1
|
Yang T, Hu X, Cao F, Yun F, Jia K, Zhang M, Kong G, Nie B, Liu Y, Zhang H, Li X, Gao H, Shi J, Liang G, Hu G, Kasper DL, Song X, Qian Y. Targeting symbionts by apolipoprotein L proteins modulates gut immunity. Nature 2025:10.1038/s41586-025-08990-4. [PMID: 40369072 DOI: 10.1038/s41586-025-08990-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 04/07/2025] [Indexed: 05/16/2025]
Abstract
The mammalian gut harbours trillions of commensal bacteria that interact with their hosts through various bioactive molecules1,2. However, the mutualistic strategies that hosts evolve to benefit from these symbiotic relationships are largely unexplored. Here we report that mouse enterocytes secrete apolipoprotein L9a and b (APOL9a/b) in the presence of microbiota. By integrating flow cytometry sorting of APOL9-binding bacterial taxa with 16S ribosomal RNA gene sequencing (APOL9-seq), we identify that APOL9a/b, as well as their human equivalent APOL2, coat gut bacteria belonging to the order of Bacteroidales with a high degree of specificity through commensal ceramide-1-phosphate (Cer1P) lipids. Genetic abolition of ceramide-1-phosphate synthesis pathways in gut-dominant symbiote Bacteroides thetaiotaomicron significantly decreases the binding of APOL9a/b to the bacterium. Instead of lysing the bacterial cells, coating of APOL9a/b induces the production of outer membrane vesicles (OMVs) from the target bacteria. Subsequently, the Bacteroides-elicited outer membrane vesicles enhance the host's interferon-γ signalling to promote major histocompatibility complex class II expression in the intestinal epithelial cells. In mice, the loss of Apol9a/b compromises the gut major histocompatibility complex class II-instructed immune barrier function, leading to early mortality from infection by intestinal pathogens. Our data show how a host-elicited factor benefits gut immunological homeostasis by selectively targeting commensal ceramide molecules.
Collapse
Affiliation(s)
- Tao Yang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaohu Hu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Fei Cao
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Fenglin Yun
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Kaiwen Jia
- Key Laboratory of RNA Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Mingxiang Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Gaohui Kong
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Biyu Nie
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuexing Liu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Haohao Zhang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xiaoyu Li
- Center for Inflammatory Bowel Disease Research and Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hongyan Gao
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China
| | - Jiantao Shi
- Key Laboratory of RNA Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Guanxiang Liang
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China
| | - Guohong Hu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Dennis L Kasper
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Xinyang Song
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
| | - Youcun Qian
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
2
|
Wu H, Lv B, Zhi L, Shao Y, Liu X, Mitteregger M, Chakaroun R, Tremaroli V, Hazen SL, Wang R, Bergström G, Bäckhed F. Microbiome-metabolome dynamics associated with impaired glucose control and responses to lifestyle changes. Nat Med 2025:10.1038/s41591-025-03642-6. [PMID: 40200054 DOI: 10.1038/s41591-025-03642-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 03/05/2025] [Indexed: 04/10/2025]
Abstract
Type 2 diabetes (T2D) is a complex disease shaped by genetic and environmental factors, including the gut microbiome. Recent research revealed pathophysiological heterogeneity and distinct subgroups in both T2D and prediabetes, prompting exploration of personalized risk factors. Using metabolomics in two Swedish cohorts (n = 1,167), we identified over 500 blood metabolites associated with impaired glucose control, with approximately one-third linked to an altered gut microbiome. Our findings identified metabolic disruptions in microbiome-metabolome dynamics as potential mediators of compromised glucose homeostasis, as illustrated by the potential interactions between Hominifimenecus microfluidus and Blautia wexlerae via hippurate. Short-term lifestyle changes, for example, diet and exercise, modulated microbiome-associated metabolites in a lifestyle-specific manner. This study suggests that the microbiome-metabolome axis is a modifiable target for T2D management, with optimal health benefits achievable through a combination of lifestyle modifications.
Collapse
Affiliation(s)
- Hao Wu
- Center for Obesity and Hernia Surgery, Department of General Surgery, Huashan Hospital, and State Key Laboratory of Genetic Engineering, Fudan Microbiome Center, Human Phenome Institute, Fudan University, Shanghai, China.
| | - Bomin Lv
- Center for Obesity and Hernia Surgery, Department of General Surgery, Huashan Hospital, and State Key Laboratory of Genetic Engineering, Fudan Microbiome Center, Human Phenome Institute, Fudan University, Shanghai, China
| | - Luqian Zhi
- Center for Obesity and Hernia Surgery, Department of General Surgery, Huashan Hospital, and State Key Laboratory of Genetic Engineering, Fudan Microbiome Center, Human Phenome Institute, Fudan University, Shanghai, China
| | - Yikai Shao
- Center for Obesity and Hernia Surgery, Department of General Surgery, Huashan Hospital, and State Key Laboratory of Genetic Engineering, Fudan Microbiome Center, Human Phenome Institute, Fudan University, Shanghai, China
| | - Xinyan Liu
- Center for Obesity and Hernia Surgery, Department of General Surgery, Huashan Hospital, and State Key Laboratory of Genetic Engineering, Fudan Microbiome Center, Human Phenome Institute, Fudan University, Shanghai, China
| | - Matthias Mitteregger
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Rima Chakaroun
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Valentina Tremaroli
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Stanley L Hazen
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland, OH, USA
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH, USA
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ru Wang
- School of Kinesiology, Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Göran Bergström
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Fredrik Bäckhed
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
3
|
Lee J, Son S, Lee M, Park SB. Development of potential immunomodulatory ligands targeting natural killer T cells inspired by gut symbiont-derived glycolipids. Commun Chem 2025; 8:98. [PMID: 40169880 PMCID: PMC11961698 DOI: 10.1038/s42004-025-01497-z] [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: 01/21/2025] [Accepted: 03/20/2025] [Indexed: 04/03/2025] Open
Abstract
α-Galactosylceramide (α-GalCer) is a prototypical antigen recognized by natural killer T (NKT) cells, a subset of T cells crucial for immune regulation. Despite its significance, the complex structure-activity relationship of α-GalCer and its analogs remains poorly understood, particularly in defining the structural determinants of NKT cell responses. In this study, we designed and synthesized potential immunomodulatory ligands targeting NKT cells, inspired by glycolipids derived from the gut symbiont Bacteroides fragilis. A series of α-GalCer analogs with terminal iso-branched sphinganine backbones was developed through rational modification of the acyl chain. Our results identified the C3' hydroxyl group as a structural element that impairs glycolipid presentation by CD1d, as evidenced by reduced IL-2 secretion and weak competition with a potent CD1d ligand. Notably, among C3'-deoxy α-GalCer analogs, those containing an α-chloroacetamide group exhibited robust NKT cell activation with Th2 selectivity. Computational docking and mass spectrometry analyses further confirmed the substantial interaction of α-chloroacetamide analogs to CD1d. These findings underscore the potential of leveraging microbiota-derived glycolipid structures to selectively modulate NKT cell functions for therapeutic purposes.
Collapse
Affiliation(s)
- Jesang Lee
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Sumin Son
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Minha Lee
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Seung Bum Park
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Cao Y, Fan X, Zang T, Li Y, Tu Y, Wei Y, Bai J, Liu Y. Gut microbiota causes depressive phenotype by modulating glycerophospholipid and sphingolipid metabolism via the gut-brain axis. Psychiatry Res 2025; 346:116392. [PMID: 39933221 DOI: 10.1016/j.psychres.2025.116392] [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: 11/12/2024] [Revised: 01/26/2025] [Accepted: 02/06/2025] [Indexed: 02/13/2025]
Abstract
Emerging evidence suggests that changes in the gut microbiota (GM) are related to prenatal depression onset, but the underlying molecular mechanisms remain obscure. This study was conducted to explore how disordered GM is involved in the onset of prenatal depression through the microbiome-gut-brain (MGB) axis. We transplanted fecal microbiota from women with and without prenatal depression into germ-free mice. Fecal metagenomic sequencing and LC-MS untargeted metabolomics analysis were performed to identify the GM composition, function, and metabolites in mice. Lipid metabolomics analysis was then used to characterize the lipid metabolism of brain tissue in mice. We found that mice transplanted with fecal microbiota from women with prenatal depression exhibited depressive-like behaviors as well as characteristic disorders of the phylum Firmicutes. Weighted Gene Correlation Network Analysis identified three microbial and one metabolic module in the gut, alongside two lipid metabolic modules in the brain, as significantly related to all depressive-like behaviors. These modules were enriched for glycerophospholipid and sphingolipid metabolism. In addition, the GM of mice with depressive-like behaviors were enriched and deficient in relevant functions and enzymes in the glycerophospholipid (mainly phosphatidylethanolamine) and sphingolipid (mainly hexosyl-ceramide) metabolic pathways, respectively. Consistently, glycerophospholipid and sphingolipid metabolites in the brains of depressive-like mice were up- and down-regulated. Increased phosphatidylethanolamine and decreased hexosyl-ceramide were significantly related to differential genera in the gut. Collectively, our findings provide a novel microbial and metabolic framework for understanding the role of the MGB axis in prenatal depression, indicating that the GM may be involved in the onset of depressive phenotypes by modulating central glycerophospholipid and sphingolipid metabolic homeostasis.
Collapse
Affiliation(s)
- Yanan Cao
- Renmin Hospital of Wuhan University, Wuhan, 430060, PR China
| | - Xiaoxiao Fan
- Center for Women's and Children's Health, Wuhan University School of Nursing, Wuhan University, Wuhan, 430071, PR China
| | - Tianzi Zang
- Center for Women's and Children's Health, Wuhan University School of Nursing, Wuhan University, Wuhan, 430071, PR China
| | - Yanting Li
- Center for Women's and Children's Health, Wuhan University School of Nursing, Wuhan University, Wuhan, 430071, PR China
| | - Yiming Tu
- Center for Women's and Children's Health, Wuhan University School of Nursing, Wuhan University, Wuhan, 430071, PR China
| | - Yi Wei
- Center for Women's and Children's Health, Wuhan University School of Nursing, Wuhan University, Wuhan, 430071, PR China
| | - Jinbing Bai
- Emory University Nell Hodgson Woodruff School of Nursing, 1520 Clifton Road, Atlanta, GA, 30322, USA
| | - Yanqun Liu
- Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Center for Women's and Children's Health, Wuhan University School of Nursing, Wuhan University, Wuhan, 430071, PR China.
| |
Collapse
|
5
|
Li L, Li T, Liang X, Zhu L, Fang Y, Dong L, Zheng Y, Xu X, Li M, Cai T, Zhao F, Xin M, Shao M, Guan Y, Liu M, Li F, Zhang C, Wang Q, Sun W, Zheng Y. A decrease in Flavonifractor plautii and its product, phytosphingosine, predisposes individuals with phlegm-dampness constitution to metabolic disorders. Cell Discov 2025; 11:25. [PMID: 40097405 PMCID: PMC11914097 DOI: 10.1038/s41421-025-00789-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/28/2025] [Indexed: 03/19/2025] Open
Abstract
According to traditional Chinese medicine (TCM) constitutional theory, individuals with phlegm-dampness constitution (PDC) are at increased risk for metabolic disorders. Previous studies have indicated that PDC individuals exhibit gene expression changes associated with metabolic disorders, even individuals with normal metabolic indices. However, the biological mechanisms underlying these changes remain unclear. The gut microbiota has recently emerged as a promising avenue for elucidating TCM principles. Here, we revealed that individuals with PDC have distinct gut microbiota and serum metabolite profiles. A decrease in phytosphingosine was associated with increased PDC scores and metabolic disorder severity. Subsequent experiments demonstrated that Flavonifractor plautii can biosynthesize phytosphingosine, which was also negatively correlated with the PDC score. Interestingly, both F. plautii and phytosphingosine levels decreased in PDC subjects with normal metabolic indices. Fecal transplantation from these individuals accelerated the development of metabolic disorders in mice. However, supplementation with F. plautii and phytosphingosine ameliorated metabolic disorders by increasing phytosphingosine levels in the gut‒hepatic axis. Mechanistic investigations confirmed that phytosphingosine can directly bind to hepatic peroxisome proliferator-activated receptor α (PPARα) and activate its nuclear transcription activity, thereby regulating downstream gene expression related to glucose‒lipid metabolism. Our research indicates that the decrease in F. plautii and its product, phytosphingosine, contributes to gene expression changes related to metabolic disorders in PDC individuals and increases their susceptibility to metabolic disorders. These findings suggest that diagnosing PDC may be beneficial for identifying at-risk populations among apparently healthy individuals, thereby advancing the broader field of metabolic disorder prevention and TCM integration.
Collapse
Affiliation(s)
- Lingru Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, China
| | - Tianxing Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, China
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Xue Liang
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, China
| | - Linghui Zhu
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, China
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yini Fang
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, China
| | - Ling Dong
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Yi Zheng
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoxue Xu
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Mingrui Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, China
| | - Tianqi Cai
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Fufangyu Zhao
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, China
| | - Meiling Xin
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, China
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Mingyan Shao
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, China
| | - Yuanyuan Guan
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, China
| | - Meiyi Liu
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, China
| | - Fangli Li
- Beijing University of Chinese Medicine Affiliated Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Chenhong Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| | - Qi Wang
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, China.
| | - Wenlong Sun
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China.
| | - Yanfei Zheng
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
6
|
Zhao F, Shao M, Li M, Li T, Zheng Y, Sun W, Ni C, Li L. Sphingolipid metabolites involved in the pathogenesis of atherosclerosis: perspectives on sphingolipids in atherosclerosis. Cell Mol Biol Lett 2025; 30:18. [PMID: 39920588 PMCID: PMC11804087 DOI: 10.1186/s11658-024-00679-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 12/17/2024] [Indexed: 02/09/2025] Open
Abstract
Atherosclerosis, with its complex pathogenesis, is a leading underlying cause of many cardiovascular diseases, which are increasingly prevalent in the population. Sphingolipids play an important role in the development of atherosclerosis. Key metabolites and enzymes in sphingolipid metabolism influence the pathogenesis of atherosclerosis in a variety of ways, including inflammatory responses and oxidative stress. Thus, an investigation of sphingolipid metabolism-related metabolites and key enzymes may provide novel insights and treatment targets for atherosclerosis. This review discusses various mechanisms and research progress on the relationship between various sphingolipid metabolites, related enzymes, and atherosclerosis. Finally, we look into the future research direction of phytosphingolipids.
Collapse
Affiliation(s)
- Fufangyu Zhao
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Mingyan Shao
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Mingrui Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Tianxing Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yanfei Zheng
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Wenlong Sun
- Institute of Biomedical Research, School of Life Sciences, Shandong University of Technology, Zibo, 255000, Shandong, China.
| | - Cheng Ni
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Lingru Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
7
|
Teng Y, Luo C, Qiu X, Mu J, Sriwastva MK, Xu Q, Liu M, Hu X, Xu F, Zhang L, Park JW, Hwang JY, Kong M, Liu Z, Zhang X, Xu R, Yan J, Merchant ML, McClain CJ, Zhang HG. Plant-nanoparticles enhance anti-PD-L1 efficacy by shaping human commensal microbiota metabolites. Nat Commun 2025; 16:1295. [PMID: 39900923 PMCID: PMC11790884 DOI: 10.1038/s41467-025-56498-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
Diet has emerged as a key impact factor for gut microbiota function. However, the complexity of dietary components makes it difficult to predict specific outcomes. Here we investigate the impact of plant-derived nanoparticles (PNP) on gut microbiota and metabolites in context of cancer immunotherapy with the humanized gnotobiotic mouse model. Specifically, we show that ginger-derived exosome-like nanoparticle (GELN) preferentially taken up by Lachnospiraceae and Lactobacillaceae mediated by digalactosyldiacylglycerol (DGDG) and glycine, respectively. We further demonstrate that GELN aly-miR159a-3p enhances anti-PD-L1 therapy in melanoma by inhibiting the expression of recipient bacterial phospholipase C (PLC) and increases the accumulation of docosahexaenoic acid (DHA). An increased level of circulating DHA inhibits PD-L1 expression in tumor cells by binding the PD-L1 promoter and subsequently prevents c-myc-initiated transcription of PD-L1. Colonization of germ-free male mice with gut bacteria from anti-PD-L1 non-responding patients supplemented with DHA enhances the efficacy of anti-PD-L1 therapy compared to controls. Our findings reveal a previously unknown mechanistic impact of PNP on human tumor immunotherapy by modulating gut bacterial metabolic pathways.
Collapse
Affiliation(s)
- Yun Teng
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA.
| | - Chao Luo
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
- Department of Central Laboratory, The affiliated Huai'an First People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Xiaolan Qiu
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
- Department of Breast and Thyroid Surgery, The affiliated Huai'an First People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Jingyao Mu
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Mukesh K Sriwastva
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Qingbo Xu
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA
| | - Minmin Liu
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
- Department of Breast and Thyroid Surgery, The affiliated Huai'an First People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Xin Hu
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fangyi Xu
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Lifeng Zhang
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Juw Won Park
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
- Department of Bioinformatics and Biostatistics, SPHIS, University of Louisville, Louisville, KY, USA
| | - Jae Yeon Hwang
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Maiying Kong
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
- Department of Bioinformatics and Biostatistics, SPHIS, University of Louisville, Louisville, KY, USA
| | - Zhanxu Liu
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Xiang Zhang
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Raobo Xu
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Jun Yan
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Michael L Merchant
- Kidney Disease Program and Clinical Proteomics Center, University of Louisville, Louisville, KY, USA
| | - Craig J McClain
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY, USA
| | - Huang-Ge Zhang
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA.
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA.
- Robley Rex Veterans Affairs Medical Center, Louisville, KY, USA.
| |
Collapse
|
8
|
Costa A, Lucarini E. Treating chronic stress and chronic pain by manipulating gut microbiota with diet: can we kill two birds with one stone? Nutr Neurosci 2025; 28:221-244. [PMID: 38889540 DOI: 10.1080/1028415x.2024.2365021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Background: Chronic stress and chronic pain are closely linked by the capacity to exacerbate each other, sharing common roots in the brain and in the gut. The strict intersection between these two neurological diseases makes important to have a therapeutic strategy aimed at preventing both to maintain mental health in patients. Diet is an modifiable lifestyle factor associated with gut-brain axis diseases and there is growing interest in its use as adjuvant to main therapies. Several evidence attest the impact of specific diets or nutrients on chronic stress-related disorders and pain with a good degree of certainty. A daily adequate intake of foods containing micronutrients such as amino acids, minerals and vitamins, as well as the reduction in the consumption of processed food products can have a positive impact on microbiota and gut health. Many nutrients are endowed of prebiotic, anti-inflammatory, immunomodulatory and neuroprotective potential which make them useful tools helping the management of chronic stress and pain in patients. Dietary regimes, as intermittent fasting or caloric restriction, are promising, although further studies are needed to optimize protocols according to patient's medical history, age and sex. Moreover, by supporting gut microbiota health with diet is possible to attenuate comorbidities such as obesity, gastrointestinal dysfunction and mood disorders, thus reducing healthcare costs related to chronic stress or pain.Objective: This review summarize the most recent evidence on the microbiota-mediated beneficial effects of macro- and micronutrients, dietary-related factors, specific nutritional regimens and dietary intervention on these pathological conditions.
Collapse
Affiliation(s)
- Alessia Costa
- Department of Neuroscience, Psychology, Drug Area and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Elena Lucarini
- Department of Neuroscience, Psychology, Drug Area and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| |
Collapse
|
9
|
Castaño AR, Porcelli SA. Editorial: Cognate recognition, functional properties and immunotherapeutic applications of iNKT cells: leveling the road to the clinic. Front Immunol 2025; 16:1552225. [PMID: 39917293 PMCID: PMC11798922 DOI: 10.3389/fimmu.2025.1552225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 01/09/2025] [Indexed: 02/09/2025] Open
Affiliation(s)
- A. Raul Castaño
- Department of Cellular Biology, Physiology and Immunology, Autonomous University of Barcelona, Barcelona, Spain
| | - Steven Anthony Porcelli
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, New York City, NY, United States
| |
Collapse
|
10
|
Kang J, Hong B, Ma S, Wu J, Yang Z, Fan X, Shao L, Sun K, Zhao J, Fang H, Wu T. Ecological factors affecting toluene biosynthesis from bacterial communities. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 959:178186. [PMID: 39708473 DOI: 10.1016/j.scitotenv.2024.178186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/08/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
As a highly emitted volatile organic compound, toluene significantly contributes to atmospheric pollution and poses high risks to human health. Its anthropogenic source is well understood, while its biosynthesis remains poorly understood, especially by bacterial communities. This research attempted to reveal the temporal changes of bacterial community structure during toluene biosynthesis and identify key bacterial factors using 16S rRNA sequencing gene and machine learning methods. The results showed that toluene biosynthesis by the bacterial consortium nonlinearly increased with phenylacetic acid concentration with the optimal temperature of 25-30 °C and pH of 7-7.5. Diversity and richness of the bacterial communities increased over time, as well as the abundance and composition of phyla (e.g. Bacteroidota and Synergistota), families (e.g. Acidaminococcaceae and Oscillospiraceae), species (e.g. Bacteroides and Parabacteroides), and functional genes (e.g. phenylalanine, tyrosine, and tryptophan biosynthesis and fatty acid metabolism). They were significantly related to toluene biosynthesis, of which the Shannon and Simpson indices and the abundances of Synergistaceae, Bacteroidaceae, and Spirochaetaceae species and functional genes related to metabolic pathways, biosynthesis of secondary metabolites, and alanine aspartate and glutamate metabolism were identified as key factors. Findings of this study contributed to new understandings of the underlying mechanisms of toluene biosynthesis by the bacterial community.
Collapse
Affiliation(s)
- Jian Kang
- School of Ecology and Environment, Anhui Normal University, Wuhu 241000, China
| | - Bing Hong
- School of Ecology and Environment, Anhui Normal University, Wuhu 241000, China; Anhui Provincial Engineering Laboratory of Water and Soil Pollution Control and Remediation, Anhui Normal University, Wuhu 241000, China; Center of Cooperative Innovation for Recovery and Reconstruction of Degraded Ecosystem in Wanjiang City Belt, Wuhu 241000, China.
| | - Shutan Ma
- School of Ecology and Environment, Anhui Normal University, Wuhu 241000, China; Anhui Provincial Engineering Laboratory of Water and Soil Pollution Control and Remediation, Anhui Normal University, Wuhu 241000, China; Center of Cooperative Innovation for Recovery and Reconstruction of Degraded Ecosystem in Wanjiang City Belt, Wuhu 241000, China
| | - Jiangping Wu
- School of Ecology and Environment, Anhui Normal University, Wuhu 241000, China; Anhui Provincial Engineering Laboratory of Water and Soil Pollution Control and Remediation, Anhui Normal University, Wuhu 241000, China; Center of Cooperative Innovation for Recovery and Reconstruction of Degraded Ecosystem in Wanjiang City Belt, Wuhu 241000, China
| | - Zhi Yang
- School of Ecology and Environment, Anhui Normal University, Wuhu 241000, China
| | - Xiaoyu Fan
- School of Ecology and Environment, Anhui Normal University, Wuhu 241000, China
| | - Luyi Shao
- School of Ecology and Environment, Anhui Normal University, Wuhu 241000, China
| | - Kun Sun
- Wuhu Institute of Technology, Wuhu 241006, China
| | - Juan Zhao
- School of Ecology and Environment, Anhui Normal University, Wuhu 241000, China; Anhui Provincial Engineering Laboratory of Water and Soil Pollution Control and Remediation, Anhui Normal University, Wuhu 241000, China; Center of Cooperative Innovation for Recovery and Reconstruction of Degraded Ecosystem in Wanjiang City Belt, Wuhu 241000, China
| | - Hua Fang
- School of Ecology and Environment, Anhui Normal University, Wuhu 241000, China; Anhui Provincial Engineering Laboratory of Water and Soil Pollution Control and Remediation, Anhui Normal University, Wuhu 241000, China; Center of Cooperative Innovation for Recovery and Reconstruction of Degraded Ecosystem in Wanjiang City Belt, Wuhu 241000, China
| | - Ting Wu
- School of Ecology and Environment, Anhui Normal University, Wuhu 241000, China; Anhui Provincial Engineering Laboratory of Water and Soil Pollution Control and Remediation, Anhui Normal University, Wuhu 241000, China; Center of Cooperative Innovation for Recovery and Reconstruction of Degraded Ecosystem in Wanjiang City Belt, Wuhu 241000, China.
| |
Collapse
|
11
|
Cui L, Xie Y, Luo K, Wang M, Liu L, Li C, Tian X. Physiological and intestinal microbiota responses of sea cucumber Apostichopus japonicus to various stress and signatures of intestinal microbiota dysbiosis. Front Microbiol 2024; 15:1528275. [PMID: 39780943 PMCID: PMC11708840 DOI: 10.3389/fmicb.2024.1528275] [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: 11/14/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
Identifying the signatures of intestinal dysbiosis caused by common stresses is fundamental to establishing efficient health monitoring strategies for sea cucumber. This study investigated the impact of six common stress experienced frequently in aquaculture on the growth performance, intestinal homeostasis and microbiota of sea cucumber, including thermal (23°C), hypoosmotic (22‰ salinity), ammonium (0.5 mg/L NH4 +-N), and nitrite (0.25 mg/L NO2 --N) stress exposure for 30 days, as well as starvation and crowding (6 kg/m3 density) stress exposure for 60 days. Results demonstrated that all stress led to reduced growth performance and digestive capacity of sea cucumber, along with varying degrees of oxidative stress and immune responses. Various stresses significantly altered the diversity, community structure (except for crowding stress), and composition of intestinal microbiota. The ratios of Bacteroidota: Proteobacteria (B: P) and Firmicutes: Proteobacteria (F: P) declined markedly compared to the control. Potentially pathogenic bacteria of Shewanellaceae, Vibrionaceae, and Moraxellaceae significantly increased under crowding, ammonium, and nitrite stress, respectively, whereas beneficial microbes of Achromobacter and Rhodobacteraceae were, respectively, enriched under hypoosmotic and starvation stresses. The complexity and stability of microbial ecological networks were further altered by these stresses. KEGG predictions revealed the reduced functional pathways of intestinal microbiota involved in host immunity under different stresses. Correlation analysis further confirmed a strong link between microbiota response and host immunity under different stresses. The increased abundance of Verrucomicrobia species could also be identified as the sensitive indicator for diagnosing whether the host was under stressful pressure by random forest analysis.
Collapse
Affiliation(s)
- Liang Cui
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Yumeng Xie
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Kai Luo
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Mingyang Wang
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Longzhen Liu
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
- The Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| | - Changlin Li
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Xiangli Tian
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| |
Collapse
|
12
|
He Y, Mohapatra G, Asokan S, Nobs SP, Elinav E. Microbiome modulation of antigen presentation in tolerance and inflammation. Curr Opin Immunol 2024; 91:102471. [PMID: 39277909 DOI: 10.1016/j.coi.2024.102471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/17/2024]
Abstract
The microbiome regulates mammalian immune responses from early life to adulthood. Antigen presentation, orchestrating these responses, integrates commensal and pathogenic signals. However, the temporal and spatial specificity of microbiome impacts on antigen presentation and downstream tolerance versus inflammation remain incompletely understood. Herein, we review the influences of antigen presentation of microbiome-related epitopes on immunity; impacts of microbiome-based modulation of antigen presentation on innate and adaptive immune responses; and their ramifications on homeostasis and immune-related disease, ranging from auto-inflammation to tumorigenesis. We highlight mechanisms driving these influences, such as 'molecular mimicry', in which microbiome auto-antigen presentation aberrantly triggers an immune response driving autoimmunity or influences conferred by microbiome-derived metabolites on antigen-presenting cells in inflammatory bowel disease. We discuss unknowns, controversies, and challenges associated with the study of microbiome regulation of antigen presentation while demonstrating how increasing knowledge may contribute to the development of microbiome-based therapeutics modulating immune responses in a variety of clinical contexts.
Collapse
Affiliation(s)
- Yiming He
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Gayatree Mohapatra
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Sahana Asokan
- Microbiome & Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Samuel Philip Nobs
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel.
| | - Eran Elinav
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Microbiome & Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
13
|
Green CR, Kolar MJ, McGregor GH, Nelson AT, Wallace M, Metallo CM. Quantifying acyl-chain diversity in isobaric compound lipids containing monomethyl branched-chain fatty acids. J Lipid Res 2024; 65:100677. [PMID: 39490922 PMCID: PMC11621494 DOI: 10.1016/j.jlr.2024.100677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 11/05/2024] Open
Abstract
Compound lipids comprise a diverse group of metabolites present in living systems, and metabolic- and environmentally-driven structural distinctions across this family are increasingly linked to biological function. However, methods for deconvoluting these often isobaric lipid species are lacking or require specialized instrumentation. Notably, acyl-chain diversity within cells may be influenced by nutritional states, metabolic dysregulation, or genetic alterations. Therefore, a reliable, validated method of quantifying structurally similar even-, odd-, and branched-chain acyl groups within intact compound lipids will be invaluable for gaining molecular insights into their biological functions. Here we demonstrate the chromatographic resolution of isobaric lipids containing distinct combinations of straight-chain and branched-chain acyl groups via ultra-high-pressure liquid chromatography (UHPLC)-mass spectrometry (MS) using a C30 liquid chromatography column. Using metabolically engineered adipocytes lacking branched-keto acid dehydrogenase A (Bckdha), we validate this approach through a combination of fatty acid supplementation and metabolic tracing using monomethyl branched-chain fatty acids and valine. We observe the resolution of numerous isobaric triacylglycerols and other compound lipids, demonstrating the resolving utility of this method. This approach adds to the toolbox for laboratories to quantify and characterize acyl chain diversity across the lipidome.
Collapse
Affiliation(s)
- Courtney R Green
- Molecular and Cellular Biology Laboratory, The Salk Institute for Biological Studies, CA, USA
| | - Matthew J Kolar
- Molecular and Cellular Biology Laboratory, The Salk Institute for Biological Studies, CA, USA; Department of Dermatology, University of California, San Diego, CA, USA
| | - Grace H McGregor
- Molecular and Cellular Biology Laboratory, The Salk Institute for Biological Studies, CA, USA
| | - Andrew T Nelson
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Martina Wallace
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland; Conway Institute of Biomolecular and Biomedical Research, Dublin, Ireland
| | - Christian M Metallo
- Molecular and Cellular Biology Laboratory, The Salk Institute for Biological Studies, CA, USA.
| |
Collapse
|
14
|
Praveena T, Le Nours J. State of play in the molecular presentation and recognition of anti-tumor lipid-based analogues. Front Immunol 2024; 15:1479382. [PMID: 39669569 PMCID: PMC11635198 DOI: 10.3389/fimmu.2024.1479382] [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: 08/12/2024] [Accepted: 11/11/2024] [Indexed: 12/14/2024] Open
Abstract
The Natural Killer T cells (NKT) are a unique subset of T lymphocytes that recognize lipid-based antigens that are presented by the monomorphic MHC-I-like molecule, CD1d. Over 30 years ago, the discovery of the glycolipid α-Galactosylceramide (α-GalCer) from the marine sponge Agelas mauritianus, as a potent activator of the invariant Natural Killer T (iNKT) cells, has attracted great attention for its use in cancer immunotherapy. However, α-GalCer can initiate both pro-inflammatory T helper cell 1 (Th1) and anti-inflammatory Th2 type immune responses that can result in either enhanced or suppressed immunity in a somewhat unpredictable manner. Th1 polarized immune response is often correlated with an optimal anti-tumor immunity, and therefore α-GalCer did not fully offer the desired potential as an anti-tumor therapeutic. Over the past decades, considerable efforts have then been invested into the design and development of novel synthetic α-GalCer analogues that will direct a more efficient immune response towards the production of Th1 biased cytokines. In this minireview, we will discuss how subtle modifications in the chemical nature of a number of α-GalCer derivatives varied immune responses. Whilst some of these analogues showed potential in enhancing stability within CD1d and directing favourable immune responses for tumor immunotherapy, their responses in mice also highlighted the need for further research in humanized models to overcome translational challenges and optimize therapeutic efficacy.
Collapse
Affiliation(s)
| | - Jérôme Le Nours
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
15
|
Yu L, Zhang C, Zhai Q. Gut Microbes Participate in Host Polyamine Metabolism. Proc Natl Acad Sci U S A 2024; 121:e2419368121. [PMID: 39467143 PMCID: PMC11551406 DOI: 10.1073/pnas.2419368121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024] Open
Affiliation(s)
- Leilei Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi214122, China
| | - Chengcheng Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi214122, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi214122, China
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, School of Medicine, Jiangnan University, Wuxi214122, China
| |
Collapse
|
16
|
Li W, Sheng R, Cao M, Rui Y. Exploring the Relationship Between Gut Microbiota and Sarcopenia Based on Gut-Muscle Axis. Food Sci Nutr 2024; 12:8779-8792. [PMID: 39619957 PMCID: PMC11606894 DOI: 10.1002/fsn3.4550] [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: 05/31/2024] [Revised: 09/18/2024] [Accepted: 10/05/2024] [Indexed: 01/04/2025] Open
Abstract
Sarcopenia, as a disease characterized by progressive decline of quality, strength, and function of muscles, has posed an increasingly significant threat to the health of middle-aged and elderly individuals in recent years. With the continuous deepening of studies, the concept of gut-muscle axis has attracted widespread attention worldwide, and the occurrence and development of sarcopenia are believed to be closely related to the composition and functional alterations of gut microbiota. In this review, combined with existing literatures and clinical reports, we have summarized the role and impacts of gut microbiota on the muscle, the relevance between gut microbiota and sarcopenia, potential mechanisms of gut microbiota in the modulation of sarcopenia, potential methods to alleviate sarcopenia by modulating gut microbiota, and relevant advances and perspectives, thus contributing to adding more novel knowledge to this research direction and providing certain reference for future related studies.
Collapse
Affiliation(s)
- Wei Li
- Department of Spinal Surgery Unit 1Hanzhong Central Hospital of Shaanxi ProvinceHanzhongShaanxiChina
- Department of OrthopaedicsTianjin Hospital of NingqiangHanzhongShaanxiChina
| | - Ren‐Wang Sheng
- Department of Orthopaedics, School of Medicine, Zhongda HospitalSoutheast UniversityNanjingJiangsuChina
- School of MedicineSoutheast UniversityNanjingJiangsuChina
- Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, School of Medicine, Zhongda HospitalSoutheast UniversityNanjingJiangsuChina
- Orthopaedic Trauma Institute (OTI)Southeast UniversityNanjingJiangsuChina
| | - Mu‐Min Cao
- Department of Orthopaedics, School of Medicine, Zhongda HospitalSoutheast UniversityNanjingJiangsuChina
- School of MedicineSoutheast UniversityNanjingJiangsuChina
- Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, School of Medicine, Zhongda HospitalSoutheast UniversityNanjingJiangsuChina
- Orthopaedic Trauma Institute (OTI)Southeast UniversityNanjingJiangsuChina
| | - Yun‐Feng Rui
- Department of Orthopaedics, School of Medicine, Zhongda HospitalSoutheast UniversityNanjingJiangsuChina
- School of MedicineSoutheast UniversityNanjingJiangsuChina
- Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, School of Medicine, Zhongda HospitalSoutheast UniversityNanjingJiangsuChina
- Orthopaedic Trauma Institute (OTI)Southeast UniversityNanjingJiangsuChina
| |
Collapse
|
17
|
Xiao X, Singh A, Giometto A, Brito IL. Segatella clades adopt distinct roles within a single individual's gut. NPJ Biofilms Microbiomes 2024; 10:114. [PMID: 39465298 PMCID: PMC11514259 DOI: 10.1038/s41522-024-00590-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024] Open
Abstract
Segatella is a prevalent genus within individuals' gut microbiomes worldwide, especially in non-Western populations. Although metagenomic assembly and genome isolation have shed light on its genetic diversity, the lack of available isolates from this genus has resulted in a limited understanding of how members' genetic diversity translates into phenotypic diversity. Within the confines of a single gut microbiome, we have isolated 63 strains from diverse lineages of Segatella. We performed comparative analyses that exposed differences in cellular morphologies, preferences in polysaccharide utilization, yield of short-chain fatty acids, and antibiotic resistance across isolates. We further show that exposure to Segatella isolates either evokes strong or muted transcriptional responses in human intestinal epithelial cells. Our study exposes large phenotypic differences within related Segatella isolates, extending this to host-microbe interactions.
Collapse
Affiliation(s)
- Xieyue Xiao
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Adarsh Singh
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Andrea Giometto
- School of Civil and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Ilana L Brito
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
18
|
Cheng TY, Praveena T, Govindarajan S, Almeida CF, Pellicci DG, Arkins WC, Van Rhijn I, Venken K, Elewaut D, Godfrey DI, Rossjohn J, Moody DB. Lipidomic scanning of self-lipids identifies headless antigens for natural killer T cells. Proc Natl Acad Sci U S A 2024; 121:e2321686121. [PMID: 39141352 PMCID: PMC11348285 DOI: 10.1073/pnas.2321686121] [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: 01/08/2024] [Accepted: 06/12/2024] [Indexed: 08/15/2024] Open
Abstract
To broadly measure the spectrum of cellular self-antigens for natural killer T cells (NKT), we developed a sensitive lipidomics system to analyze lipids trapped between CD1d and NKT T cell receptors (TCRs). We captured diverse antigen complexes formed in cells from natural endogenous lipids, with or without inducing endoplasmic reticulum (ER) stress. After separating protein complexes with no, low, or high CD1d-TCR interaction, we eluted lipids to establish the spectrum of self-lipids that facilitate this interaction. Although this unbiased approach identified fifteen molecules, they clustered into only two related groups: previously known phospholipid antigens and unexpected neutral lipid antigens. Mass spectrometry studies identified the neutral lipids as ceramides, deoxyceramides, and diacylglycerols, which can be considered headless lipids because they lack polar headgroups that usually form the TCR epitope. The crystal structure of the TCR-ceramide-CD1d complex showed how the missing headgroup allowed the TCR to predominantly contact CD1d, supporting a model of CD1d autoreactivity. Ceramide and related headless antigens mediated physiological TCR binding affinity, weak NKT cell responses, and tetramer binding to polyclonal human and mouse NKT cells. Ceramide and sphingomyelin are oppositely regulated components of the "sphingomyelin cycle" that are altered during apoptosis, transformation, and ER stress. Thus, the unique molecular link of ceramide to NKT cell response, along with the recent identification of sphingomyelin blockers of NKT cell activation, provide two mutually reinforcing links for NKT cell response to sterile cellular stress conditions.
Collapse
Affiliation(s)
- Tan-Yun Cheng
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA02210
| | - T. Praveena
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC3800, Australia
| | - Srinath Govindarajan
- Molecular Immunology and Inflammation Unit, Vlaams Instituut voor Biotechnologie, Center for Inflammation Research, Ghent University, 9052Ghent, Belgium
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics (Rheumatology unit), Ghent University, 9000Ghent, Belgium
| | - Catarina F. Almeida
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC3010, Australia
| | - Daniel G. Pellicci
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC3010, Australia
| | - Wellington C. Arkins
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA02210
| | - Ildiko Van Rhijn
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA02210
| | - Koen Venken
- Molecular Immunology and Inflammation Unit, Vlaams Instituut voor Biotechnologie, Center for Inflammation Research, Ghent University, 9052Ghent, Belgium
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics (Rheumatology unit), Ghent University, 9000Ghent, Belgium
| | - Dirk Elewaut
- Molecular Immunology and Inflammation Unit, Vlaams Instituut voor Biotechnologie, Center for Inflammation Research, Ghent University, 9052Ghent, Belgium
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics (Rheumatology unit), Ghent University, 9000Ghent, Belgium
| | - Dale I. Godfrey
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC3010, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC3800, Australia
- Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, CardiffCF14 4XN, UK
| | - D. Branch Moody
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA02210
| |
Collapse
|
19
|
Pelissier A, Laragione T, Gulko PS, Rodríguez Martínez M. Cell-specific gene networks and drivers in rheumatoid arthritis synovial tissues. Front Immunol 2024; 15:1428773. [PMID: 39161769 PMCID: PMC11330812 DOI: 10.3389/fimmu.2024.1428773] [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: 05/07/2024] [Accepted: 06/24/2024] [Indexed: 08/21/2024] Open
Abstract
Rheumatoid arthritis (RA) is a common autoimmune and inflammatory disease characterized by inflammation and hyperplasia of the synovial tissues. RA pathogenesis involves multiple cell types, genes, transcription factors (TFs) and networks. Yet, little is known about the TFs, and key drivers and networks regulating cell function and disease at the synovial tissue level, which is the site of disease. In the present study, we used available RNA-seq databases generated from synovial tissues and developed a novel approach to elucidate cell type-specific regulatory networks on synovial tissue genes in RA. We leverage established computational methodologies to infer sample-specific gene regulatory networks and applied statistical methods to compare network properties across phenotypic groups (RA versus osteoarthritis). We developed computational approaches to rank TFs based on their contribution to the observed phenotypic differences between RA and controls across different cell types. We identified 18 (fibroblast-like synoviocyte), 16 (T cells), 19 (B cells) and 11 (monocyte) key regulators in RA synovial tissues. Interestingly, fibroblast-like synoviocyte (FLS) and B cells were driven by multiple independent co-regulatory TF clusters that included MITF, HLX, BACH1 (FLS) and KLF13, FOSB, FOSL1 (B cells). However, monocytes were collectively governed by a single cluster of TF drivers, responsible for the main phenotypic differences between RA and controls, which included RFX5, IRF9, CREB5. Among several cell subset and pathway changes, we also detected reduced presence of Natural killer T (NKT) cells and eosinophils in RA synovial tissues. Overall, our novel approach identified new and previously unsuspected Key driver genes (KDG), TF and networks and should help better understanding individual cell regulation and co-regulatory networks in RA pathogenesis, as well as potentially generate new targets for treatment.
Collapse
Affiliation(s)
- Aurelien Pelissier
- Institute of Computational Life Sciences, Zürich University of Applied Sciences (ZHAW), Wädenswil, Switzerland
- AI for Scientific Discovery, IBM Research Europe, Rüschlikon, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Teresina Laragione
- Division of Rheumatology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Percio S. Gulko
- Division of Rheumatology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - María Rodríguez Martínez
- AI for Scientific Discovery, IBM Research Europe, Rüschlikon, Switzerland
- Department of Biomedical Informatics & Data Science, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
20
|
Huang YY, Price MN, Hung A, Gal-Oz O, Tripathi S, Smith CW, Ho D, Carion H, Deutschbauer AM, Arkin AP. Barcoded overexpression screens in gut Bacteroidales identify genes with roles in carbon utilization and stress resistance. Nat Commun 2024; 15:6618. [PMID: 39103350 DOI: 10.1038/s41467-024-50124-3] [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: 05/15/2024] [Accepted: 06/28/2024] [Indexed: 08/07/2024] Open
Abstract
A mechanistic understanding of host-microbe interactions in the gut microbiome is hindered by poorly annotated bacterial genomes. While functional genomics can generate large gene-to-phenotype datasets to accelerate functional discovery, their applications to study gut anaerobes have been limited. For instance, most gain-of-function screens of gut-derived genes have been performed in Escherichia coli and assayed in a small number of conditions. To address these challenges, we develop Barcoded Overexpression BActerial shotgun library sequencing (Boba-seq). We demonstrate the power of this approach by assaying genes from diverse gut Bacteroidales overexpressed in Bacteroides thetaiotaomicron. From hundreds of experiments, we identify new functions and phenotypes for 29 genes important for carbohydrate metabolism or tolerance to antibiotics or bile salts. Highlights include the discovery of a D-glucosamine kinase, a raffinose transporter, and several routes that increase tolerance to ceftriaxone and bile salts through lipid biosynthesis. This approach can be readily applied to develop screens in other strains and additional phenotypic assays.
Collapse
Affiliation(s)
- Yolanda Y Huang
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
- Department of Microbiology and Immunology, University at Buffalo, State University of New York, Buffalo, NY, USA.
| | - Morgan N Price
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Allison Hung
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley, CA, USA
| | - Omree Gal-Oz
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Surya Tripathi
- Department of Plant and Microbial Biology, University of California-Berkeley, Berkeley, CA, USA
| | - Christopher W Smith
- Department of Microbiology and Immunology, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Davian Ho
- Department of Bioengineering, University of California-Berkeley, Berkeley, CA, USA
| | - Héloïse Carion
- Department of Bioengineering, University of California-Berkeley, Berkeley, CA, USA
| | - Adam M Deutschbauer
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Department of Plant and Microbial Biology, University of California-Berkeley, Berkeley, CA, USA
| | - Adam P Arkin
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
- Department of Bioengineering, University of California-Berkeley, Berkeley, CA, USA.
| |
Collapse
|
21
|
Chen C, Han P, Qing Y. Metabolic heterogeneity in tumor microenvironment - A novel landmark for immunotherapy. Autoimmun Rev 2024; 23:103579. [PMID: 39004158 DOI: 10.1016/j.autrev.2024.103579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/10/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024]
Abstract
The surrounding non-cancer cells and tumor cells that make up the tumor microenvironment (TME) have various metabolic rhythms. TME metabolic heterogeneity is influenced by the intricate network of metabolic control within and between cells. DNA, protein, transport, and microbial levels are important regulators of TME metabolic homeostasis. The effectiveness of immunotherapy is also closely correlated with alterations in TME metabolism. The response of a tumor patient to immunotherapy is influenced by a variety of variables, including intracellular metabolic reprogramming, metabolic interaction between cells, ecological changes within and between tumors, and general dietary preferences. Although immunotherapy and targeted therapy have made great strides, their use in the accurate identification and treatment of tumors still has several limitations. The function of TME metabolic heterogeneity in tumor immunotherapy is summarized in this article. It focuses on how metabolic heterogeneity develops and is regulated as a tumor progresses, the precise molecular mechanisms and potential clinical significance of imbalances in intracellular metabolic homeostasis and intercellular metabolic coupling and interaction, as well as the benefits and drawbacks of targeted metabolism used in conjunction with immunotherapy. This offers insightful knowledge and important implications for individualized tumor patient diagnosis and treatment plans in the future.
Collapse
Affiliation(s)
- Chen Chen
- The First Affiliated Hospital of Ningbo University, Ningbo 315211, Zhejiang, China
| | - Peng Han
- Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang, China.
| | - Yanping Qing
- The First Affiliated Hospital of Ningbo University, Ningbo 315211, Zhejiang, China.
| |
Collapse
|
22
|
Hayashizaki K, Kamii Y, Kinjo Y. Glycolipid antigen recognition by invariant natural killer T cells and its role in homeostasis and antimicrobial responses. Front Immunol 2024; 15:1402412. [PMID: 38863694 PMCID: PMC11165115 DOI: 10.3389/fimmu.2024.1402412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 05/14/2024] [Indexed: 06/13/2024] Open
Abstract
Due to the COVID-19 pandemic, the importance of developing effective vaccines has received more attention than ever before. To maximize the effects of vaccines, it is important to select adjuvants that induce strong and rapid innate and acquired immune responses. Invariant natural killer T (iNKT) cells, which constitute a small population among lymphocytes, bypass the innate and acquired immune systems through the rapid production of cytokines after glycolipid recognition; hence, their activation could be used as a vaccine strategy against emerging infectious diseases. Additionally, the diverse functions of iNKT cells, including enhancing antibody production, are becoming more understood in recent years. In this review, we briefly describe the functional subset of iNKT cells and introduce the glycolipid antigens recognized by them. Furthermore, we also introduce novel vaccine development taking advantages of iNKT cell activation against infectious diseases.
Collapse
Affiliation(s)
- Koji Hayashizaki
- Department of Bacteriology, The Jikei University School of Medicine, Tokyo, Japan
- Jikei Center for Biofilm Science and Technology, The Jikei University School of Medicine, Tokyo, Japan
| | - Yasuhiro Kamii
- Department of Bacteriology, The Jikei University School of Medicine, Tokyo, Japan
- Division of Respiratory Diseases, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yuki Kinjo
- Department of Bacteriology, The Jikei University School of Medicine, Tokyo, Japan
- Jikei Center for Biofilm Science and Technology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
23
|
Green CR, Kolar MJ, McGregor GH, Nelson AT, Wallace M, Metallo CM. Quantifying acyl-chain diversity in isobaric compound lipids containing monomethyl branched-chain fatty acids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.28.596332. [PMID: 38853874 PMCID: PMC11160641 DOI: 10.1101/2024.05.28.596332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Compound lipids comprise a diverse group of metabolites present in living systems, and metabolic- and environmentally-driven structural distinctions across this family is increasingly linked to biological function. However, methods for deconvoluting these often isobaric lipid species are lacking or require specialized instrumentation. Notably, acyl-chain diversity within cells may be influenced by nutritional states, metabolic dysregulation, or genetic alterations. Therefore, a reliable, validated method of quantifying structurally similar even-, odd-, and branched-chain acyl groups within intact compound lipids will be invaluable for gaining molecular insights into their biological functions. Here we demonstrate the chromatographic resolution of isobaric lipids containing distinct combinations of straight-chain and branched-chain acyl groups via ultra-high-pressure liquid chromatography (UHPLC)-mass spectrometry (MS) using a C30 liquid chromatography column. Using metabolically-engineered adipocytes lacking branched-keto acid dehydrogenase A (Bckdha), we validate this approach through a combination of fatty acid supplementation and metabolic tracing using monomethyl branched-chain fatty acids and valine. We observe resolution of numerous isobaric triacylglycerols and other compound lipids, demonstrating the resolving utility of this method. This approach strengthens our ability to quantify and characterize the inherent diversity of acyl chains across the lipidome.
Collapse
Affiliation(s)
- CR Green
- Molecular and Cellular Biology Laboratory, The Salk Institute for Biological Studies, 10010N. Torrey Pines Rd., La Jolla, 92037, CA, USA
| | - MJ Kolar
- Molecular and Cellular Biology Laboratory, The Salk Institute for Biological Studies, 10010N. Torrey Pines Rd., La Jolla, 92037, CA, USA
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92037, USA
| | - GH McGregor
- Molecular and Cellular Biology Laboratory, The Salk Institute for Biological Studies, 10010N. Torrey Pines Rd., La Jolla, 92037, CA, USA
| | - AT Nelson
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642
| | - M Wallace
- School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8, Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Research, Dublin, Ireland
| | - CM Metallo
- Molecular and Cellular Biology Laboratory, The Salk Institute for Biological Studies, 10010N. Torrey Pines Rd., La Jolla, 92037, CA, USA
| |
Collapse
|
24
|
Bao B, Wang Y, Boudreau P, Song X, Wu M, Chen X, Patik I, Tang Y, Ouahed J, Ringel A, Barends J, Wu C, Balskus E, Thiagarajah J, Liu J, Wessels MR, Lencer WI, Kasper DL, An D, Horwitz BH, Snapper SB. Bacterial Sphingolipids Exacerbate Colitis by Inhibiting ILC3-derived IL-22 Production. Cell Mol Gastroenterol Hepatol 2024; 18:101350. [PMID: 38704148 PMCID: PMC11222953 DOI: 10.1016/j.jcmgh.2024.04.007] [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: 11/20/2023] [Revised: 04/25/2024] [Accepted: 04/25/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND & AIMS Gut bacterial sphingolipids, primarily produced by Bacteroidetes, have dual roles as bacterial virulence factors and regulators of the host mucosal immune system, including regulatory T cells and invariant natural killer T cells. Patients with inflammatory bowel disease display altered sphingolipids profiles in fecal samples. However, how bacterial sphingolipids modulate mucosal homeostasis and regulate intestinal inflammation remains unclear. METHODS We used dextran sodium sulfate (DSS)-induced colitis in mice monocolonized with Bacteroides fragilis strains expressing or lacking sphingolipids to assess the influence of bacterial sphingolipids on intestinal inflammation using transcriptional, protein, and cellular analyses. Colonic explant and organoid were used to study the function of bacterial sphingolipids. Host mucosal immune cells and cytokines were profiled and characterized using flow cytometry, enzyme-linked immunosorbent assay, and Western blot, and cytokine function in vivo was investigated by monoclonal antibody injection. RESULTS B fragilis sphingolipids exacerbated intestinal inflammation. Mice monocolonized with B fragilis lacking sphingolipids exhibited less severe DSS-induced colitis. This amelioration of colitis was associated with increased production of interleukin (IL)-22 by ILC3. Mice colonized with B fragilis lacking sphingolipids following DSS treatment showed enhanced epithelial STAT3 activity, intestinal cell proliferation, and antimicrobial peptide production. Protection against DSS colitis associated with B fragilis lacking sphingolipids was reversed on IL22 blockade. Furthermore, bacterial sphingolipids restricted epithelial IL18 production following DSS treatment and interfered with IL22 production by a subset of ILC3 cells expressing both IL18R and major histocompatibility complex class II. CONCLUSIONS B fragilis-derived sphingolipids exacerbate mucosal inflammation by impeding epithelial IL18 expression and concomitantly suppressing the production of IL22 by ILC3 cells.
Collapse
Affiliation(s)
- Bin Bao
- Division of Gastroenterology, Hepatology, and Nutrition; Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts; Division of Infectious Diseases, Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts; School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei, Anhui, China.
| | - Youyuan Wang
- Division of Infectious Diseases, Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts; Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Pavl Boudreau
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts
| | - Xinyang Song
- Department of Immunology, Harvard Medical School, Boston, Massachusetts; Shanghai Institute of Biochemistry and Cell Biology, CAS, Shanghai, China
| | - Meng Wu
- Department of Immunology, Harvard Medical School, Boston, Massachusetts
| | - Xi Chen
- Division of Gastroenterology, Hepatology, and Nutrition; Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Izabel Patik
- Division of Gastroenterology, Hepatology, and Nutrition; Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Ying Tang
- Division of Gastroenterology, Hepatology, and Nutrition; Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Jodie Ouahed
- Division of Gastroenterology, Hepatology, and Nutrition; Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Amit Ringel
- Division of Gastroenterology, Hepatology, and Nutrition; Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Jared Barends
- Division of Gastroenterology, Hepatology, and Nutrition; Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Emily Balskus
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts
| | - Jay Thiagarajah
- Division of Gastroenterology, Hepatology, and Nutrition; Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Jian Liu
- School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Michael R Wessels
- Division of Infectious Diseases, Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Wayne Isaac Lencer
- Division of Gastroenterology, Hepatology, and Nutrition; Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Dennis L Kasper
- Department of Immunology, Harvard Medical School, Boston, Massachusetts
| | - Dingding An
- Division of Infectious Diseases, Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Bruce Harold Horwitz
- Division of Gastroenterology, Hepatology, and Nutrition; Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Scott B Snapper
- Division of Gastroenterology, Hepatology, and Nutrition; Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
25
|
Cang W, Li X, Tang J, Wang Y, Mu D, Wu C, Shi H, Shi L, Wu J, Wu R. Therapeutic Potential of Bacteroides fragilis SNBF-1 as a Next-Generation Probiotic: In Vitro Efficacy in Lipid and Carbohydrate Metabolism and Antioxidant Activity. Foods 2024; 13:735. [PMID: 38472847 DOI: 10.3390/foods13050735] [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: 01/20/2024] [Revised: 02/11/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
This study explores the potential of aerotolerant Bacteroides fragilis (B. fragilis) strains as next-generation probiotics (NGPs), focusing on their adaptability in the gastrointestinal environment, safety profile, and probiotic functions. From 23 healthy infant fecal samples, we successfully isolated 56 beneficial B. fragilis strains. Notably, the SNBF-1 strain demonstrated superior cholesterol removal efficiency in HepG2 cells, outshining all other strains by achieving a remarkable reduction in cholesterol by 55.38 ± 2.26%. Comprehensive genotype and phenotype analyses were conducted, including sugar utilization and antibiotic sensitivity tests, leading to the development of an optimized growth medium for SNBF-1. SNBF-1 also demonstrated robust and consistent antioxidant activity, particularly in cell-free extracts, as evidenced by an average oxygen radical absorbance capacity value of 1.061 and a 2,2-diphenyl-1-picrylhydrazyl scavenging ability of 94.53 ± 7.31%. The regulation of carbohydrate metabolism by SNBF-1 was assessed in the insulin-resistant HepG2 cell line. In enzyme inhibition assays, SNBF-1 showed significant α-amylase and α-glucosidase inhibition, with rates of 87.04 ± 2.03% and 37.82 ± 1.36%, respectively. Furthermore, the cell-free supernatant (CFS) of SNBF-1 enhanced glucose consumption and glycogen synthesis in insulin-resistant HepG2 cells, indicating improved cellular energy metabolism. This was consistent with the observation that the CFS of SNBF-1 increased the proliferation of HepG2 cells by 123.77 ± 0.82% compared to that of the control. Overall, this research significantly enhances our understanding of NGPs and their potential therapeutic applications in modulating the gut microbiome.
Collapse
Affiliation(s)
- Weihe Cang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, China
| | - Xuan Li
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Jiayi Tang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, China
| | - Ying Wang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, China
| | - Delun Mu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, China
| | - Chunting Wu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Haisu Shi
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Lin Shi
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Junrui Wu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, China
| | - Rina Wu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, China
| |
Collapse
|
26
|
Xiao X, Le HH, Lee MT, Lamm D, Johnson EL, Brito IL. Prevotella copri variants among a single host diverge in sphingolipid production. mBio 2024; 15:e0240923. [PMID: 38236049 PMCID: PMC10865984 DOI: 10.1128/mbio.02409-23] [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: 09/05/2023] [Accepted: 12/15/2023] [Indexed: 01/19/2024] Open
Abstract
Sphingolipids serve as vital structural and signaling components of the cell membranes in both eukaryotes and prokaryotes. Within the gut microbiome, Bacteroides species have been identified as major producers of sphingolipids, and Bacteroides-produced sphingolipids have been shown to be modulators of host immune and metabolic functions. While Bacteroides species are a prominent feature of the gut microbiomes of populations living in industrialized countries, Prevotella copri, a member of the same phyla, albeit a different family, is the dominant feature across the remainder of the global population, although their sphingolipid-producing capabilities have not been as thoroughly investigated. To fill this gap, we examined the genomes of over 60 diverse isolates of P. copri and identified several key enzymes involved in sphingolipid synthesis in P. copri. Combining bioorthogonal labeling and liquid chromatography-mass spectrometry (LC-MS) based lipidomics, we functionally characterized the first step in P. copri de novo sphingolipid synthesis in addition to profiling the sphingolipidomes of P. copri strains, identifying key enzymes that may play roles in producing a diverse set of P. copri sphingolipids. Given the limited genetic engineering tools amenable for use in P. copri, our approach takes advantage of comparative genomics and phenotypic profiling to explore sphingolipid production in these understudied, yet highly prevalent, organisms.IMPORTANCESphingolipids are important signaling molecules for maintaining metabolic and immune homeostasis in the host. These lipids are also produced by gut commensals, most notably by Bacteroides species. Despite the global prevalence of Prevotella copri in gut microbiomes of individuals, little is known about the types of sphingolipids they produce and whether they are similar in composition and structure to those produced by Bacteroides. Given the varied associations of P. copri with diverse sphingolipid-related health outcomes, such as rheumatoid arthritis and glucose intolerance, it is important to first characterize the specific sphingolipids produced by individual strains of P. copri and to identify the genes involved in their pathways of production. This characterization of P. copri-derived sphingolipids provides further insight into how bacterial sphingolipid production can serve as a mechanism for microbial modulation of host phenotypes.
Collapse
Affiliation(s)
- Xieyue Xiao
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York State, USA
| | - Henry H. Le
- Division of Nutritional Sciences, Cornell University, Ithaca, New York State, USA
| | - Min-Ting Lee
- Division of Nutritional Sciences, Cornell University, Ithaca, New York State, USA
| | - Daniel Lamm
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York State, USA
| | - Elizabeth L. Johnson
- Division of Nutritional Sciences, Cornell University, Ithaca, New York State, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Ilana L. Brito
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York State, USA
| |
Collapse
|
27
|
O'Sullivan A, Brady E, Lafferty L, O'Shea F, O'Regan Z, Meurs N, Baldini M, Gengatharan J, Metallo CM, Wallace M. Long chain monomethyl branched-chain fatty acid levels in human milk vary with gestational weight gain. Prostaglandins Leukot Essent Fatty Acids 2024; 201:102607. [PMID: 38277883 DOI: 10.1016/j.plefa.2024.102607] [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: 10/13/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 01/28/2024]
Abstract
Breastfeeding is an important determinant of infant health and there is immense interest in understanding its metabolite composition so that key beneficial components can be identified. The aim of this research was to measure the fatty acid composition of human milk in an Irish cohort where we examined changes depending on lactation stage and gestational weight gain trajectory. Utilizing a chromatography approach optimal for isomer separation, we identified 44 individual fatty acid species via GCMS and showed that monomethyl branched-chain fatty acids(mmBCFA's), C15:0 and C16:1 are lower in women with excess gestational weight gain versus low gestational weight gain. To further explore the potential contribution of the activity of endogenous metabolic pathways to levels of these fatty acids in milk, we administered D2O to C57BL/6J dams fed a purified lard based high fat diet (HFD) or low-fat diet during gestation and quantified the total and de novo synthesized levels of fatty acids in their milk. We found that de novo synthesis over three days can account for between 10 and 50 % of mmBCFAs in milk from dams on the low-fat diet dependent on the branched-chain fatty acid species. However, HFD fed mice had significantly decreased de novo synthesized fatty acids in milk resulting in lower total mmBCFAs and medium chain fatty acid levels. Overall, our findings highlight the diverse fatty acid composition of human milk and that human milk mmBCFA levels differ between gestational weight gain phenotypes. In addition, our data indicates that de novo synthesis contributes to mmBCFA levels in mice milk and thus may also be a contributory factor to mmBCFA levels in human milk. Given emerging data indicating mmBCFAs may be beneficial components of milk, this study contributes to our knowledge around the phenotypic factors that may impact their levels.
Collapse
Affiliation(s)
- Aifric O'Sullivan
- Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8, Dublin, Ireland
| | - Emer Brady
- Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8, Dublin, Ireland
| | - Lucy Lafferty
- Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8, Dublin, Ireland
| | - Fiona O'Shea
- Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8, Dublin, Ireland
| | - Zoe O'Regan
- Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8, Dublin, Ireland
| | - Noah Meurs
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, 92093, CA, USA
| | - Michelle Baldini
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, 92093, CA, USA
| | - Jivani Gengatharan
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, 92093, CA, USA
| | - Christian M Metallo
- Molecular and Cellular Biology Laboratory, Salk Institute, 10010N. Torrey Pines Rd., La Jolla, 92037, CA, USA
| | - Martina Wallace
- Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8, Dublin, Ireland.
| |
Collapse
|
28
|
Moriyama T, Yoritate M, Kato N, Saika A, Kusuhara W, Ono S, Nagatake T, Koshino H, Kiya N, Moritsuka N, Tanabe R, Hidaka Y, Usui K, Chiba S, Kudo N, Nakahashi R, Igawa K, Matoba H, Tomooka K, Ishikawa E, Takahashi S, Kunisawa J, Yamasaki S, Hirai G. Linkage-Editing Pseudo-Glycans: A Reductive α-Fluorovinyl- C-Glycosylation Strategy to Create Glycan Analogs with Altered Biological Activities. J Am Chem Soc 2024; 146:2237-2247. [PMID: 38196121 DOI: 10.1021/jacs.3c12581] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
The acetal (O-glycoside) bonds of glycans and glycoconjugates are chemically and biologically vulnerable, and therefore C-glycosides are of interest as more stable analogs. We hypothesized that, if the O-glycoside linkage plays a vital role in glycan function, the biological activities of C-glycoside analogs would vary depending on their substituents. Based on this idea, we adopted a "linkage-editing strategy" for the creation of glycan analogs (pseudo-glycans). We designed three types of pseudo-glycans with CH2 and CHF linkages, which resemble the O-glycoside linkage in terms of bond lengths, angles, and bulkiness, and synthesized them efficiently by means of fluorovinyl C-glycosylation and selective hydrogenation reactions. Application of this strategy to isomaltose (IM), an inducer of amylase expression, and α-GalCer, which activates iNKT cells, resulted in the discovery of CH2-IM, which shows increased amylase production ability, and CHF-α-GalCer, which shows activity opposite that of native α-GalCer, serving as an antagonist of iNKT cells.
Collapse
Affiliation(s)
- Takahiro Moriyama
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Makoto Yoritate
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Naoki Kato
- Faculty of Agriculture, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka 573-0101, Japan
- RIKEN Center for Sustainable Resource Science, 2-1, Hirosawa, Wako, Saitama 351-0198, Japan
| | - Azusa Saika
- Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Asagi-Saito, Ibaraki, Osaka 567-0085, Japan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 11 Biopolis Way, Helios, Singapore 138667, Singapore
| | - Wakana Kusuhara
- Research Institute for Microbial Diseases, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan
- Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Shunsuke Ono
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takahiro Nagatake
- Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Asagi-Saito, Ibaraki, Osaka 567-0085, Japan
- Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashi-Mita, Tama, Kawasaki, Kanagawa 214-8571, Japan
| | - Hiroyuki Koshino
- RIKEN Center for Sustainable Resource Science, 2-1, Hirosawa, Wako, Saitama 351-0198, Japan
| | - Noriaki Kiya
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Natsuho Moritsuka
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Riko Tanabe
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yu Hidaka
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kazuteru Usui
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Suzuka Chiba
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Noyuri Kudo
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Rintaro Nakahashi
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kazunobu Igawa
- Institute for Materials Chemistry and Engineering, IRCCS, Kyushu University, Kasuga, Fukuoka 816-8580, Japan
| | - Hiroaki Matoba
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Katsuhiko Tomooka
- Institute for Materials Chemistry and Engineering, IRCCS, Kyushu University, Kasuga, Fukuoka 816-8580, Japan
| | - Eri Ishikawa
- Research Institute for Microbial Diseases, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan
- Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Shunji Takahashi
- RIKEN Center for Sustainable Resource Science, 2-1, Hirosawa, Wako, Saitama 351-0198, Japan
| | - Jun Kunisawa
- Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Asagi-Saito, Ibaraki, Osaka 567-0085, Japan
| | - Sho Yamasaki
- Research Institute for Microbial Diseases, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan
- Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Go Hirai
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
- RIKEN Center for Sustainable Resource Science, 2-1, Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
29
|
Clausen U, Vital ST, Lambertus P, Gehler M, Scheve S, Wöhlbrand L, Rabus R. Catabolic Network of the Fermentative Gut Bacterium Phocaeicola vulgatus (Phylum Bacteroidota) from a Physiologic-Proteomic Perspective. Microb Physiol 2024; 34:88-107. [PMID: 38262373 DOI: 10.1159/000536327] [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: 08/28/2023] [Accepted: 01/10/2024] [Indexed: 01/25/2024]
Abstract
INTRODUCTION Phocaeicola vulgatus (formerly Bacteroides vulgatus) is a prevalent member of human and animal guts, where it influences by its dietary-fiber-fueled, fermentative metabolism the microbial community as well as the host health. Moreover, the fermentative metabolism of P. vulgatus bears potential for a sustainable production of bulk chemicals. The aim of the present study was to refine the current understanding of the P. vulgatus physiology. METHODS P. vulgatus was adapted to anaerobic growth with 14 different carbohydrates, ranging from hexoses, pentoses, hemicellulose, via an uronic acid to deoxy sugars. These substrate-adapted cells formed the basis to define the growth stoichiometries by quantifying growth/fermentation parameters and to reconstruct the catabolic network by applying differential proteomics. RESULTS The determination of growth performance revealed, e.g., doubling times (h) from 1.39 (arabinose) to 14.26 (glucuronate), biomass yields (gCDW/mmolS) from 0.01 (fucose) to 0.27 (α-cyclodextrin), and ATP yields (mMATP/mMC) from 0.21 (rhamnose) to 0.60 (glucuronate/xylan). Furthermore, fermentation product spectra were determined, ranging from broad and balanced (with xylan: acetate, succinate, formate, and propanoate) to rather one sided (with rhamnose or fucose: mainly propane-1,2-diol). The fermentation network serving all tested compounds is composed of 56 proteins (all identified), with several peripheral reaction sequences formed with high substrate specificity (e.g., conversion of arabinose to d-xylulose-3-phosphate) implicating a fine-tuned regulation. By contrast, central modules (e.g., glycolysis or the reaction sequence from PEP to succinate) were constitutively formed. Extensive formation of propane-1,2-diol from rhamnose and fucose involves rhamnulokinase (RhaB), rhamnulose-1-phosphate kinase (RhaD), and lactaldehyde reductase (FucO). Furthermore, Sus-like systems are apparently the most relevant uptake systems and a complex array of transmembrane electron-transfer systems (e.g., Na+-pumping Rnf and Nqr complexes, fumarate reductase) as well as F- and V-type ATP-synthases were detected. CONCLUSIONS The present study provides insights into the potential contribution of P. vulgatus to the gut metabolome and into the strain's biotechnological potential for sustainable production of short-chain fatty acids and alcohols.
Collapse
Affiliation(s)
- Urte Clausen
- General and Molecular Microbiology, Institute for Chemistry and Biology of the Marine Environment (ICBM), Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Sören-Tobias Vital
- General and Molecular Microbiology, Institute for Chemistry and Biology of the Marine Environment (ICBM), Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Pia Lambertus
- General and Molecular Microbiology, Institute for Chemistry and Biology of the Marine Environment (ICBM), Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Martina Gehler
- General and Molecular Microbiology, Institute for Chemistry and Biology of the Marine Environment (ICBM), Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Sabine Scheve
- General and Molecular Microbiology, Institute for Chemistry and Biology of the Marine Environment (ICBM), Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Lars Wöhlbrand
- General and Molecular Microbiology, Institute for Chemistry and Biology of the Marine Environment (ICBM), Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Ralf Rabus
- General and Molecular Microbiology, Institute for Chemistry and Biology of the Marine Environment (ICBM), Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| |
Collapse
|
30
|
Lee SW, Park HJ, Van Kaer L, Hong S. Role of CD1d and iNKT cells in regulating intestinal inflammation. Front Immunol 2024; 14:1343718. [PMID: 38274786 PMCID: PMC10808723 DOI: 10.3389/fimmu.2023.1343718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024] Open
Abstract
Invariant natural killer T (iNKT) cells, a subset of unconventional T cells that recognize glycolipid antigens in a CD1d-dependent manner, are crucial in regulating diverse immune responses such as autoimmunity. By engaging with CD1d-expressing non-immune cells (such as intestinal epithelial cells and enterochromaffin cells) and immune cells (such as type 3 innate lymphoid cells, B cells, monocytes and macrophages), iNKT cells contribute to the maintenance of immune homeostasis in the intestine. In this review, we discuss the impact of iNKT cells and CD1d in the regulation of intestinal inflammation, examining both cellular and molecular factors with the potential to influence the functions of iNKT cells in inflammatory bowel diseases such as Crohn's disease and ulcerative colitis.
Collapse
Affiliation(s)
- Sung Won Lee
- Department of Biomedical Laboratory Science, College of Health and Biomedical Services, Sangji University, Wonju, Republic of Korea
| | - Hyun Jung Park
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, Republic of Korea
| | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Seokmann Hong
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, Republic of Korea
| |
Collapse
|
31
|
Pelissier A, Laragione T, Gulko PS, Rodríguez Martínez M. Cell-Specific Gene Networks and Drivers in Rheumatoid Arthritis Synovial Tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.28.573505. [PMID: 38234732 PMCID: PMC10793435 DOI: 10.1101/2023.12.28.573505] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Rheumatoid arthritis (RA) is a common autoimmune and inflammatory disease characterized by inflammation and hyperplasia of the synovial tissues. RA pathogenesis involves multiple cell types, genes, transcription factors (TFs) and networks. Yet, little is known about the TFs, and key drivers and networks regulating cell function and disease at the synovial tissue level, which is the site of disease. In the present study, we used available RNA-seq databases generated from synovial tissues and developed a novel approach to elucidate cell type-specific regulatory networks on synovial tissue genes in RA. We leverage established computational methodologies to infer sample-specific gene regulatory networks and applied statistical methods to compare network properties across phenotypic groups (RA versus osteoarthritis). We developed computational approaches to rank TFs based on their contribution to the observed phenotypic differences between RA and controls across different cell types. We identified 18,16,19,11 key regulators of fibroblast-like synoviocyte (FLS), T cells, B cells, and monocyte signatures and networks, respectively, in RA synovial tissues. Interestingly, FLS and B cells were driven by multiple independent co-regulatory TF clusters that included MITF, HLX, BACH1 (FLS) and KLF13, FOSB, FOSL1 (synovial B cells). However, monocytes were collectively governed by a single cluster of TF drivers, responsible for the main phenotypic differences between RA and controls, which included RFX5, IRF9, CREB5. Among several cell subset and pathway changes, we also detected reduced presence of NKT cell and eosinophils in RA synovial tissues. Overall, our novel approach identified new and previously unsuspected KDG, TF and networks and should help better understanding individual cell regulation and co-regulatory networks in RA pathogenesis, as well as potentially generate new targets for treatment.
Collapse
Affiliation(s)
- Aurelien Pelissier
- IBM Research Europe, 8803 Rüschlikon, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
- Currently at Institute of Computational Life Sciences, ZHAW, 8400 Winterthur, Switzerland
| | - Teresina Laragione
- Division of Rheumatology, Icahn School of Medicine at Mount Sinai, 10029 New York, United States
| | - Percio S. Gulko
- Division of Rheumatology, Icahn School of Medicine at Mount Sinai, 10029 New York, United States
| | - María Rodríguez Martínez
- IBM Research Europe, 8803 Rüschlikon, Switzerland
- Currently at Yale School of Medicine, 06510 New Haven, United States
| |
Collapse
|
32
|
Zhang H, Xie Y, Cao F, Song X. Gut microbiota-derived fatty acid and sterol metabolites: biotransformation and immunomodulatory functions. Gut Microbes 2024; 16:2382336. [PMID: 39046079 PMCID: PMC11271093 DOI: 10.1080/19490976.2024.2382336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 05/26/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024] Open
Abstract
Commensal microorganisms in the human gut produce numerous metabolites by using small molecules derived from the host or diet as precursors. Host or dietary lipid molecules are involved in energy metabolism and maintaining the structural integrity of cell membranes. Notably, gut microbes can convert these lipids into bioactive signaling molecules through their biotransformation and synthesis pathways. These microbiota-derived lipid metabolites can affect host physiology by influencing the body's immune and metabolic processes. This review aims to summarize recent advances in the microbial transformation and host immunomodulatory functions of these lipid metabolites, with a special focus on fatty acids and steroids produced by our gut microbiota.
Collapse
Affiliation(s)
- Haohao Zhang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yadong Xie
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Fei Cao
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xinyang Song
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
33
|
Castelo J, Araujo-Aris S, Barriales D, Tanner Pasco S, Seoane I, Peña-Cearra A, Palacios A, Simó C, Garcia-Cañas V, Khamwong M, Martín-Ruiz I, Gonzalez-Lopez M, Barcena L, Martín Rodríguez JE, Lavín JL, Gutiez N, Marcos R, Atondo E, Cobela A, Plaza-Vinuesa L, Plata A, Santos-Fernandez E, Fernandez-Tejada A, Villarán MC, Mancheño JM, Maria Flores J, María Aransay A, Pellón A, de Las Rivas B, Muñoz R, Margolles A, Ruas-Madiedo P, Victoria Selma M, Gomez de Agüero M, Abecia L, Anguita J, Rodríguez H. The microbiota metabolite, phloroglucinol, confers long-term protection against inflammation. Gut Microbes 2024; 16:2438829. [PMID: 39676480 DOI: 10.1080/19490976.2024.2438829] [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: 07/17/2024] [Revised: 10/16/2024] [Accepted: 11/26/2024] [Indexed: 12/17/2024] Open
Abstract
Phloroglucinol is a key byproduct of gut microbial metabolism that has been widely used as a treatment for irritable bowel syndrome. Here, we demonstrate that phloroglucinol tempers macrophage responses to pro-inflammatory pathogens and stimuli. In vivo, phloroglucinol administration decreases gut and extraintestinal inflammation in murine models of inflammatory bowel disease and systemic infection. The metabolite induces modest modifications in the microbiota. However, the presence of an active microbiota is required to preserve its anti-inflammatory activity. Remarkably, the protective effect of phloroglucinol lasts partially at least 6 months. Single-cell transcriptomic analysis of bone marrow progenitors demonstrates the capacity of the metabolite to induce long-lasting innate immune training in hematopoietic lineages, at least partially through the participation of the receptor and transcription factor, aryl hydrocarbon receptor (AhR). Phloroglucinol induces alterations in metabolic and epigenetic pathways that are most prevalent in upstream progenitors as hallmarks of central trained immunity. These data identify phloroglucinol as a dietary-derived compound capable of inducing central trained immunity and modulating the response of the host to inflammatory insults.
Collapse
Affiliation(s)
- Janire Castelo
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Sarai Araujo-Aris
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Diego Barriales
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | | | - Iratxe Seoane
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
- Department of Immunology, Microbiology and Parasitology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Ainize Peña-Cearra
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
- Department of Immunology, Microbiology and Parasitology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Ainhoa Palacios
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Carolina Simó
- Molecular Nutrition and Metabolism, Institute of Food Science Research (CIAL), Spanish National Research Council (CSIC), Madrid, Spain
| | - Virginia Garcia-Cañas
- Molecular Nutrition and Metabolism, Institute of Food Science Research (CIAL), Spanish National Research Council (CSIC), Madrid, Spain
| | - Muthita Khamwong
- Würzburg Institute of Systems Immunology, Max-Planck Research Group at the Julius-Maximilians Universität, Würzburg, Germany
| | - Itziar Martín-Ruiz
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | | | - Laura Barcena
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | | | - José Luís Lavín
- Applied Mathematics Department - Bioinformatics Unit, NEIKER-BRTA, Derio, Spain
| | - Naiara Gutiez
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Raquel Marcos
- Departamento de Microbiología y Bioquímica, Instituto de Productos Lácteos de Asturias, CSIC, Villaviciosa, Spain
- Functionality and Ecology of Beneficial Microbes (MicroHealth) Group, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Estibaliz Atondo
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Arantza Cobela
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Laura Plaza-Vinuesa
- Departamento de PRocesos Tecnológicos y Biotecnología, Instituto de Ciencia y Tecnología de Alimentos y Nutrición (ICTAN), CSIC, Madrid, Spain
| | - Adrián Plata
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | | | - Alberto Fernandez-Tejada
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | | | - José Miguel Mancheño
- Department of Crystallography and Structural Biology, Institute of Physical Chemistry Blas Cabrera (IQF), CSIC, Madrid, Spain
| | - Juana Maria Flores
- Department of Animal Medicine and Surgery, Veterinary Faculty, Universidad Complutense de Madrid, Madrid, Spain
| | - Ana María Aransay
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
- CIBERehd, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Aize Pellón
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Blanca de Las Rivas
- Departamento de PRocesos Tecnológicos y Biotecnología, Instituto de Ciencia y Tecnología de Alimentos y Nutrición (ICTAN), CSIC, Madrid, Spain
| | - Rosario Muñoz
- Departamento de PRocesos Tecnológicos y Biotecnología, Instituto de Ciencia y Tecnología de Alimentos y Nutrición (ICTAN), CSIC, Madrid, Spain
| | - Abelardo Margolles
- Departamento de Microbiología y Bioquímica, Instituto de Productos Lácteos de Asturias, CSIC, Villaviciosa, Spain
- Functionality and Ecology of Beneficial Microbes (MicroHealth) Group, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Patricia Ruas-Madiedo
- Departamento de Microbiología y Bioquímica, Instituto de Productos Lácteos de Asturias, CSIC, Villaviciosa, Spain
- Functionality and Ecology of Beneficial Microbes (MicroHealth) Group, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Maria Victoria Selma
- Laboratory of Food & Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, CEBAS-CSIC, Murcia, Spain
| | - Mercedes Gomez de Agüero
- Würzburg Institute of Systems Immunology, Max-Planck Research Group at the Julius-Maximilians Universität, Würzburg, Germany
| | - Leticia Abecia
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
- Department of Immunology, Microbiology and Parasitology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Juan Anguita
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Héctor Rodríguez
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| |
Collapse
|
34
|
Cuaycal AE, Teixeira LD, Lorca GL, Gonzalez CF. Lactobacillus johnsonii N6.2 phospholipids induce immature-like dendritic cells with a migratory-regulatory-like transcriptional signature. Gut Microbes 2023; 15:2252447. [PMID: 37675983 PMCID: PMC10486300 DOI: 10.1080/19490976.2023.2252447] [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: 02/23/2023] [Revised: 07/12/2023] [Accepted: 08/23/2023] [Indexed: 09/08/2023] Open
Abstract
Shifts in the gut microbiota composition, called dysbiosis, have been directly associated with acute and chronic diseases. However, the underlying biological systems connecting gut dysbiosis to systemic inflammatory pathologies are not well understood. Phospholipids (PLs) act as precursors of both, bioactive inflammatory and resolving mediators. Their dysregulation is associated with chronic diseases including cancer. Gut microbial-derived lipids are structurally unique and capable of modulating host's immunity. Lactobacillus johnsonii N6.2 is a Gram-positive gut symbiont with probiotic characteristics. L. johnsonii N6.2 reduces the incidence of autoimmunity in animal models of Type 1 Diabetes and improves general wellness in healthy volunteers by promoting, in part, local and systemic anti-inflammatory responses. By utilizing bioassay-guided fractionation methods with bone marrow-derived dendritic cells (BMDCs), we report here that L. johnsonii N6.2 purified lipids induce a transcriptional signature that resembles that of migratory (mig) DCs. RNAseq-based analysis showed that BMDCs stimulated with L. johnsonii N6.2 total lipids upregulate maturation-mig related genes Cd86, Cd40, Ccr7, Icam1 along with immunoregulatory genes including Itgb8, Nfkbiz, Jag1, Adora2a, IL2ra, Arg1, and Cd274. Quantitative reverse transcription (qRT)-PCR analysis indicated that PLs are the bioactive lipids triggering the BMDCs response. Antibody-blocking of surface Toll-like receptor (TLR)2 resulted in boosted PL-mediated upregulation of pro-inflammatory Il6. Chemical inhibition of the IKKα kinase from the non-canonical NF-κB pathway specifically restricted upregulation of Il6 and Tnf. Phenotypically, PL-stimulated BMDCs displayed an immature like-phenotype with significantly increased surface ICAM-1. This study provides insight into the immunoregulatory capacity of Gram-positive, gut microbial-derived phospholipids on innate immune responses.
Collapse
Affiliation(s)
- Alexandra E. Cuaycal
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, USA
| | - Leandro Dias Teixeira
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, USA
| | - Graciela L. Lorca
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, USA
| | - Claudio F. Gonzalez
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, USA
| |
Collapse
|
35
|
Tan Y, Liang J, Lai M, Wan S, Luo X, Li F. Advances in synthetic biology toolboxes paving the way for mechanistic understanding and strain engineering of gut commensal Bacteroides spp. and Clostridium spp. Biotechnol Adv 2023; 69:108272. [PMID: 37844770 DOI: 10.1016/j.biotechadv.2023.108272] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
The gut microbiota plays a significant role in influencing human immunity, metabolism, development, and behavior by producing a wide range of metabolites. While there is accumulating data on several microbiota-derived small molecules that contribute to host health and disease, our knowledge regarding the molecular mechanisms underlying metabolite-mediated microbe-host interactions remains limited. This is primarily due to the lack of efficient genetic tools for most commensal bacteria, especially those belonging to the dominant phyla Bacteroides spp. and Clostridium spp., which hinders the application of synthetic biology to these gut commensal bacteria. In this review, we provide an overview of recent advances in synthetic biology tools developed for the two dominant genera, as well as their applications in deciphering the mechanisms of microbe-host interactions mediated by microbiota-derived small molecules. We also discuss the potential biomedical applications of engineering commensal bacteria using these toolboxes. Finally, we share our perspective on the future development of synthetic biology tools for a better understanding of small molecule-mediated microbe-host interactions and their engineering for biomedical purposes.
Collapse
Affiliation(s)
- Yang Tan
- Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China; Shandong Energy Institute, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, Qingdao 266101, China.
| | - Jing Liang
- Center for Synthetic Biochemistry, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Mingchi Lai
- College of Life Sciences, Qingdao Agricultural University, Qingdao 266109, China
| | - Sai Wan
- Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China; Shandong Energy Institute, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, Qingdao 266101, China
| | - Xiaozhou Luo
- Center for Synthetic Biochemistry, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Fuli Li
- Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China; Shandong Energy Institute, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, Qingdao 266101, China.
| |
Collapse
|
36
|
Van Dingenen L, Segers C, Wouters S, Mysara M, Leys N, Kumar-Singh S, Malhotra-Kumar S, Van Houdt R. Dissecting the role of the gut microbiome and fecal microbiota transplantation in radio- and immunotherapy treatment of colorectal cancer. Front Cell Infect Microbiol 2023; 13:1298264. [PMID: 38035338 PMCID: PMC10687483 DOI: 10.3389/fcimb.2023.1298264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most commonly diagnosed cancers and poses a major burden on the human health worldwide. At the moment, treatment of CRC consists of surgery in combination with (neo)adjuvant chemotherapy and/or radiotherapy. More recently, immune checkpoint blockers (ICBs) have also been approved for CRC treatment. In addition, recent studies have shown that radiotherapy and ICBs act synergistically, with radiotherapy stimulating the immune system that is activated by ICBs. However, both treatments are also associated with severe toxicity and efficacy issues, which can lead to temporary or permanent discontinuation of these treatment programs. There's growing evidence pointing to the gut microbiome playing a role in these issues. Some microorganisms seem to contribute to radiotherapy-associated toxicity and hinder ICB efficacy, while others seem to reduce radiotherapy-associated toxicity or enhance ICB efficacy. Consequently, fecal microbiota transplantation (FMT) has been applied to reduce radio- and immunotherapy-related toxicity and enhance their efficacies. Here, we have reviewed the currently available preclinical and clinical data in CRC treatment, with a focus on how the gut microbiome influences radio- and immunotherapy toxicity and efficacy and if these treatments could benefit from FMT.
Collapse
Affiliation(s)
- Lena Van Dingenen
- Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | - Charlotte Segers
- Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
| | - Shari Wouters
- Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
- Molecular Pathology Group, Laboratory of Cell Biology and Histology, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | - Mohamed Mysara
- Bioinformatics Group, Center for Informatics Science, School of Information Technology and Computer Science, Nile University, Giza, Egypt
| | - Natalie Leys
- Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
| | - Samir Kumar-Singh
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
- Molecular Pathology Group, Laboratory of Cell Biology and Histology, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | - Surbhi Malhotra-Kumar
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | - Rob Van Houdt
- Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
| |
Collapse
|
37
|
Fan L, Xia Y, Wang Y, Han D, Liu Y, Li J, Fu J, Wang L, Gan Z, Liu B, Fu J, Zhu C, Wu Z, Zhao J, Han H, Wu H, He Y, Tang Y, Zhang Q, Wang Y, Zhang F, Zong X, Yin J, Zhou X, Yang X, Wang J, Yin Y, Ren W. Gut microbiota bridges dietary nutrients and host immunity. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2466-2514. [PMID: 37286860 PMCID: PMC10247344 DOI: 10.1007/s11427-023-2346-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 04/05/2023] [Indexed: 06/09/2023]
Abstract
Dietary nutrients and the gut microbiota are increasingly recognized to cross-regulate and entrain each other, and thus affect host health and immune-mediated diseases. Here, we systematically review the current understanding linking dietary nutrients to gut microbiota-host immune interactions, emphasizing how this axis might influence host immunity in health and diseases. Of relevance, we highlight that the implications of gut microbiota-targeted dietary intervention could be harnessed in orchestrating a spectrum of immune-associated diseases.
Collapse
Affiliation(s)
- Lijuan Fan
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yaoyao Xia
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Youxia Wang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yanli Liu
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China
| | - Jiahuan Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jie Fu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Leli Wang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Zhending Gan
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Bingnan Liu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jian Fu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Congrui Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Zhenhua Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jinbiao Zhao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Hui Han
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Hao Wu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yiwen He
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Yulong Tang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Qingzhuo Zhang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yibin Wang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China
| | - Fan Zhang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China
| | - Xin Zong
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China.
| | - Xihong Zhou
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China.
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China.
| | - Wenkai Ren
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
38
|
Bai X, Ya R, Tang X, Cai M. Role and interaction of bacterial sphingolipids in human health. Front Microbiol 2023; 14:1289819. [PMID: 37937219 PMCID: PMC10626005 DOI: 10.3389/fmicb.2023.1289819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/09/2023] [Indexed: 11/09/2023] Open
Abstract
Sphingolipids, present in both higher animals and prokaryotes, involving in cell differentiation, pathogenesis and apoptosis in human physiological health. With increasing attention on the gut microbiome and its impact on wellbeing, there is a renewed focus on exploring bacterial sphingolipids. This review aims to consolidate the current understanding of bacterial sphingolipids and their impact on host health. Compared to mammalian sphingolipids, bacterial sphingolipids are characterized by odd chain lengths due to the presence of branched alkyl chains. Additionally, intestinal microbial sphingolipids can migrate from the gut to various host organs, affecting the immune system and metabolism. Furthermore, the intricate interplay between dietary sphingolipids and the gut microbiota is explored, shedding light on their complex relationship. Despite limited knowledge in this area, this review aims to raise awareness about the importance of bacterial sphingolipids and further our understanding of more uncharacterized bacterial sphingolipids and their significant role in maintaining host health.
Collapse
Affiliation(s)
- Xiaoye Bai
- School of Medicine, Sun Yat-sen University, Shenzhen, China
- Shenzhen Bay Laboratory, Institute of Chemical Biology, Shenzhen, China
| | - Ru Ya
- Inner Mongolia Academy of Science and Technology, Hohhot, China
| | - Xiaoyu Tang
- Shenzhen Bay Laboratory, Institute of Chemical Biology, Shenzhen, China
| | - Mingwei Cai
- Shenzhen Bay Laboratory, Institute of Chemical Biology, Shenzhen, China
| |
Collapse
|
39
|
Di Ciaula A, Bonfrate L, Khalil M, Garruti G, Portincasa P. Contribution of the microbiome for better phenotyping of people living with obesity. Rev Endocr Metab Disord 2023; 24:839-870. [PMID: 37119391 PMCID: PMC10148591 DOI: 10.1007/s11154-023-09798-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2023] [Indexed: 05/01/2023]
Abstract
Obesity has reached epidemic proportion worldwide and in all ages. Available evidence points to a multifactorial pathogenesis involving gene predisposition and environmental factors. Gut microbiota plays a critical role as a major interface between external factors, i.e., diet, lifestyle, toxic chemicals, and internal mechanisms regulating energy and metabolic homeostasis, fat production and storage. A shift in microbiota composition is linked with overweight and obesity, with pathogenic mechanisms involving bacterial products and metabolites (mainly endocannabinoid-related mediators, short-chain fatty acids, bile acids, catabolites of tryptophan, lipopolysaccharides) and subsequent alterations in gut barrier, altered metabolic homeostasis, insulin resistance and chronic, low-grade inflammation. Although animal studies point to the links between an "obesogenic" microbiota and the development of different obesity phenotypes, the translational value of these results in humans is still limited by the heterogeneity among studies, the high variation of gut microbiota over time and the lack of robust longitudinal studies adequately considering inter-individual confounders. Nevertheless, available evidence underscores the existence of several genera predisposing to obesity or, conversely, to lean and metabolically health phenotype (e.g., Akkermansia muciniphila, species from genera Faecalibacterium, Alistipes, Roseburia). Further longitudinal studies using metagenomics, transcriptomics, proteomics, and metabolomics with exact characterization of confounders are needed in this field. Results must confirm that distinct genera and specific microbial-derived metabolites represent effective and precision interventions against overweight and obesity in the long-term.
Collapse
Affiliation(s)
- Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70124 Bari, Italy
| | - Leonilde Bonfrate
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70124 Bari, Italy
| | - Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70124 Bari, Italy
| | - Gabriella Garruti
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70124 Bari, Italy
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70124 Bari, Italy
| |
Collapse
|
40
|
Kim S, Cho S, Kim JH. CD1-mediated immune responses in mucosal tissues: molecular mechanisms underlying lipid antigen presentation system. Exp Mol Med 2023; 55:1858-1871. [PMID: 37696897 PMCID: PMC10545705 DOI: 10.1038/s12276-023-01053-6] [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: 02/28/2023] [Revised: 05/02/2023] [Accepted: 05/07/2023] [Indexed: 09/13/2023] Open
Abstract
The cluster of differentiation 1 (CD1) molecule differs from major histocompatibility complex class I and II because it presents glycolipid/lipid antigens. Moreover, the CD1-restricted T cells that recognize these self and foreign antigens participate in both innate and adaptive immune responses. CD1s are constitutively expressed by professional and nonprofessional antigen-presenting cells in mucosal tissues, namely, the skin, lung, and intestine. This suggests that CD1-reactive T cells are involved in the immune responses of these tissues. Indeed, evidence suggests that these cells play important roles in diverse diseases, such as inflammation, autoimmune disease, and infection. Recent studies elucidating the molecular mechanisms by which CD1 presents lipid antigens suggest that defects in these mechanisms could contribute to the activities of CD1-reactive T cells. Thus, improving our understanding of these mechanisms could lead to new and effective therapeutic approaches to CD1-associated diseases. In this review, we discuss the CD1-mediated antigen presentation system and its roles in mucosal tissue immunity.
Collapse
Affiliation(s)
- Seohyun Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Sumin Cho
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Ji Hyung Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
41
|
Yoo JS, Oh SF. Unconventional immune cells in the gut mucosal barrier: regulation by symbiotic microbiota. Exp Mol Med 2023; 55:1905-1912. [PMID: 37696893 PMCID: PMC10545787 DOI: 10.1038/s12276-023-01088-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/19/2023] [Accepted: 08/08/2023] [Indexed: 09/13/2023] Open
Abstract
The mammalian gut is the most densely colonized organ by microbial species, which are in constant contact with the host throughout life. Hosts have developed multifaceted cellular and molecular mechanisms to distinguish and respond to benign and pathogenic bacteria. In addition to relatively well-characterized innate and adaptive immune cells, a growing body of evidence shows additional important players in gut mucosal immunity. Among them, unconventional immune cells, including innate lymphoid cells (ILCs) and unconventional T cells, are essential for maintaining homeostasis. These cells rapidly respond to bacterial signals and bridge the innate immunity and adaptive immunity in the mucosal barrier. Here, we focus on the types and roles of these immune cells in physiological and pathological conditions as prominent mechanisms by which the host immune system communicates with the gut microbiota in health and diseases.
Collapse
Affiliation(s)
- Ji-Sun Yoo
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Sungwhan F Oh
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, USA.
- Graduate Program in Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
42
|
Park JS, Gazzaniga FS, Kasper DL, Sharpe AH. Microbiota-dependent regulation of costimulatory and coinhibitory pathways via innate immune sensors and implications for immunotherapy. Exp Mol Med 2023; 55:1913-1921. [PMID: 37696895 PMCID: PMC10545783 DOI: 10.1038/s12276-023-01075-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 07/11/2023] [Accepted: 07/19/2023] [Indexed: 09/13/2023] Open
Abstract
Our bodies are inhabited by trillions of microorganisms. The host immune system constantly interacts with the microbiota in barrier organs, including the intestines. Over decades, numerous studies have shown that our mucosal immune system is dynamically shaped by a variety of microbiota-derived signals. Elucidating the mediators of these interactions is an important step for understanding how the microbiota is linked to mucosal immune homeostasis and gut-associated diseases. Interestingly, the efficacy of cancer immunotherapies that manipulate costimulatory and coinhibitory pathways has been correlated with the gut microbiota. Moreover, adverse effects of these therapies in the gut are linked to dysregulation of the intestinal immune system. These findings suggest that costimulatory pathways in the immune system might serve as a bridge between the host immune system and the gut microbiota. Here, we review mechanisms by which commensal microorganisms signal immune cells and their potential impact on costimulation. We highlight how costimulatory pathways modulate the mucosal immune system through not only classical antigen-presenting cells but also innate lymphocytes, which are highly enriched in barrier organs. Finally, we discuss the adverse effects of immune checkpoint inhibitors in the gut and the possible relationship with the gut microbiota.
Collapse
Affiliation(s)
- Joon Seok Park
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Francesca S Gazzaniga
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA, 02129, USA
- Department of Pathology, Harvard Medical School, Boston, MA, 02115, USA
| | - Dennis L Kasper
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Arlene H Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
43
|
Vallet N, Salmona M, Malet-Villemagne J, Bredel M, Bondeelle L, Tournier S, Mercier-Delarue S, Cassonnet S, Ingram B, Peffault de Latour R, Bergeron A, Socié G, Le Goff J, Lepage P, Michonneau D. Circulating T cell profiles associate with enterotype signatures underlying hematological malignancy relapses. Cell Host Microbe 2023; 31:1386-1403.e6. [PMID: 37463582 DOI: 10.1016/j.chom.2023.06.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 05/23/2023] [Accepted: 06/19/2023] [Indexed: 07/20/2023]
Abstract
Early administration of azithromycin after allogeneic hematopoietic stem cell transplantation was shown to increase the relapse of hematological malignancies. To determine the impact of azithromycin on the post-transplant gut ecosystem and its influence on relapse, we characterized overtime gut bacteriome, virome, and metabolome of 55 patients treated with azithromycin or a placebo. We describe four enterotypes and the network of associated bacteriophage species and metabolic pathways. One enterotype associates with sustained remission. One taxon from Bacteroides specifically associates with relapse, while two from Bacteroides and Prevotella correlate with complete remission. These taxa are associated with lipid, pentose, and branched-chain amino acid metabolic pathways and several bacteriophage species. Enterotypes and taxa associate with exhausted T cells and the functional status of circulating immune cells. These results illustrate how an antibiotic influences a complex network of gut bacteria, viruses, and metabolites and may promote cancer relapse through modifications of immune cells.
Collapse
Affiliation(s)
- Nicolas Vallet
- Université de Paris Cité, INSERM U976, 75010 Paris, France
| | - Maud Salmona
- Université de Paris Cité, INSERM U976, 75010 Paris, France; Virology Department, AP-HP, Saint-Louis Hospital, 75010 Paris, France
| | - Jeanne Malet-Villemagne
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Domaine de Vilvert, 78350 Jouy-en-Josas, France
| | - Maxime Bredel
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Domaine de Vilvert, 78350 Jouy-en-Josas, France
| | - Louise Bondeelle
- Pneumology Unit, AP-HP, Saint-Louis Hospital, 75010 Paris, France
| | - Simon Tournier
- Core Facilities, Saint-Louis Research Institute, Université de Paris Cité, UAR 2030/US 53, 75010 Paris, France
| | | | - Stéphane Cassonnet
- Service de Biostatistique et Information Médicale, AP-HP, Saint-Louis Hospital, 75010 Paris, France
| | | | - Régis Peffault de Latour
- Hematology Transplantation, AP-HP, Saint-Louis Hospital, 1 avenue Claude Vellefaux, 75010 Paris, France; Cryostem Consortium, 13382 Marseille, France
| | - Anne Bergeron
- Pneumology Department, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Gérard Socié
- Université de Paris Cité, INSERM U976, 75010 Paris, France; Hematology Transplantation, AP-HP, Saint-Louis Hospital, 1 avenue Claude Vellefaux, 75010 Paris, France
| | - Jérome Le Goff
- Université de Paris Cité, INSERM U976, 75010 Paris, France; Virology Department, AP-HP, Saint-Louis Hospital, 75010 Paris, France
| | - Patricia Lepage
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Domaine de Vilvert, 78350 Jouy-en-Josas, France
| | - David Michonneau
- Université de Paris Cité, INSERM U976, 75010 Paris, France; Hematology Transplantation, AP-HP, Saint-Louis Hospital, 1 avenue Claude Vellefaux, 75010 Paris, France.
| |
Collapse
|
44
|
Cameron G, Nguyen T, Ciula M, Williams SJ, Godfrey DI. Glycolipids from the gut symbiont Bacteroides fragilis are agonists for natural killer T cells and induce their regulatory differentiation. Chem Sci 2023; 14:7887-7896. [PMID: 37502334 PMCID: PMC10370605 DOI: 10.1039/d3sc02124f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/26/2023] [Indexed: 07/29/2023] Open
Abstract
Natural Killer T (NKT) cells are a lipid-antigen reactive T cell subset that is restricted to the antigen presenting molecule CD1d. They possess diverse functional properties that contribute to inflammatory and regulatory immune responses. The most studied lipid antigen target for these T cells is α-galactosylceramide (αGC). The commensal organism Bacteroides fragilis (B. fragilis) produces several forms of αGC, but conflicting information exists about the influence of these lipids on NKT cells. Herein, we report the total synthesis of a major form of αGC from B. fragilis (Bf αGC), and several analogues thereof. We confirm the T cell receptor (TCR)-mediated recognition of these glycolipids by mouse and human NKT cells. Despite the natural structure of Bf αGC containing lipid branching that limits potency, we demonstrate that Bf αGC drives mouse NKT cells to proliferate and differentiate into producers of the immunoregulatory cytokine, interleukin-10 (IL-10). These Bf αGC-experienced NKT cells display regulatory function by inhibiting the expansion of naïve NKT cells upon subsequent exposure to this antigen. Moreover, this regulatory activity impacts more than just NKT cells, as demonstrated by the NKT cell-mediated inhibition of antigen-stimulated mucosal-associated invariant T (MAIT) cells (a T cell subset restricted to a different antigen presenting molecule, MR1). These findings reveal that B. fragilis-derived NKT cell agonists may have broad immunoregulatory activity, providing insight into the mechanisms influencing immune tolerance to commensal bacteria and highlighting a potential means to manipulate NKT cell function for therapeutic benefit.
Collapse
Affiliation(s)
- Garth Cameron
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne Melbourne VIC 3000 Australia
| | - Tram Nguyen
- School of Chemistry, University of Melbourne Parkville VIC 3010 Australia
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne Parkville VIC 3010 Australia
| | - Marcin Ciula
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne Melbourne VIC 3000 Australia
| | - Spencer J Williams
- School of Chemistry, University of Melbourne Parkville VIC 3010 Australia
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne Parkville VIC 3010 Australia
| | - Dale I Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne Melbourne VIC 3000 Australia
| |
Collapse
|
45
|
Luo J, Chen Z, Castellano D, Bao B, Han W, Li J, Kim G, An D, Lu W, Wu C. Lipids regulate peripheral serotonin release via gut CD1d. Immunity 2023; 56:1533-1547.e7. [PMID: 37354904 PMCID: PMC10527042 DOI: 10.1016/j.immuni.2023.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/04/2023] [Accepted: 06/01/2023] [Indexed: 06/26/2023]
Abstract
The crosstalk between the immune and neuroendocrine systems is critical for intestinal homeostasis and gut-brain communications. However, it remains unclear how immune cells participate in gut sensation of hormones and neurotransmitters release in response to environmental cues, such as self-lipids and microbial lipids. We show here that lipid-mediated engagement of invariant natural killer T (iNKT) cells with enterochromaffin (EC) cells, a subset of intestinal epithelial cells, promoted peripheral serotonin (5-HT) release via a CD1d-dependent manner, regulating gut motility and hemostasis. We also demonstrated that inhibitory sphingolipids from symbiotic microbe Bacteroides fragilis represses 5-HT release. Mechanistically, CD1d ligation on EC cells transduced a signal and restrained potassium conductance through activation of protein tyrosine kinase Pyk2, leading to calcium influx and 5-HT secretion. Together, our data reveal that by engaging with iNKT cells, gut chemosensory cells selectively perceive lipid antigens via CD1d to control 5-HT release, modulating intestinal and systemic homeostasis.
Collapse
Affiliation(s)
- Jialie Luo
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Zuojia Chen
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - David Castellano
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Bin Bao
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wenyan Han
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Jian Li
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Girak Kim
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Dingding An
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wei Lu
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
46
|
Wegrecki M. CD1a-mediated immunity from a molecular perspective. Mol Immunol 2023; 158:43-53. [PMID: 37116273 DOI: 10.1016/j.molimm.2023.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 03/24/2023] [Accepted: 04/24/2023] [Indexed: 04/30/2023]
Abstract
Human CD1a is a non-polymorphic glycoprotein that presents lipid antigens to T cells. The most obvious role of CD1a is associated with its expression on Langerhans cells in epidermis, where it is involved in responses to pathogens. Antigen-specific T cells are believed to co-recognise CD1a presenting bacterial antigens such as species of lipopeptides from Mycobacterium tuberculosis. Further, human skin contains large amount of endogenous lipids that can activate distinct subsets of CD1a-restricted autoreactive T cells, mostly belonging to the αβ lineage, which are abundant in human blood and skin and are important for skin homeostasis in healthy individuals. CD1a and CD1a-restricted T cells have been linked to certain autoimmune conditions such as psoriasis, atopic dermatitis and contact hypersensitivity becoming a potential candidate for clinical interventions. A significant progress has been made in the last twenty years towards our understanding of the molecular processes that orchestrate CD1a-lipid binding, antigen presentation and mechanism of CD1a recognition by αβ and γδ T cells. This review summarises the recent developments within the field of CD1a-mediated immunity from a molecular perspective.
Collapse
Affiliation(s)
- Marcin Wegrecki
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
47
|
Dang X, Huang Q, He YQ, Gaitán-Espitia JD, Zhang T, Thiyagarajan V. Ocean acidification drives gut microbiome changes linked to species-specific immune defence. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 256:106413. [PMID: 36801178 DOI: 10.1016/j.aquatox.2023.106413] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 12/10/2022] [Accepted: 01/28/2023] [Indexed: 06/18/2023]
Abstract
Ocean acidification (OA) has important effects on the intrinsic phenotypic characteristics of many marine organisms. Concomitantly, OA can alter the extended phenotypes of these organisms by perturbing the structure and function of their associated microbiomes. It is unclear, however, the extent to which interactions between these levels of phenotypic change can modulate the capacity for resilience to OA. Here, we explored this theoretical framework assessing the influence of OA on intrinsic (immunological responses and energy reserve) and extrinsic (gut microbiome) phenotypic characteristics and the survival of important calcifiers, the edible oysters Crassostrea angulata and C. hongkongensis. After one-month exposure to experimental OA (pH 7.4) and control (pH 8.0) conditions, we found species-specific responses characterised by elevated stress (hemocyte apoptosis) and decreased survival in the coastal species (C. angulata) compared with the estuarine species (C. hongkongensis). Phagocytosis of hemocytes was not affected by OA but in vitro bacterial clearance capability decreased in both species. Gut microbial diversity decreased in C. angulata but not in C. hongkongensis. Overall, C. hongkongensis was capable of maintaining the homeostasis of the immune system and energy supply under OA. In contrast, C. angulata's immune function was suppressed, and the energy reserve was imbalanced, which might be attributed to the declined microbial diversity and the functional loss of essential bacteria in the guts. This study highlights a species-specific response to OA determined by genetic background and local adaptation, shedding light on the understanding of host-microbiota-environment interactions in future coastal acidification.
Collapse
Affiliation(s)
- Xin Dang
- The Swire Institute of Marine Science, School of Biological Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Qi Huang
- The Swire Institute of Marine Science, School of Biological Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China; Department of Civil Engineering, Environmental Microbiome Engineering and Biotechnology Lab, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yuan-Qiu He
- The Swire Institute of Marine Science, School of Biological Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Juan Diego Gaitán-Espitia
- The Swire Institute of Marine Science, School of Biological Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Tong Zhang
- Department of Civil Engineering, Environmental Microbiome Engineering and Biotechnology Lab, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Vengatesen Thiyagarajan
- The Swire Institute of Marine Science, School of Biological Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| |
Collapse
|
48
|
Li Y, Yan Y, Fu H, Jin S, He S, Wang Z, Dong G, Li B, Guo S. Does diet or macronutrients intake drive the structure and function of gut microbiota? Front Microbiol 2023; 14:1126189. [PMID: 36860485 PMCID: PMC9970161 DOI: 10.3389/fmicb.2023.1126189] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 01/23/2023] [Indexed: 02/16/2023] Open
Abstract
Shift of ingestive behavior is an important strategy for animals to adapt to change of the environment. We knew that shifts in animal dietary habits lead to changes in the structure of the gut microbiota, but we are not sure about if changes in the composition and function of the gut microbiota respond to changes in the nutrient intake or food items. To investigate how animal feeding strategies affect nutrient intakes and thus alter the composition and digestion function of gut microbiota, we selected a group of wild primate group for the study. We quantified their diet and macronutrients intake in four seasons of a year, and instant fecal samples were analyzed by high-throughput sequencing of 16S rRNA and metagenomics. These results demonstrated that the main reason that causes seasonal shifts of gut microbiota is the macronutrient variation induced by seasonal dietary differences. Gut microbes can help to compensate for insufficient macronutrients intake of the host through microbial metabolic functions. This study contributes to a deeper understanding of the causes of seasonal variation in host-microbial variation in wild primates.
Collapse
Affiliation(s)
- Yuhang Li
- Shaanxi Key Laboratory for Animal Conservation, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Yujie Yan
- Shaanxi Key Laboratory for Animal Conservation, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Hengguang Fu
- Shaanxi Key Laboratory for Animal Conservation, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Shiyu Jin
- Shaanxi Key Laboratory for Animal Conservation, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Shujun He
- Shaanxi Key Laboratory for Animal Conservation, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Zi Wang
- Shaanxi Key Laboratory for Animal Conservation, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Guixin Dong
- Guangdong Chimelong Group Co., Ltd., Guangzhou, China,Guangdong South China Rare Wild Animal Species Conservation Center, Zhuhai, China
| | - Baoguo Li
- Shaanxi Key Laboratory for Animal Conservation, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China,Shaanxi Institute of Zoology, Xi’an, China
| | - Songtao Guo
- Shaanxi Key Laboratory for Animal Conservation, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China,Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, China,*Correspondence: Songtao Guo,
| |
Collapse
|
49
|
Abstract
Metabolites produced by commensal gut microbes impact host health through their recognition by the immune system and their influence on numerous metabolic pathways. Notably, the gut microbiota can both transform and synthesize lipids as well as break down dietary lipids to generate secondary metabolites with host modulatory properties. Although lipids have largely been consigned to structural roles, particularly in cell membranes, recent research has led to an increased appreciation of their signaling activities, with potential impacts on host health and physiology. This review focuses on studies that highlight the functions of bioactive lipids in mammalian physiology, with a special emphasis on immunity and metabolism.
Collapse
Affiliation(s)
- Eric M Brown
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Jon Clardy
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Blavatnik Institute, Boston, MA 02115, USA
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
50
|
Shi L, Xu Y, Feng M. Role of Gut Microbiome in Immune Regulation and Immune Checkpoint Therapy of Colorectal Cancer. Dig Dis Sci 2023; 68:370-379. [PMID: 36575326 DOI: 10.1007/s10620-022-07689-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/01/2022] [Indexed: 12/29/2022]
Abstract
Colorectal cancer (CRC) is one of the most frequent gastrointestinal malignant tumors worldwide. Immune checkpoint therapies (ICTs) have been proven to be a reliable treatment for some subtypes of CRC. Gut microbiome is closely involved in intestinal carcinogenesis through the regulation of local immune and inflammation of colonic mucosa. Numerous studies have demonstrated that the immunotherapeutic efficacy of CRC and other kinds of cancer is influenced by the immunosuppressive microenvironment constituted by intestinal microbiome and their metabolites. This Review will discuss the recent advances in how gut microbiome can modify the immune microenvironment and its potential role in ICTs of CRC.
Collapse
Affiliation(s)
- Linsen Shi
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Yumei Xu
- Department of Radiation Oncology Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Min Feng
- Department of Gastrointestinal Surgery, The Affiliated Drum Tower Hospital of NanJing Medical University, 321 Zhongshan Road, Nanjing, 210002, People's Republic of China.
| |
Collapse
|