chosen as instrumental variables. The causal association between exposure factor metabolites and the outcome variable ESUS was assessed using two methods: MR-Egger regression and inverse variance-weighted (IVW) analysis. Results A causal relationship was observed between X-11593--O-methylascorbate* and ESUS, indicating a protective factor. Moreover, a causal relationship was identified between cholesterol esters in large very-low-density lipoprotein (VLDL), cholesterol esters in medium low-density lipoprotein (LDL), concentration of medium LDL particles, phospholipids in medium LDL, phenylalanine, total cholesterol in small LDL, total lipids in medium LDL and ESUS, representing risk factor. Funnel plots exhibited a symmetrical distribution of SNPs, while pleiotropic tests (p > 0.05) and leave-one-out tests indicated that the results were relatively stable. Conclusion Metabolites are causally associated with ESUS. LDL and VLDL-related metabolites are identified as risk factors for ESUS.
Collapse
Affiliation(s)
- Ying Hang
- Radiography Center - Interventional Catheter Room, Yixing People’s Hospital, Yixing, China
| | - Zanhao Chen
- Department of Medicine, Xinglin College, Nantong University, Nantong, China
| | - Jiayi Ren
- Department of Medicine, Xinglin College, Nantong University, Nantong, China
| | - Yu Wang
- Department of Medicine, Xinglin College, Nantong University, Nantong, China
| | - Kangle Zhu
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Qianhong Zhu
- Department of Emergency Medicine, Yixing People’s Hospital, Yixing, China
| |
Collapse
|
30
|
Vich Vila A, Zhang J, Liu M, Faber KN, Weersma RK. Untargeted faecal metabolomics for the discovery of biomarkers and treatment targets for inflammatory bowel diseases. Gut 2024; 73:1909-1920. [PMID: 39002973 PMCID: PMC11503092 DOI: 10.1136/gutjnl-2023-329969] [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: 05/11/2024] [Accepted: 06/23/2024] [Indexed: 07/15/2024]
Abstract
The gut microbiome has been recognised as a key component in the pathogenesis of inflammatory bowel diseases (IBD), and the wide range of metabolites produced by gut bacteria are an important mechanism by which the human microbiome interacts with host immunity or host metabolism. High-throughput metabolomic profiling and novel computational approaches now allow for comprehensive assessment of thousands of metabolites in diverse biomaterials, including faecal samples. Several groups of metabolites, including short-chain fatty acids, tryptophan metabolites and bile acids, have been associated with IBD. In this Recent Advances article, we describe the contribution of metabolomics research to the field of IBD, with a focus on faecal metabolomics. We discuss the latest findings on the significance of these metabolites for IBD prognosis and therapeutic interventions and offer insights into the future directions of metabolomics research.
Collapse
Affiliation(s)
- Arnau Vich Vila
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Jingwan Zhang
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong (SAR), People's Republic of China
- Microbiota I-Center (MagIC), Hong Kong (SAR), People's Republic of China
| | - Moting Liu
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Klaas Nico Faber
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
31
|
Spencer KD, Bline H, Chen HJ, Verosky BG, Hilt ME, Jaggers RM, Gur TL, Mathé EA, Bailey MT. Modulation of anxiety-like behavior in galactooligosaccharide-fed mice: A potential role for bacterial tryptophan metabolites and reduced microglial reactivity. Brain Behav Immun 2024; 121:229-243. [PMID: 39067620 DOI: 10.1016/j.bbi.2024.07.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 07/02/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024] Open
Abstract
Prebiotic galactooligosaccharides (GOS) reduce anxiety-like behaviors in mice and humans. However, the biological pathways behind these behavioral changes are not well understood. To begin to study these pathways, we utilized C57BL/6 mice that were fed a standard diet with or without GOS supplementation for 3 weeks prior to testing on the open field. After behavioral testing, colonic contents and serum were collected for bacteriome (16S rRNA gene sequencing, colonic contents only) and metabolome (UPLC-MS, colonic contents and serum data) analyses. As expected, GOS significantly reduced anxiety-like behavior (i.e., increased time in the center) and decreased cytokine gene expression (Tnfa and Ccl2) in the prefrontal cortex. Notably, time in the center of the open field was significantly correlated with serum methyl-indole-3-acetic acid (methyl-IAA). This metabolite is a methylated form of indole-3-acetic acid (IAA) that is derived from bacterial metabolism of tryptophan. Sequencing analyses showed that GOS significantly increased Lachnospiraceae UCG006 and Akkermansia; these taxa are known to metabolize both GOS and tryptophan. To determine the extent to which methyl-IAA can affect anxiety-like behavior, mice were intraperitoneally injected with methyl-IAA. Mice given methyl-IAA had a reduction in anxiety-like behavior in the open field, along with lower Tnfa in the prefrontal cortex. Methyl-IAA was also found to reduce TNF-α (as well as CCL2) production by LPS-stimulated BV2 microglia. Together, these data support a novel pathway through which GOS reduces anxiety-like behaviors in mice and suggests that the bacterial metabolite methyl-IAA reduces microglial cytokine and chemokine production, which in turn reduces anxiety-like behavior.
Collapse
Affiliation(s)
- Kyle D Spencer
- Graduate Partnership Program, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA; Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA; Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Heather Bline
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Helen J Chen
- Medical Scientist Training Program, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Branden G Verosky
- Medical Scientist Training Program, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Miranda E Hilt
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Robert M Jaggers
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Tamar L Gur
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Psychiatry & Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ewy A Mathé
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Michael T Bailey
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Oral and GI Research Affinity Group, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
32
|
Carrasco-Zanini J, Wheeler E, Uluvar B, Kerrison N, Koprulu M, Wareham NJ, Pietzner M, Langenberg C. Mapping biological influences on the human plasma proteome beyond the genome. Nat Metab 2024; 6:2010-2023. [PMID: 39327534 PMCID: PMC11496106 DOI: 10.1038/s42255-024-01133-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 08/23/2024] [Indexed: 09/28/2024]
Abstract
Broad-capture proteomic platforms now enable simultaneous assessment of thousands of plasma proteins, but most of these are not actively secreted and their origins are largely unknown. Here we integrate genomic with deep phenomic information to identify modifiable and non-modifiable factors associated with 4,775 plasma proteins in ~8,000 mostly healthy individuals. We create a data-driven map of biological influences on the human plasma proteome and demonstrate segregation of proteins into clusters based on major explanatory factors. For over a third (N = 1,575) of protein targets, joint genetic and non-genetic factors explain 10-77% of the variation in plasma (median 19.88%, interquartile range 14.01-31.09%), independent of technical factors (median 2.48%, interquartile range 0.78-6.41%). Together with genetically anchored causal inference methods, our map highlights potential causal associations between modifiable risk factors and plasma proteins for hundreds of protein-disease associations, for example, COL6A3, which possibly mediates the association between reduced kidney function and cardiovascular disease. We provide a map of biological and technical influences on the human plasma proteome to help contextualize findings from proteomic studies.
Collapse
Affiliation(s)
- Julia Carrasco-Zanini
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Eleanor Wheeler
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Burulça Uluvar
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Nicola Kerrison
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Mine Koprulu
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Maik Pietzner
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Claudia Langenberg
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK.
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany.
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK.
| |
Collapse
|
33
|
Ruze R, Chen Y, Song J, Xu R, Yin X, Xu Q, Wang C, Zhao Y. Enhanced cytokine signaling and ferroptosis defense interplay initiates obesity-associated pancreatic ductal adenocarcinoma. Cancer Lett 2024; 601:217162. [PMID: 39127339 DOI: 10.1016/j.canlet.2024.217162] [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: 05/13/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Obesity is a significant risk factor for various cancers, including pancreatic cancer (PC), but the underlying mechanisms are still unclear. In our study, pancreatic ductal epithelial cells were cultured using serum from human subjects with diverse metabolic statuses, revealing that serum from patients with obesity alters inflammatory cytokine signaling and ferroptosis, where a mutual enhancement between interleukin 34 (IL-34) expression and ferroptosis defense was observed in these cells. Notably, oncogenic KRASG12D amplified their interaction and this leads to the initiation of pancreatic ductal adenocarcinoma (PDAC) in diet-induced obese mice via macrophage-mediated immunosuppression. Single-cell RNA sequencing (scRNA-seq) of human samples showed that cytokine signaling, ferroptosis defense, and immunosuppression are correlated with the patients' body mass index (BMI) during PDAC progression. Our findings provide a mechanistic link between obesity, inflammation, ferroptosis defense, and pancreatic cancer, suggesting novel therapeutic targets for the prevention and treatment of obesity-associated PDAC.
Collapse
Affiliation(s)
- Rexiati Ruze
- Department of General Surgery, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS&PUMC), Beijing, 100730, China; Department of Hepatobiliary and Echinococcosis Surgery, Digestive and Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China; General Surgery Laboratory, Key Laboratory of Research in Pancreatic Tumor, CAMS, Beijing, 100023, China; National Science and Technology Key Infrastructure on Translational Medicine in PUMCH, Beijing, 100023, China; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH, CAMS&PUMC, Beijing, 100023, China.
| | - Yuan Chen
- Department of General Surgery, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS&PUMC), Beijing, 100730, China; General Surgery Laboratory, Key Laboratory of Research in Pancreatic Tumor, CAMS, Beijing, 100023, China; National Science and Technology Key Infrastructure on Translational Medicine in PUMCH, Beijing, 100023, China; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH, CAMS&PUMC, Beijing, 100023, China.
| | - Jianlu Song
- Department of General Surgery, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS&PUMC), Beijing, 100730, China; General Surgery Laboratory, Key Laboratory of Research in Pancreatic Tumor, CAMS, Beijing, 100023, China; National Science and Technology Key Infrastructure on Translational Medicine in PUMCH, Beijing, 100023, China; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH, CAMS&PUMC, Beijing, 100023, China.
| | - Ruiyuan Xu
- Department of General Surgery, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS&PUMC), Beijing, 100730, China; General Surgery Laboratory, Key Laboratory of Research in Pancreatic Tumor, CAMS, Beijing, 100023, China; National Science and Technology Key Infrastructure on Translational Medicine in PUMCH, Beijing, 100023, China; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH, CAMS&PUMC, Beijing, 100023, China.
| | - Xinpeng Yin
- Department of General Surgery, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS&PUMC), Beijing, 100730, China; General Surgery Laboratory, Key Laboratory of Research in Pancreatic Tumor, CAMS, Beijing, 100023, China; National Science and Technology Key Infrastructure on Translational Medicine in PUMCH, Beijing, 100023, China; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH, CAMS&PUMC, Beijing, 100023, China.
| | - Qiang Xu
- Department of General Surgery, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS&PUMC), Beijing, 100730, China; General Surgery Laboratory, Key Laboratory of Research in Pancreatic Tumor, CAMS, Beijing, 100023, China; National Science and Technology Key Infrastructure on Translational Medicine in PUMCH, Beijing, 100023, China; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH, CAMS&PUMC, Beijing, 100023, China.
| | - Chengcheng Wang
- General Surgery Laboratory, Key Laboratory of Research in Pancreatic Tumor, CAMS, Beijing, 100023, China; National Science and Technology Key Infrastructure on Translational Medicine in PUMCH, Beijing, 100023, China; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH, CAMS&PUMC, Beijing, 100023, China; Medical Research Center, PUMCH, CAMS&PUMC, Beijing, 100730, China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS&PUMC), Beijing, 100730, China; General Surgery Laboratory, Key Laboratory of Research in Pancreatic Tumor, CAMS, Beijing, 100023, China; National Science and Technology Key Infrastructure on Translational Medicine in PUMCH, Beijing, 100023, China; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH, CAMS&PUMC, Beijing, 100023, China.
| |
Collapse
|
34
|
Franks PW, Coral DE. Causal drivers of human proteome variation in health and disease. Nat Metab 2024; 6:1854-1855. [PMID: 39327533 DOI: 10.1038/s42255-024-01138-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Affiliation(s)
- Paul W Franks
- Department of Clinical Sciences, Lund University, Helsingborg Hospital, Helsingborg, Sweden.
| | - Daniel E Coral
- Department of Clinical Sciences, Lund University, Helsingborg Hospital, Helsingborg, Sweden
| |
Collapse
|
35
|
Martínez-González MA, Planes FJ, Ruiz-Canela M, Toledo E, Estruch R, Salas-Salvadó J, Valdés-Más R, Mena P, Castañer O, Fitó M, Clish C, Landberg R, Wittenbecher C, Liang L, Guasch-Ferré M, Lamuela-Raventós RM, Wang DD, Forouhi N, Razquin C, Hu FB. Recent advances in precision nutrition and cardiometabolic diseases. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2024:S1885-5857(24)00279-2. [PMID: 39357800 DOI: 10.1016/j.rec.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024]
Abstract
A growing body of research on nutrition omics has led to recent advances in cardiovascular disease epidemiology and prevention. Within the PREDIMED trial, significant associations between diet-related metabolites and cardiovascular disease were identified, which were subsequently replicated in independent cohorts. Some notable metabolites identified include plasma levels of ceramides, acyl-carnitines, branched-chain amino acids, tryptophan, urea cycle pathways, and the lipidome. These metabolites and their related pathways have been associated with incidence of both cardiovascular disease and type 2 diabetes. Future directions in precision nutrition research include: a) developing more robust multimetabolomic scores to predict long-term risk of cardiovascular disease and mortality; b) incorporating more diverse populations and a broader range of dietary patterns; and c) conducting more translational research to bridge the gap between precision nutrition studies and clinical applications.
Collapse
Affiliation(s)
- Miguel A Martínez-González
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States.
| | - Francisco J Planes
- Tecnun Escuela de Ingeniería, Departamento de Ingeniería Biomédica y Ciencias, Universidad de Navarra, San Sebastián, Guipúzcoa, Spain
| | - Miguel Ruiz-Canela
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Estefanía Toledo
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Ramón Estruch
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Departamento de Medicina Interna, Instituto de Investigaciones Biomédicas August Pi Sunyer (IDIBAPS), Hospital Clínico, Universidad de Barcelona, Barcelona, Spain
| | - Jordi Salas-Salvadó
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria Pere i Virgili, Departamento de Bioquímica y Biotecnología, Unidad de Nutrición Humana Universidad Rovira i Virgili, Reus, Tarragona, Spain
| | - Rafael Valdés-Más
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Pedro Mena
- Dipartimento di Scienze degli Alimenti e del Farmaco, Universitá di Parma, Parma, Italy
| | - Olga Castañer
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain
| | - Montse Fitó
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Unidad de Riesgo Cardiovascular y Nutrición, Instituto Hospital del Mar de Investigaciones Médicas (IMIM), Barcelona, Spain
| | - Clary Clish
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States
| | - Rikard Landberg
- Division of Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Clemens Wittenbecher
- Department of Life Sciences, SciLifeLab, Chalmers University of Technology, Gothenburg, Sweden
| | - Liming Liang
- Departments of Epidemiology and Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States
| | - Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Department of Public Health and Novo Nordisk Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Rosa M Lamuela-Raventós
- Grup de recerca antioxidants naturals: polifenols, Departament de Nutrició, Ciències de l'Alimentació i Gastronomia, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain; Institut de Nutrició i Seguretat Alimentària (INSA), Universitat de Barcelona (UB), Barcelona, Spain
| | - Dong D Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Nita Forouhi
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Cristina Razquin
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
36
|
Martínez-González MA, Planes FJ, Ruiz-Canela M, Toledo E, Estruch R, Salas-Salvadó J, Valdés-Más R, Mena P, Castañer O, Fitó M, Clish C, Landberg R, Wittenbecher C, Liang L, Guasch-Ferré M, Lamuela-Raventós RM, Wang DD, Forouhi N, Razquin C, Hu FB. Recent advances in precision nutrition and cardiometabolic diseases. Rev Esp Cardiol 2024:S1885-5857(24)00279-2. [PMID: 39357800 DOI: 10.1016/j.recesp.2024.09.005] [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: 08/31/2024] [Accepted: 09/17/2024] [Indexed: 01/11/2025]
Abstract
A growing body of research on nutrition omics has led to recent advances in cardiovascular disease epidemiology and prevention. Within the PREDIMED trial, significant associations between diet-related metabolites and cardiovascular disease were identified, which were subsequently replicated in independent cohorts. Some notable metabolites identified include plasma levels of ceramides, acyl-carnitines, branched-chain amino acids, tryptophan, urea cycle pathways, and the lipidome. These metabolites and their related pathways have been associated with incidence of both cardiovascular disease and type 2 diabetes. Future directions in precision nutrition research include: a) developing more robust multimetabolomic scores to predict long-term risk of cardiovascular disease and mortality; b) incorporating more diverse populations and a broader range of dietary patterns; and c) conducting more translational research to bridge the gap between precision nutrition studies and clinical applications.
Collapse
Affiliation(s)
- Miguel A Martínez-González
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States.
| | - Francisco J Planes
- Tecnun Escuela de Ingeniería, Departamento de Ingeniería Biomédica y Ciencias, Universidad de Navarra, San Sebastián, Guipúzcoa, Spain
| | - Miguel Ruiz-Canela
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Estefanía Toledo
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Ramón Estruch
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Departamento de Medicina Interna, Instituto de Investigaciones Biomédicas August Pi Sunyer (IDIBAPS), Hospital Clínico, Universidad de Barcelona, Barcelona, Spain
| | - Jordi Salas-Salvadó
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria Pere i Virgili, Departamento de Bioquímica y Biotecnología, Unidad de Nutrición Humana Universidad Rovira i Virgili, Reus, Tarragona, Spain
| | - Rafael Valdés-Más
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Pedro Mena
- Dipartimento di Scienze degli Alimenti e del Farmaco, Universitá di Parma, Parma, Italy
| | - Olga Castañer
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain
| | - Montse Fitó
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Unidad de Riesgo Cardiovascular y Nutrición, Instituto Hospital del Mar de Investigaciones Médicas (IMIM), Barcelona, Spain
| | - Clary Clish
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States
| | - Rikard Landberg
- Division of Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Clemens Wittenbecher
- Department of Life Sciences, SciLifeLab, Chalmers University of Technology, Gothenburg, Sweden
| | - Liming Liang
- Departments of Epidemiology and Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States
| | - Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Department of Public Health and Novo Nordisk Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Rosa M Lamuela-Raventós
- Grup de recerca antioxidants naturals: polifenols, Departament de Nutrició, Ciències de l'Alimentació i Gastronomia, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain; Institut de Nutrició i Seguretat Alimentària (INSA), Universitat de Barcelona (UB), Barcelona, Spain
| | - Dong D Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Nita Forouhi
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Cristina Razquin
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
37
|
Xu P, Zeng L, Wang C, Chai J, Yin J, Xu J. Metabolomic characterization of COVID-19 survivors in Jilin province. Respir Res 2024; 25:343. [PMID: 39300427 DOI: 10.1186/s12931-024-02974-0] [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: 03/05/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND The COVID-19 pandemic has escalated into a severe global public health crisis, with persistent sequelae observed in some patients post-discharge. However, metabolomic characterization of the reconvalescent remains unclear. METHODS In this study, serum and urine samples from COVID-19 survivors (n = 16) and healthy subjects (n = 16) underwent testing via the non-targeted metabolomics approach using UPLC-MS/MS. Univariate and multivariate statistical analyses were conducted to delineate the separation between the two sample groups and identify differentially expressed metabolites. By integrating random forest and cluster analysis, potential biomarkers were screened, and the differential metabolites were subsequently subjected to KEGG pathway enrichment analysis. RESULTS Significant differences were observed in the serum and urine metabolic profiles between the two groups. In serum samples, 1187 metabolites were detected, with 874 identified as significant (457 up-regulated, 417 down-regulated); in urine samples, 960 metabolites were detected, with 39 deemed significant (12 up-regulated, 27 down-regulated). Eight potential biomarkers were identified, with KEGG analysis revealing significant enrichment in several metabolic pathways, including arginine biosynthesis. CONCLUSIONS This study offers an overview of the metabolic profiles in serum and urine of COVID-19 survivors, providing a reference for post-discharge monitoring and the prognosis of COVID-19 patients.
Collapse
Affiliation(s)
- Panyang Xu
- Department of Laboratory Medicine, First Hospital of Jilin University, Changchun, China
| | - Lei Zeng
- Bethune Institute of Epigenetic Medicine, First Hospital of Jilin University, Changchun, Jilin, China
- International Center of Future Science, Jilin University, Changchun, China
| | - Chunyu Wang
- State Key Laboratory of Supramolecular Structure and Material Jilin University, Changchun, China
| | - Jiatong Chai
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Junguo Yin
- Department of Clinical Laboratory, Changchun Hospital of Traditional Chinese Medicine, Changchun, China
| | - Jiancheng Xu
- Department of Laboratory Medicine, First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
38
|
Wang X, Shang D, Chen J, Cheng S, Chen D, Zhang Z, Liu C, Yu J, Cao H, Li L, Li L. Serum metabolomics reveals the effectiveness of human placental mesenchymal stem cell therapy for Crohn's disease. Talanta 2024; 277:126442. [PMID: 38897006 DOI: 10.1016/j.talanta.2024.126442] [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: 02/02/2024] [Revised: 06/10/2024] [Accepted: 06/15/2024] [Indexed: 06/21/2024]
Abstract
Mesenchymal stem cell (MSC) therapy offers a promising cure for Crohn's disease (CD), however, its therapeutic effects vary significantly due to individual differences. Therefore, identifying easily detectable biomarkers is essential to assess the efficacy of MSC therapy. In this study, SAMP1/Yit mice were used as a model of CD, which develop spontaneous chronic ileitis, closely resembling the characteristics present in CD patients. Serum metabolic alterations during treatment were analyzed, through the application of differential 12C-/13C-dansylation labeling liquid chromatography-mass spectrometry. Based on the significant differences and time-varying trends of serum amine/phenol-containing metabolites abundance between the control group, the model group, and the treatment group, four serum biomarkers were ultimately screened for evaluating the efficacy of MSC treatment for CD, namely 4-hydroxyphenylpyruvate, 4-hydroxyphenylacetaldehyde, caffeate, and N-acetyltryptamine, whose abundances both increased in the serum of CD model mice and decreased after MSC treatment. These metabolic alterations were associated with tyrosine metabolism, which was validated by the dysregulation of related enzymes. The discovery of biomarkers may help to improve the targeting and effectiveness of treatment and provide innovative prospects for the clinical application of MSC for CD.
Collapse
Affiliation(s)
- Xiao Wang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan City 250117, China
| | - Dandan Shang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan City 250117, China
| | - Junyao Chen
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Sheng Cheng
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Deying Chen
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Zhehua Zhang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Chaoxu Liu
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Jiong Yu
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan City 250117, China; State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China; Zhejiang Key Laboratory for Diagnosis and Treatment of Physic-chemical and Aging-related Injuries, 79 Qingchun Rd, Hangzhou City 310003, China.
| | - Hongcui Cao
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan City 250117, China; State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China; Zhejiang Key Laboratory for Diagnosis and Treatment of Physic-chemical and Aging-related Injuries, 79 Qingchun Rd, Hangzhou City 310003, China.
| | - Liang Li
- Department of Chemistry, University of Alberta, Edmonton, Alberta, T6G 2G2, Canada
| | - Lanjuan Li
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan City 250117, China; State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| |
Collapse
|
39
|
Lokhov PG, Balashova EE, Trifonova OP, Maslov DL, Lokhov AP, Ponomarenko EA, Lisitsa AV, Ugrumov MV, Stilidi IS, Kushlinskii NE, Nikityuk DB, Tutelyan VA, Shestakova MV, Dedov II, Archakov AI. Clinical metabolomics: current state and prospects in Russia. BIOMEDITSINSKAIA KHIMIIA 2024; 70:329-341. [PMID: 39324197 DOI: 10.18097/pbmc20247005329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Using analytical technologies it is possible now to measure the entire diversity of molecules even in a small amount of biological samples. Metabolomic technologies simultaneously analyze thousands of low-molecular substances in a single drop of blood. Such analytical performance opens new possibilities for clinical laboratory diagnostics, still relying on the measurement of only a limited number of clinically significant substances. However, there are objective difficulties hampering introduction of metabolomics into clinical practice. The Institute of Biomedical Chemistry (IBMC), consolidating the efforts of leading scientific and medical organizations, has achieved success in this area by developing a clinical blood metabogram (CBM). CBM opens opportunities to obtain overview on the state of the body with the detailed individual metabolic characteristics of the patient. A number of scientific studies have shown that the CBM is an effective tool for monitoring the state of the body, and based on the CBM patterns (signatures), it is possible to diagnose and monitor the treatment of many diseases. Today, the CBM creation determines the current state and prospects of clinical metabolomics in Russia. This article, dedicated to the 80th anniversary of IBMC, is a review of these achievements focused on a discussion of their implementation in clinical practice.
Collapse
Affiliation(s)
- P G Lokhov
- Institute of Biomedical Chemistry, Moscow, Russia
| | | | | | - D L Maslov
- Institute of Biomedical Chemistry, Moscow, Russia
| | - A P Lokhov
- MIREA - Russian Technological University, Moscow, Russia
| | | | - A V Lisitsa
- Institute of Biomedical Chemistry, Moscow, Russia
| | - M V Ugrumov
- Koltzov Institute of Developmental Biology, Moscow, Russia
| | - I S Stilidi
- Blokhin National Medical Research Center of Oncology, Moscow, Russia
| | - N E Kushlinskii
- Blokhin National Medical Research Center of Oncology, Moscow, Russia
| | - D B Nikityuk
- Federal Research Centre of Nutrition, Biotechnology and Food Safety, Moscow, Russia
| | - V A Tutelyan
- Federal Research Centre of Nutrition, Biotechnology and Food Safety, Moscow, Russia
| | | | - I I Dedov
- Endocrinology Research Centre, Moscow, Russia
| | - A I Archakov
- Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
40
|
Cañadas-Garre M, Maqueda JJ, Baños-Jaime B, Hill C, Skelly R, Cappa R, Brennan E, Doyle R, Godson C, Maxwell AP, McKnight AJ. Mitochondrial related variants associated with cardiovascular traits. Front Physiol 2024; 15:1395371. [PMID: 39258111 PMCID: PMC11385366 DOI: 10.3389/fphys.2024.1395371] [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: 03/03/2024] [Accepted: 08/05/2024] [Indexed: 09/12/2024] Open
Abstract
Introduction Cardiovascular disease (CVD) is responsible for over 30% of mortality worldwide. CVD arises from the complex influence of molecular, clinical, social, and environmental factors. Despite the growing number of autosomal genetic variants contributing to CVD, the cause of most CVDs is still unclear. Mitochondria are crucial in the pathophysiology, development and progression of CVDs; the impact of mitochondrial DNA (mtDNA) variants and mitochondrial haplogroups in the context of CVD has recently been highlighted. Aims We investigated the role of genetic variants in both mtDNA and nuclear-encoded mitochondrial genes (NEMG) in CVD, including coronary artery disease (CAD), hypertension, and serum lipids in the UK Biobank, with sub-group analysis for diabetes. Methods We investigated 371,542 variants in 2,527 NEMG, along with 192 variants in 32 mitochondrial genes in 381,994 participants of the UK Biobank, stratifying by presence of diabetes. Results Mitochondrial variants showed associations with CVD, hypertension, and serum lipids. Mitochondrial haplogroup J was associated with CAD and serum lipids, whereas mitochondrial haplogroups T and U were associated with CVD. Among NEMG, variants within Nitric Oxide Synthase 3 (NOS3) showed associations with CVD, CAD, hypertension, as well as diastolic and systolic blood pressure. We also identified Translocase Of Outer Mitochondrial Membrane 40 (TOMM40) variants associated with CAD; Solute carrier family 22 member 2 (SLC22A2) variants associated with CAD and CVD; and HLA-DQA1 variants associated with hypertension. Variants within these three genes were also associated with serum lipids. Conclusion Our study demonstrates the relevance of mitochondrial related variants in the context of CVD. We have linked mitochondrial haplogroup U to CVD, confirmed association of mitochondrial haplogroups J and T with CVD and proposed new markers of hypertension and serum lipids in the context of diabetes. We have also evidenced connections between the etiological pathways underlying CVDs, blood pressure and serum lipids, placing NOS3, SLC22A2, TOMM40 and HLA-DQA1 genes as common nexuses.
Collapse
Affiliation(s)
- Marisa Cañadas-Garre
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
- MRC Integrative Epidemiology Unit, Bristol Medical School (Population Health Sciences), University of Bristol Oakfield House, Belfast, United Kingdom
| | - Joaquín J Maqueda
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Blanca Baños-Jaime
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
- Instituto de Investigaciones Químicas (IIQ), Centro de Investigaciones Científicas Isla de la Cartuja (cicCartuja), Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Sevilla, Spain
| | - Claire Hill
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
| | - Ryan Skelly
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
| | - Ruaidhri Cappa
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
| | - Eoin Brennan
- UCD Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Ross Doyle
- UCD Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
- Mater Misericordiae University Hospital, Dublin, Ireland
| | - Catherine Godson
- UCD Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Alexander P Maxwell
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
- Regional Nephrology Unit, Belfast City Hospital Belfast, Belfast, United Kingdom
| | - Amy Jayne McKnight
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
| |
Collapse
|
41
|
Wang Z, Gao B, Liu X, Li A. The mediating role of metabolites between gut microbiome and Hirschsprung disease: a bidirectional two-step Mendelian randomization study. Front Pediatr 2024; 12:1371933. [PMID: 39258147 PMCID: PMC11384983 DOI: 10.3389/fped.2024.1371933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024] Open
Abstract
Background Gut microbiome (GM) was observed to be associated with the incidence of Hirschsprung disease (HD). However, the effect and mechanism of GM in HD is still unclear. To investigate the relationship between GM and HD and the effect of metabolites as mediators, a bidirectional two-step Mendelian randomization (MR) study was conducted. Methods The study selected instrument variables (IVs) from summary-level genome-wide association studies (GWAS). The MiBioGen consortium provided the GWAS data for GM, while the GWAS data for metabolites and HD were obtained from the GWAS Catalog consortium. Two-sample MR analyses were performed to estimate bidirectional correlations between IVs associated with GM and HD. Then, genetic variants related to 1,400 metabolite traits were selected for further mediation analyses using the Product method. Results This study found that seven genus bacteria had a significant causal relationship with the incidence of HD but not vice versa. 27 metabolite traits were significantly correlated with HD. After combining the significant results, three significant GM-metabolites-HD lines have been identified. In the Peptococcus-Stearoyl sphingomyelin (d18:1/18:0)-HD line, the Stearoyl sphingomyelin (d18:1/18:0) levels showed a mediation proportion of 14.5%, while in the Peptococcus-lysine-HD line, the lysine levels had a mediation proportion of 12.9%. Additionally, in the Roseburia-X-21733-HD line, the X-21733 levels played a mediation proportion of 23.5%. Conclusion Our MR study indicates a protective effect of Peptococcus on HD risk that is partially mediated through serum levels of stearoyl sphingomyelin (d18:1/18:0) and lysine, and a risk effect of Roseburia on HD that is partially mediated by X-21733 levels. These findings could serve as novel biomarkers and therapeutic targets for HD.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Bingjun Gao
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Xiao Liu
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China
| | - Aiwu Li
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
42
|
Safari-Alighiarloo N, Bostanghadiri N, Sedighi M, Mohebbi A, Vafaei E, Mirshekar M, Razavi S, Alaei-Shahmiri F. Quantification and Correlation Analysis of Bacteroides Species with Diabetes-Related Amino Acids in Individuals with Prediabetes and Type 2 Diabetes Mellitus. Metab Syndr Relat Disord 2024. [PMID: 39133120 DOI: 10.1089/met.2024.0071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024] Open
Abstract
Background: The relationship between gut microbiota and diabetes-related amino acids significantly impacts insulin resistance and obesity. We aimed to quantify two Bacteroidetes species and their correlation with branched-chain amino acids, aromatic amino acids, and glutamate in prediabetes (preDM) and type 2 diabetes mellitus (T2DM). Methods: Fecal samples were collected from 68 participants, including 21 with T2DM, 23 with preDM, and 24 with normal glycemic tolerance (NGT). The abundance of Bacteroides vulgatus and Bacteroides thetaiotaomicron was determined by quantitative real-time polymerase chain reaction. Plasma amino acid measurements were performed using liquid chromatography coupled with tandem mass spectrometry. Results: The quantities of B. vulgatus and B. thetaiotaomicron were reduced in preDM and T2DM than in NGT subjects, but it was not statistically significant. The concentrations of leucine, valine, and tyrosine were significantly higher in preDM and T2DM than in NGT subjects (P < 0.05). A negative correlation was observed between B. thetaiotaomicron abundance and two aromatic amino acids (tyrosine, r = -0.28, P = 0.04; phenylalanine, r = -0.26, P = 0.05). Conclusions: These findings imply that, since gut microbiota varies throughout ethnic groups, further research with many participants will be required to determine the abundance of B. vulgatus and B. thetaiotaomicron in preDM and T2DM and their association with diabetes-related amino acids.
Collapse
Affiliation(s)
- Nahid Safari-Alighiarloo
- Institute of Endocrinology and Metabolism, Endocrine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Narjess Bostanghadiri
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mansour Sedighi
- Department of Microbiology, School of Medicine, Kurdistan University of Medical Sciences, Tehran, Iran
- Research Institute for Health Development, Zoonoses Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Ali Mohebbi
- Stem Cell and Regenerative Medicine Innovation Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Vafaei
- Fetal and Pediatric Cardiovascular Research Center, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Mirshekar
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shabnam Razavi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fariba Alaei-Shahmiri
- Institute of Endocrinology and Metabolism, Endocrine Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
43
|
da Silva Rosa Freire S, Padilha M, Lima Ferreira AL, Machado Schincaglia R, Cunha Figueiredo AC, Freitas-Costa NC, Yin X, Brennan L, Kac G. Association between the third trimester maternal serum metabolome and child growth and development through the first year of life. Sci Rep 2024; 14:18360. [PMID: 39112666 PMCID: PMC11306240 DOI: 10.1038/s41598-024-69247-0] [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: 04/10/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024] Open
Abstract
Evidence suggests that maternal metabolome may be associated with child health outcomes. We analyzed the association between the maternal metabolome between 28-35 gestational weeks and child growth and development during the first year. A prospective cohort of 98 mother-child dyads was followed at birth, 1, 6, and 12 months. Maternal serum samples were collected for targeted LC-MS/MS analysis, which measured 132 metabolites. The child's growth and development were assessed at each time-point. Z-scores were calculated based on WHO growth standards, and the domains of development were assessed using the Ages and Stages Questionnaires (ASQ-3). Multiple linear mixed-effects models were performed and confounders were identified using a Diagram Acyclic Graph. The Benjamini-Hochberg correction was used for multiple comparison adjustments. We found a positive association between lysophosphatidylcholines (14:0; 16:0; 16:1; 17:0; 18:0; 18:1; 18:2; 20:4) with the z-score of weight-for-age, and lysophosphatidylcholines (14:0; 16:0; 16:1; 18:0) and taurine with the z-score of weight-for-length, and lysophosphatidylcholines (14:0; 16:0; 16:1; 17:0; 18:0; 18:1; 18:2; 20:4) and glycine with the z-score of BMI-for-age. The leucine, methionine, tryptophan, and valine were negatively associated with the fine motor skills domain. We observed an association between maternal metabolome and the growth and child's development throughout the first year.
Collapse
Affiliation(s)
- Samary da Silva Rosa Freire
- Department of Social and Applied Nutrition, Josué de Castro Nutrition Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Marina Padilha
- Department of Social and Applied Nutrition, Josué de Castro Nutrition Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ana Lorena Lima Ferreira
- Department of Social and Applied Nutrition, Josué de Castro Nutrition Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Amanda Caroline Cunha Figueiredo
- Department of Social and Applied Nutrition, Josué de Castro Nutrition Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Nathalia Cristina Freitas-Costa
- Department of Social and Applied Nutrition, Josué de Castro Nutrition Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Xiaofei Yin
- UCD School of Agriculture and Food Science, Conway Institute, UCD Institute of Food and Health, University College Dublin, Dublin, Ireland
| | - Lorraine Brennan
- UCD School of Agriculture and Food Science, Conway Institute, UCD Institute of Food and Health, University College Dublin, Dublin, Ireland
| | - Gilberto Kac
- Department of Social and Applied Nutrition, Josué de Castro Nutrition Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
- Nutritional Epidemiology, Josué de Castro Nutrition Institute, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, J2, Room 29, Cidade Universitária, Rio de Janeiro, RJ, 21941902, Brazil.
| |
Collapse
|
44
|
Wang H, Zhan J, Jiang H, Jia H, Pan Y, Zhong X, Huo J, Zhao S. Metagenomics-Metabolomics Exploration of Three-Way-Crossbreeding Effects on Rumen to Provide Basis for Crossbreeding Improvement of Sheep Microbiome and Metabolome of Sheep. Animals (Basel) 2024; 14:2256. [PMID: 39123781 PMCID: PMC11311065 DOI: 10.3390/ani14152256] [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: 06/27/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
The objective of this experiment was to explore the effects of three-way hybridization on rumen microbes and metabolites in sheep using rumen metagenomics and metabolomics. Healthy Hu and CAH (Charolais × Australian White × Hu) male lambs of similar birth weight and age were selected for short-term fattening after intensive weaning to collect rumen fluid for sequencing. Rumen metagenomics diversity showed that Hu and CAH sheep were significantly segregated at the species, KEGG-enzyme, and CAZy-family levels. Moreover, the CAH significantly increased the ACE and Chao1 indices. Further, correlation analysis of the abundance of the top 80 revealed that the microorganisms were interrelated at the species, KEGG-enzyme, and CAZy-family levels. Overall, the microbiome significantly affected metabolites of the top five pathways, with the strongest correlation found with succinic acid. Meanwhile, species-level microbial markers significantly affected rumen differential metabolites. In addition, rumen microbial markers in Hu sheep were overall positively correlated with down-regulated metabolites and negatively correlated with up-regulated metabolites. In contrast, rumen microbial markers in CAH lambs were overall negatively correlated with down-regulated metabolites and positively correlated with up-regulated metabolites. These results suggest that three-way crossbreeding significantly affects rumen microbial community and metabolite composition, and that significant interactions exist between rumen microbes and metabolites.
Collapse
Affiliation(s)
- Haibo Wang
- Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang 330200, China; (H.W.); (J.Z.); (H.J.); (H.J.); (Y.P.); (X.Z.)
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Jinshun Zhan
- Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang 330200, China; (H.W.); (J.Z.); (H.J.); (H.J.); (Y.P.); (X.Z.)
| | - Haoyun Jiang
- Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang 330200, China; (H.W.); (J.Z.); (H.J.); (H.J.); (Y.P.); (X.Z.)
| | - Haobin Jia
- Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang 330200, China; (H.W.); (J.Z.); (H.J.); (H.J.); (Y.P.); (X.Z.)
| | - Yue Pan
- Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang 330200, China; (H.W.); (J.Z.); (H.J.); (H.J.); (Y.P.); (X.Z.)
- College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Xiaojun Zhong
- Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang 330200, China; (H.W.); (J.Z.); (H.J.); (H.J.); (Y.P.); (X.Z.)
| | - Junhong Huo
- Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang 330200, China; (H.W.); (J.Z.); (H.J.); (H.J.); (Y.P.); (X.Z.)
| | - Shengguo Zhao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| |
Collapse
|
45
|
Das SK, Comeau ME, Langefeld CD. Metaboepigenetic regulation of gene expression in obesity and insulin resistance. Metabolomics 2024; 20:91. [PMID: 39096438 DOI: 10.1007/s11306-024-02159-2] [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: 04/01/2024] [Accepted: 07/23/2024] [Indexed: 08/05/2024]
Abstract
INTRODUCTION Variation in DNA methylation (DNAm) in adipose tissue is associated with the pathogenesis of obesity and insulin resistance. The activity of enzymes involved in altering DNAm levels is dependent on several metabolite cofactors. OBJECTIVES To understand the role of metabolites as mechanistic regulators of epigenetic marks, we tested the association between selected plasma metabolites and DNAm levels in the adipose tissue of African Americans. METHODS In the AAGMEx cohort (N = 256), plasma levels of metabolites were measured by untargeted liquid chromatography-mass spectrometry; adipose tissue DNAm and transcript levels were measured by reduced representation bisulfite sequencing, and expression microarray, respectively. RESULTS Among the 21 one-carbon metabolism pathway metabolites evaluated, six were associated with gluco-metabolic traits (PFDR < 0.05, for BMI, SI, or Matsuda index) in AAGMEx. Methylation levels of 196, 116, and 180 CpG-sites were associated (P < 0.0001) with S-adenosylhomocysteine (SAH), cystine, and hypotaurine, respectively. Cis-expression quantitative trait methylation (cis eQTM) analyses suggested the role of metabolite-level-associated CpG sites in regulating the expression of adipose tissue transcripts, including genes in G-protein coupled receptor signaling pathway. Plasma SAH level-associated CpG sites chr19:3403712 and chr19:3403735 were also associated with the expression of G-protein subunit alpha 15 (GNA15) in adipose. The expression of GNA15 was significantly correlated with BMI (β = 1.87, P = 1.9 × 10-16) and SI (β = -1.61, P = 2.49 × 10-5). CONCLUSION Our study suggests that a subset of metabolites modulates the methylation levels of CpG sites in specific loci and, in turn, regulates the expression of transcripts involved in obesity and insulin resistance.
Collapse
Affiliation(s)
- Swapan K Das
- Department of Internal Medicine, Section of Endocrinology and Metabolism, Medical Center Boulevard, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
| | - Mary E Comeau
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Carl D Langefeld
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Center for Precision Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
46
|
Myrmel LS, Fjære E, Han M, Jensen BAH, Rolle-Kampczyk U, Danneskiold-Samsøe NB, Ho QT, Smette A, von Bergen M, Xiao L, Kristiansen K, Madsen L. The Food Sources in Western Diets Modulate Obesity Development, Insulin Sensitivity, and the Plasma and Cecal Metabolome in Mice. Mol Nutr Food Res 2024; 68:e2400246. [PMID: 39107912 DOI: 10.1002/mnfr.202400246] [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: 04/03/2024] [Revised: 07/10/2024] [Indexed: 08/29/2024]
Abstract
SCOPE Dietary constituents modulate development of obesity and type 2 diabetes. The metabolic impact from different food sources in western diets (WD) on obesity development is not fully elucidated. This study aims to identify dietary sources that differentially affect obesity development and the metabolic processes involved. METHODS AND RESULTS Mice were fed isocaloric WDs with protein and fat from different food groups, including egg and dairy, terrestrial meat, game meat, marine, vegetarian, and a mixture of all. This study evaluates development of obesity, glucose tolerance, insulin sensitivity, and plasma and cecal metabolome. WD based on marine or vegetarian food sources protects male mice from obesity development and insulin resistance, whereas meat-based diets promote obesity. The intake of different food sources induces marked differences in the lipid-related plasma metabolome, particularly impacting phosphatidylcholines. Fifty-nine lipid-related plasma metabolites are positively associated with adiposity and a distinct cecal metabolome is found in mice fed a marine diet. CONCLUSION This study demonstrates differences in obesity development between the food groups. Diet specific metabolomic signatures in plasma and cecum associated with adiposity, where a marine based diet modulates the level of plasma and cecal phosphatidylcholines in addition to preventing obesity development.
Collapse
Affiliation(s)
| | - Even Fjære
- Institute of Marine Research, Bergen, 5817, Norway
| | - Mo Han
- BGI Research, Shenzhen, 518083, China
- China National GeneBank, BGI Research, Shenzhen, 518120, China
| | | | - Ulrike Rolle-Kampczyk
- Department of Molecular Toxicology, UFZ-Helmholtz Centre for Environmental Research, 04318, Leipzig, Germany
| | | | - Quang Tri Ho
- Institute of Marine Research, Bergen, 5817, Norway
| | - Anita Smette
- Institute of Marine Research, Bergen, 5817, Norway
| | - Martin von Bergen
- Department of Molecular Toxicology, UFZ-Helmholtz Centre for Environmental Research, 04318, Leipzig, Germany
- Institute of Biochemistry, University of Leipzig, 04103, Leipzig, Germany
| | - Liang Xiao
- BGI Research, Shenzhen, 518083, China
- China National GeneBank, BGI Research, Shenzhen, 518120, China
| | - Karsten Kristiansen
- BGI Research, Shenzhen, 518083, China
- Department of Biology, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Lise Madsen
- Institute of Marine Research, Bergen, 5817, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, 5200, Norway
| |
Collapse
|
47
|
Adolph TE, Tilg H. Western diets and chronic diseases. Nat Med 2024; 30:2133-2147. [PMID: 39085420 DOI: 10.1038/s41591-024-03165-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024]
Abstract
'Westernization', which incorporates industrial, cultural and dietary trends, has paralleled the rise of noncommunicable diseases across the globe. Today, the Western-style diet emerges as a key stimulus for gut microbial vulnerability, chronic inflammation and chronic diseases, affecting mainly the cardiovascular system, systemic metabolism and the gut. Here we review the diet of modern times and evaluate the threat it poses for human health by summarizing recent epidemiological, translational and clinical studies. We discuss the links between diet and disease in the context of obesity and type 2 diabetes, cardiovascular diseases, gut and liver diseases and solid malignancies. We collectively interpret the evidence and its limitations and discuss future challenges and strategies to overcome these. We argue that healthcare professionals and societies must react today to the detrimental effects of the Western diet to bring about sustainable change and improved outcomes in the future.
Collapse
Affiliation(s)
- Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
48
|
Sun J, Zhao J, Zhou S, Li X, Li T, Wang L, Yuan S, Chen D, Law PJ, Larsson SC, Farrington SM, Houlston RS, Dunlop MG, Theodoratou E, Li X. Systematic investigation of genetically determined plasma and urinary metabolites to discover potential interventional targets for colorectal cancer. J Natl Cancer Inst 2024; 116:1303-1312. [PMID: 38648753 PMCID: PMC11308169 DOI: 10.1093/jnci/djae089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/27/2024] [Accepted: 04/13/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND We aimed to identify plasma and urinary metabolites related to colorectal cancer (CRC) risk and elucidate their mediator role in the associations between modifiable risk factors and CRC. METHODS Metabolite quantitative trait loci were derived from 2 published metabolomics genome-wide association studies, and summary-level data were extracted for 651 plasma metabolites and 208 urinary metabolites. Genetic associations with CRC were obtained from a large-scale genome-wide association study meta-analysis (100 204 cases, 154 587 controls) and the FinnGen cohort (4957 cases, 304 197 controls). Mendelian randomization and colocalization analyses were performed to evaluate the causal roles of metabolites in CRC. Druggability evaluation was employed to prioritize potential therapeutic targets. Multivariable Mendelian randomization and mediation estimation were conducted to elucidate the mediating effects of metabolites on the associations between modifiable risk factors and CRC. RESULTS The study identified 30 plasma metabolites and 4 urinary metabolites for CRC. Plasma sphingomyelin and urinary lactose, which were positively associated with CRC risk, could be modulated by drug interventions (ie, olipudase alfa, tilactase). Thirteen modifiable risk factors were associated with 9 metabolites, and 8 of these modifiable risk factors were associated with CRC risk. These 9 metabolites mediated the effect of modifiable risk factors (Actinobacteria, body mass index, waist to hip ratio, fasting insulin, smoking initiation) on CRC. CONCLUSION This study identified key metabolite biomarkers associated with CRC and elucidated their mediator roles in the associations between modifiable risk factors and CRC. These findings provide new insights into the etiology and potential therapeutic targets for CRC and the etiological pathways of modifiable environmental factors with CRC.
Collapse
Affiliation(s)
- Jing Sun
- Department of Big Data in Health Science School of Public Health, and Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jianhui Zhao
- Department of Big Data in Health Science School of Public Health, and Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Siyun Zhou
- Department of Big Data in Health Science School of Public Health, and Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xinxuan Li
- Department of Big Data in Health Science School of Public Health, and Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Tengfei Li
- Department of Big Data in Health Science School of Public Health, and Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lijuan Wang
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Dong Chen
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Philip J Law
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Susanna C Larsson
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Susan M Farrington
- Cancer Research UK Edinburgh Centre, Medical Research Council Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Richard S Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Malcolm G Dunlop
- Cancer Research UK Edinburgh Centre, Medical Research Council Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Colon Cancer Genetics Group, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Evropi Theodoratou
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, UK
- Colon Cancer Genetics Group, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Xue Li
- Department of Big Data in Health Science School of Public Health, and Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
49
|
Lee S, Portlock T, Le Chatelier E, Garcia-Guevara F, Clasen F, Oñate FP, Pons N, Begum N, Harzandi A, Proffitt C, Rosario D, Vaga S, Park J, von Feilitzen K, Johansson F, Zhang C, Edwards LA, Lombard V, Gauthier F, Steves CJ, Gomez-Cabrero D, Henrissat B, Lee D, Engstrand L, Shawcross DL, Proctor G, Almeida M, Nielsen J, Mardinoglu A, Moyes DL, Ehrlich SD, Uhlen M, Shoaie S. Global compositional and functional states of the human gut microbiome in health and disease. Genome Res 2024; 34:967-978. [PMID: 39038849 PMCID: PMC11293553 DOI: 10.1101/gr.278637.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 06/05/2024] [Indexed: 07/24/2024]
Abstract
The human gut microbiota is of increasing interest, with metagenomics a key tool for analyzing bacterial diversity and functionality in health and disease. Despite increasing efforts to expand microbial gene catalogs and an increasing number of metagenome-assembled genomes, there have been few pan-metagenomic association studies and in-depth functional analyses across different geographies and diseases. Here, we explored 6014 human gut metagenome samples across 19 countries and 23 diseases by performing compositional, functional cluster, and integrative analyses. Using interpreted machine learning classification models and statistical methods, we identified Fusobacterium nucleatum and Anaerostipes hadrus with the highest frequencies, enriched and depleted, respectively, across different disease cohorts. Distinct functional distributions were observed in the gut microbiomes of both westernized and nonwesternized populations. These compositional and functional analyses are presented in the open-access Human Gut Microbiome Atlas, allowing for the exploration of the richness, disease, and regional signatures of the gut microbiota across different cohorts.
Collapse
Affiliation(s)
- Sunjae Lee
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), 61005, Gwangju, Republic of Korea
| | - Theo Portlock
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-171 21, Sweden
| | | | - Fernando Garcia-Guevara
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-171 21, Sweden
| | - Frederick Clasen
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
| | | | - Nicolas Pons
- University Paris-Saclay, INRAE, MetaGenoPolis, 78350 Jouy-en-Josas, France
| | - Neelu Begum
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
| | - Azadeh Harzandi
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
| | - Ceri Proffitt
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
| | - Dorines Rosario
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
| | - Stefania Vaga
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
| | - Junseok Park
- Department of Bio and Brain Engineering, KAIST, Yuseong-gu, Daejeon 305-701, Republic of Korea
| | - Kalle von Feilitzen
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-171 21, Sweden
| | - Fredric Johansson
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-171 21, Sweden
| | - Cheng Zhang
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-171 21, Sweden
| | - Lindsey A Edwards
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
- Institute of Liver Studies, Department of Inflammation Biology, School of Immunology and Microbial Sciences, King's College London, London SE5 9NU, United Kingdom
| | - Vincent Lombard
- INRAE, USC1408 Architecture et Fonction des Macromolécules Biologiques (AFMB), Marseille 13288, France
- Architecture et Fonction des Macromolécules Biologiques (AFMB), CNRS, Aix-Marseille University, Marseille 13288, France
| | - Franck Gauthier
- University Paris-Saclay, INRAE, MetaGenoPolis, 78350 Jouy-en-Josas, France
| | - Claire J Steves
- Department of Twin Research & Genetic Epidemiology, King's College London, London WC2R 2LS, United Kingdom
| | - David Gomez-Cabrero
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
- Translational Bioinformatics Unit, Navarrabiomed, Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Bernard Henrissat
- Department of Biological Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Lyngby, Denmark
| | - Doheon Lee
- Department of Bio and Brain Engineering, KAIST, Yuseong-gu, Daejeon 305-701, Republic of Korea
| | - Lars Engstrand
- Centre for Translational Microbiome Research (CTMR), Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Debbie L Shawcross
- Institute of Liver Studies, Department of Inflammation Biology, School of Immunology and Microbial Sciences, King's College London, London SE5 9NU, United Kingdom
| | - Gordon Proctor
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
| | - Mathieu Almeida
- University Paris-Saclay, INRAE, MetaGenoPolis, 78350 Jouy-en-Josas, France
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden
- BioInnovation Institute, DK-2200 Copenhagen N, Denmark
| | - Adil Mardinoglu
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-171 21, Sweden
| | - David L Moyes
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
| | - Stanislav Dusko Ehrlich
- University Paris-Saclay, INRAE, MetaGenoPolis, 78350 Jouy-en-Josas, France
- Department of Clinical and Movement Neurosciences, University College London, London NW3 2PF, United Kingdom
| | - Mathias Uhlen
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-171 21, Sweden;
| | - Saeed Shoaie
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom;
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-171 21, Sweden
| |
Collapse
|
50
|
Leclercq S. Involvement of the gut microbiome-brain axis in alcohol use disorder. Alcohol Alcohol 2024; 59:agae050. [PMID: 39042929 DOI: 10.1093/alcalc/agae050] [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: 04/18/2024] [Revised: 06/06/2024] [Accepted: 07/10/2024] [Indexed: 07/25/2024] Open
Abstract
The human intestine is colonized by a variety of microorganisms that influence the immune system, the metabolic response, and the nervous system, with consequences for brain function and behavior. Unbalance in this microbial ecosystem has been shown to be associated with psychiatric disorders, and altered gut microbiome composition related to bacteria, viruses, and fungi has been well established in patients with alcohol use disorder. This review describes the gut microbiome-brain communication pathways, including the ones related to the vagus nerve, the inflammatory cytokines, and the gut-derived metabolites. Finally, the potential benefits of microbiota-based therapies for the management of alcohol use disorder, such as probiotics, prebiotics, and fecal microbiota transplantation, are also discussed.
Collapse
Affiliation(s)
- Sophie Leclercq
- Laboratory of Nutritional Psychiatry, Institute of Neuroscience, Université Catholique de Louvain, 1200 Brussels, Belgium
| |
Collapse
|