1
|
Sun R, Zhang Y, Zhao X, Tang T, Cao Y, Yang L, Tian Y, Zhang Z, Zhang P, Xu F. Temporal and Spatial Metabolic Shifts Revealing the Transition from Ulcerative Colitis to Colitis-Associated Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2412551. [PMID: 39840505 DOI: 10.1002/advs.202412551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/17/2024] [Indexed: 01/23/2025]
Abstract
Patients with ulcerative colitis (UC) have a higher risk of developing colorectal cancer (CRC), however, the metabolic shifts during the UC-to-CRC transition remain elusive. In this study, an AOM-DSS-induced three-stage colitis-associated colorectal cancer (CAC) model is constructed and targeted metabolomics analysis and pathway enrichment are performed, uncovering the metabolic changes in this transition. Spatial metabolic trajectories in the "normal-to-normal adjacent tissue (NAT)-to-tumor" transition, and temporal metabolic trajectories in the "colitis-to-dysplasia-to-carcinoma" transition are identified through K-means clustering of 74 spatially and 77 temporally differential metabolites, respectively. The findings reveal two distinct metabolic profile categories during the inflammation-to-cancer progression: those with consistent changes, either increasing (e.g., kynurenic acid, xanthurenic acid) or decreasing (e.g., long-chain fatty acids, LCFAs), and those enriched at specific disease stages (e.g., serotonin). Further analysis of metabolites with consistent temporal trends identifies eicosapentaenoic acid (EPA) as a key metabolite, potentially exerting anti-inflammatory and anti-cancer effects by inhibiting insulin-like growth factor binding protein 5 (IGFBP5). This study reveals novel metabolic mechanisms underlying the transition from UC to CAC and suggests potential targets to delay the progression.
Collapse
Affiliation(s)
- Ruiqi Sun
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Yuanyuan Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Xian Zhao
- Department of Pharmacy, Drum Tower Hospital, China Pharmaceutical University, Nanjing, 210008, P. R. China
| | - Tian Tang
- School of Pharmacy, Air Force Medical University, Xi'an, 710032, P. R. China
| | - Yuepeng Cao
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, 210009, P. R. China
| | - Liu Yang
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, 210009, P. R. China
| | - Yuan Tian
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Zunjian Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Pei Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Fengguo Xu
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, P. R. China
| |
Collapse
|
2
|
Gong Z, Yi H, Zhang J, Li W, Wang H, Guo P, Li C, Pan A, Cao Y, Lu Z, Jiang H. Role of Arg1 + ILC2s and ILCregs in gestational diabetes progression. Sci Rep 2025; 15:1580. [PMID: 39794391 PMCID: PMC11724119 DOI: 10.1038/s41598-025-85452-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Innate lymphoid cells (ILCs) are a newly discovered subset of immune cells that are responsible for regulation of the immune microenvironment. In particular, the ILC categories ILC2s and regulatory ILCs (ILCregs) are associated with immunosuppression and chronic inflammation. Chronic low-grade inflammation leads to insulin resistance, a major etiological factor in gestational diabetes mellitus (GDM). However, the influence of ILCs on GDM remains poorly understood. Therefore, this study aims to investigate the potential role of ILCs in the development and progression of GDM. This study included 19 patients diagnosed with GDM and 19 age- and body mass index-matched individuals in the control group. Flow cytometry was employed to assess the frequency and function of ILC subsets in peripheral blood (PB), cord blood (CB), and placental tissues. Additionally, ELISA was utilized to measure the levels of the cytokines TNF-α, IFN-γ, TGF-β, and IL-4/10/13/22 in the serum samples of patients. Compared to the control group with normal pregnancy, significantly elevated levels of ILC2s, Arg1+ILC2s, and ILCregs were detected in the PB, CB, and placental tissues of the GDM group. With regard to inflammation-related cytokines, the levels of IL-13/22 in PB serum were significantly elevated, while the TGF-β levels were significantly reduced in the GDM group compared to the control group (CG). Further, in the CB serum samples, IL-13 levels were elevated in the GDM group. Additionally, a negative correlation was observed between the number of ILC3s and the number of ILCregs present in umbilical cord blood, while the IL-13 level in peripheral blood was negatively correlated with the number of ILC3s. The present findings indicate that chronic low-grade inflammation mediated by Arg-1+ILC2s and ILCregs is closely associated with the pathogenesis of GDM.
Collapse
Affiliation(s)
- Zhangyun Gong
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Haixing Yi
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jie Zhang
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Wan Li
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Hao Wang
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Peipei Guo
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Caihua Li
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Anan Pan
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yunxia Cao
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Zhimin Lu
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Huanhuan Jiang
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
3
|
Dawkins JJ, Gerber GK. MMETHANE: interpretable AI for predicting host status from microbial composition and metabolomics data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.13.628441. [PMID: 39713330 PMCID: PMC11661223 DOI: 10.1101/2024.12.13.628441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Metabolite production, consumption, and exchange are intimately involved with host health and disease, as well as being key drivers of host-microbiome interactions. Despite the increasing prevalence of datasets that jointly measure microbiome composition and metabolites, computational tools for linking these data to the status of the host remain limited. To address these limitations, we developed MMETHANE, an open-source software package that implements a purpose-built deep learning model for predicting host status from paired microbial sequencing and metabolomic data. MMETHANE incorporates prior biological knowledge, including phylogenetic and chemical relationships, and is intrinsically interpretable, outputting an English-language set of rules that explains its decisions. Using a compendium of six datasets with paired microbial composition and metabolomics measurements, we showed that MMETHANE always performed at least on par with existing methods, including blackbox machine learning techniques, and outperformed other methods on >80% of the datasets evaluated. We additionally demonstrated through two cases studies analyzing inflammatory bowel disease gut microbiome datasets that MMETHANE uncovers biologically meaningful links between microbes, metabolites, and disease status.
Collapse
|
4
|
Yuan Y, Hu H, Sun Z, Wang W, Wang Z, Zheng M, Xing Y, Zhang W, Wang M, Lu X, Li Y, Liang C, Lin Z, Xie C, Li J, Mao T. Combining Metagenomics, Network Pharmacology and RNA-Seq Strategies to Reveal the Therapeutic Effects and Mechanisms of Qingchang Wenzhong Decoction on Inflammatory Bowel Disease in Mice. Drug Des Devel Ther 2024; 18:4273-4289. [PMID: 39347539 PMCID: PMC11438451 DOI: 10.2147/dddt.s473688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/10/2024] [Indexed: 10/01/2024] Open
Abstract
Background Inflammatory bowel disease (IBD) is a chronic and recurrent inflammatory disease that lacks effective treatments. Qingchang Wenzhong Decoction (QCWZD) is a clinically effective herbal prescription that has been proven to attenuate intestinal inflammation in IBD. However, its molecular mechanism of action has not been clearly elucidated. Purpose We aimed to probe the mechanism of QCWZD for the treatment of IBD. Methods The dextran sulfate sodium (DSS)-induced mouse model of IBD was used to identify the molecular targets involved in the mechanism of action of QCWZD. Metagenomics sequencing was utilized to analyze the differences in gut microbiota and the functional consequences of these changes. Network pharmacology combined with RNA sequencing (RNA-seq) were employed to predict the molecular targets and mechanism of action of QCWZD, and were validated through in vivo experiments. Results Our results demonstrated that QCWZD treatment alleviated intestinal inflammation and accelerated intestinal mucosal healing that involved restoration of microbial homeostasis. This hypothesis was supported by the results of bacterial metagenomics sequencing that showed attenuation of gut dysbiosis by QCWZD treatment, especially the depletion of the pathogenic bacterial genus Bacteroides, while increasing the beneficial microorganism Akkermansia muciniphila that led to altered bacterial gene functions, such as metabolic regulation. Network pharmacology and RNA-seq analyses showed that Th17 cell differentiation plays an important role in QCWZD-based treatment of IBD. This was confirmed by in vivo experiments showing a marked decrease in the percentage of CD3+CD4+IL-17+ (Th17) cells. Furthermore, our results also showed that the key factors associated with Th17 cell differentiation (IL-17, NF-κB, TNF-α and IL-6) in the colon were significantly reduced in QCWZD-treated colitis mice. Conclusion QCWZD exerted beneficial effects in the treatment of IBD by modulating microbial homeostasis while inhibiting Th17 cell differentiation and its associated pathways, providing a novel and promising therapeutic strategy for the treatment of IBD.
Collapse
Affiliation(s)
- Yali Yuan
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
- Hebei North University, Zhangjiakou, Hebei, People’s Republic of China
| | - Hairong Hu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Zhongmei Sun
- Tianjin Nankai Hospital, Tianjin, People’s Republic of China
| | - Wenting Wang
- Beitaipingzhuang Community Health Service Center, Beijing, People’s Republic of China
| | - Zhibin Wang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | | | - Yunqi Xing
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Wenji Zhang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Muyuan Wang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Xinyu Lu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Yitong Li
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Chengtao Liang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Zhengdao Lin
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Chune Xie
- Shenzhen Bao’an Traditional Chinese Medicine Hospital, Shenzhen, People’s Republic of China
| | - Junxiang Li
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Tangyou Mao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| |
Collapse
|
5
|
Wang H, Chen D, Li H, Fu C, Fang L, Wang R, Xu J. Bifidobacterium regulates premature infant gut metabolites, reducing serum inflammatory factors: a randomised controlled trial. Pediatr Res 2024:10.1038/s41390-024-03552-2. [PMID: 39271904 DOI: 10.1038/s41390-024-03552-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 08/17/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Analyse the effects of Bifidobacterium BB-12 on intestinal metabolites and serum inflammatory factors in premature infants. METHODS 71 premature infants at gestational age of ≤32 weeks were randomly divided into the probiotic (n = 36) and control (n = 35) groups. Faecal and blood samples were collected from the two groups of premature infants at the 2nd and 4th week of life for intestinal metabolite detection and assessment of the level of the serum inflammatory markers TLR4, NF- κ B, IL-1β, and TNF- α. RESULTS Compared to the control group, the probiotic group contained more amino acids, these elements were enriched on multiple amino acid metabolic pathways, and the probiotic group showed significantly lower levels of the serum inflammatory markers TLR4, NF-κB, IL-1β, and TNF-α. Finally, the probiotic group showed a lower incidence of feeding intolerance. CONCLUSIONS The administration of Bifidobacterium BB-12 is associated with increasing the levels of glutamine, glutamic acid, and kynurenine in the gut of premature infants, and associated with reducing the levels of TLR4 and NF-κB in the serum, further decreasing the secretion of the pro-inflammatory factors IL-1β and TNF-α, and alleviating systemic inflammatory reactions, thereby reducing the incidence of feeding intolerance. IMPACT 1. The use of Bifidobacterium BB-12 in premature infants can increase the levels of amino acids in the intestine. 2. Increases in Bifidobacterium BB-12 may decrease the serum levels of TLR4, NF-κB, IL-1β, and TNF-α. 3. Kynurenine may improve the prognosis of preterm infants by reducing inflammation. 4. Bifidobacterium BB-12 may improve the feeding tolerance of premature infants, thus reducing the incidence of feeding intolerance.
Collapse
Affiliation(s)
- He Wang
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Dongmei Chen
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Huamei Li
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Chunyan Fu
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Lingyu Fang
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Ruiquan Wang
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Jinglin Xu
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China.
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China.
| |
Collapse
|
6
|
Zhang Y, Higgins CB, Tica S, Adams JA, Sun J, Kelly SC, Zong X, Dietzen DJ, Pietka T, Ballentine SJ, Shriver LP, Patti GJ, Cao Y, DeBosch BJ. Hierarchical tricarboxylic acid cycle regulation by hepatocyte arginase 2 links the urea cycle to oxidative metabolism. Cell Metab 2024; 36:2069-2085.e8. [PMID: 39116884 DOI: 10.1016/j.cmet.2024.07.007] [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: 08/23/2023] [Revised: 03/19/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024]
Abstract
Urea cycle impairment and its relationship to obesity and inflammation remained elusive, partly due to the dramatic clinical presentation of classical urea cycle defects. We generated mice with hepatocyte-specific arginase 2 deletion (Arg2LKO) and revealed a mild compensated urea cycle defect. Stable isotope tracing and respirometry revealed hepatocyte urea and TCA cycle flux defects, impaired mitochondrial oxidative metabolism, and glutamine anaplerosis despite normal energy and glucose homeostasis during early adulthood. Yet during middle adulthood, chow- and diet-induced obese Arg2LKO mice develop exaggerated glucose and lipid derangements, which are reversible by replacing the TCA cycle oxidative substrate nicotinamide adenine dinucleotide. Moreover, serum-based hallmarks of urea, TCA cycle, and mitochondrial derangements predict incident fibroinflammatory liver disease in 106,606 patients nearly a decade in advance. The data reveal hierarchical urea-TCA cycle control via ARG2 to drive oxidative metabolism. Moreover, perturbations in this circuit may causally link urea cycle compromise to fibroinflammatory liver disease.
Collapse
Affiliation(s)
- Yiming Zhang
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Cassandra B Higgins
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Stefani Tica
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joshua A Adams
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jiameng Sun
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shannon C Kelly
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xiaoyu Zong
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dennis J Dietzen
- Department of Pediatrics, Washington University School of Medicine, Laboratory Services, St. Louis Children's Hospital, St. Louis, MO 63110, USA
| | - Terri Pietka
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Samuel J Ballentine
- Department of Pathology & Immunology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Leah P Shriver
- Department of Pediatrics, Washington University School of Medicine, Laboratory Services, St. Louis Children's Hospital, St. Louis, MO 63110, USA; Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Gary J Patti
- Department of Pediatrics, Washington University School of Medicine, Laboratory Services, St. Louis Children's Hospital, St. Louis, MO 63110, USA; Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Yin Cao
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brian J DeBosch
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
7
|
Zhang B, Magnaye KM, Stryker E, Moltzau-Anderson J, Porsche CE, Hertz S, McCauley KE, Smith BJ, Zydek M, Pollard KS, Ma A, El-Nachef N, Lynch SV. Sustained mucosal colonization and fecal metabolic dysfunction by Bacteroides associates with fecal microbial transplant failure in ulcerative colitis patients. Sci Rep 2024; 14:18558. [PMID: 39122767 PMCID: PMC11316000 DOI: 10.1038/s41598-024-62463-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 05/16/2024] [Indexed: 08/12/2024] Open
Abstract
Fecal microbial transplantation (FMT) offers promise for treating ulcerative colitis (UC), though the mechanisms underlying treatment failure are unknown. This study harnessed longitudinally collected colonic biopsies (n = 38) and fecal samples (n = 179) from 19 adults with mild-to-moderate UC undergoing serial FMT in which antimicrobial pre-treatment and delivery mode (capsules versus enema) were assessed for clinical response (≥ 3 points decrease from the pre-treatment Mayo score). Colonic biopsies underwent dual RNA-Seq; fecal samples underwent parallel 16S rRNA and shotgun metagenomic sequencing as well as untargeted metabolomic analyses. Pre-FMT, the colonic mucosa of non-responsive (NR) patients harbored an increased burden of bacteria, including Bacteroides, that expressed more antimicrobial resistance genes compared to responsive (R) patients. NR patients also exhibited muted mucosal expression of innate immune antimicrobial response genes. Post-FMT, NR and R fecal microbiomes and metabolomes exhibited significant divergence. NR metabolomes had elevated concentrations of immunostimulatory compounds including sphingomyelins, lysophospholipids and taurine. NR fecal microbiomes were enriched for Bacteroides fragilis and Bacteroides salyersiae strains that encoded genes capable of taurine production. These findings suggest that both effective mucosal microbial clearance and reintroduction of bacteria that reshape luminal metabolism associate with FMT success and that persistent mucosal and fecal colonization by antimicrobial-resistant Bacteroides species may contribute to FMT failure.
Collapse
Affiliation(s)
- Bing Zhang
- Department of Medicine, Division of Gastrointestinal and Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Kevin M Magnaye
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- The Benioff Center for Microbiome Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Emily Stryker
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Jacqueline Moltzau-Anderson
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- The Benioff Center for Microbiome Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Cara E Porsche
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Sandra Hertz
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark
| | - Kathryn E McCauley
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Byron J Smith
- The Gladstone Institutes, Data Science and Biotechnology, San Francisco, CA, 94158, USA
| | - Martin Zydek
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Katherine S Pollard
- The Gladstone Institutes, Data Science and Biotechnology, San Francisco, CA, 94158, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, 94158, USA
- Chan Zuckerberg Biohub, San Francisco, CA University of California, San Francisco, CA, 94158, USA
| | - Averil Ma
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Najwa El-Nachef
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Division of Gastroenterology, Henry Ford Health System, Detroit, MI, 48208, USA
| | - Susan V Lynch
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA.
- The Benioff Center for Microbiome Medicine, University of California San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
8
|
Jaswal K, Todd OA, Flores Audelo RC, Santus W, Paul S, Singh M, Miao J, Underhill DM, Peters BM, Behnsen J. Commensal Yeast Promotes Salmonella Typhimurium Virulence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.08.606421. [PMID: 39211098 PMCID: PMC11360897 DOI: 10.1101/2024.08.08.606421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Enteric pathogens engage in complex interactions with the host and the resident microbiota to establish gut colonization. Although mechanistic interactions between enteric pathogens and bacterial commensals have been extensively studied, whether and how commensal fungi affect pathogenesis of enteric infections remains largely unknown. Here we show that colonization with the common human gut commensal fungus Candida albicans worsened infections with the enteric pathogen Salmonella enterica serovar Typhimurium. Presence of C. albicans in the mouse gut increased Salmonella cecum colonization and systemic dissemination. We investigated the underlying mechanism and found that Salmonella binds to C. albicans via Type 1 fimbriae and uses its Type 3 Secretion System (T3SS) to deliver effector proteins into C. albicans . A specific effector, SopB, was sufficient to manipulate C. albicans metabolism, triggering increased arginine biosynthesis in C. albicans and the release of millimolar amounts of arginine into the extracellular environment. The released arginine, in turn, induced T3SS expression in Salmonella , increasing its invasion of epithelial cells. C. albicans deficient in arginine production was unable to increase Salmonella virulence in vitro or in vivo . In addition to modulating pathogen invasion, arginine also directly influenced the host response to infection. Arginine-producing C. albicans dampened the inflammatory response during Salmonella infection, whereas C. albicans deficient in arginine production did not. Arginine supplementation in the absence of C. albicans increased the systemic spread of Salmonella and decreased the inflammatory response, phenocopying the presence of C. albicans . In summary, we identified C. albicans colonization as a susceptibility factor for disseminated Salmonella infection, and arginine as a central metabolite in the cross-kingdom interaction between fungi, bacteria, and host.
Collapse
|
9
|
Liu M, Guo S, Wang L. Systematic review of metabolomic alterations in ulcerative colitis: unveiling key metabolic signatures and pathways. Therap Adv Gastroenterol 2024; 17:17562848241239580. [PMID: 38560428 PMCID: PMC10981261 DOI: 10.1177/17562848241239580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
Background Despite numerous metabolomic studies on ulcerative colitis (UC), the results have been highly variable, making it challenging to identify key metabolic abnormalities in UC. Objectives This study aims to uncover key metabolites and metabolic pathways in UC by analyzing existing metabolomics data. Design A systematic review. Data sources and methods We conducted a comprehensive search in databases (PubMed, Cochrane Library, Embase, and Web of Science) and relevant study references for metabolomic research on UC up to 28 December 2022. Significant metabolite differences between UC patients and controls were identified, followed by an analysis of relevant metabolic pathways. Results This review incorporated 78 studies, identifying 2868 differentially expressed metabolites between UC patients and controls. The metabolites were predominantly from 'lipids and lipid-like molecules' and 'organic acids and derivatives' superclasses. We found 101 metabolites consistently altered in multiple datasets within the same sample type and 78 metabolites common across different sample types. Of these, 62 metabolites exhibited consistent regulatory trends across various datasets or sample types. Pathway analysis revealed 22 significantly altered metabolic pathways, with 6 pathways being recurrently enriched across different sample types. Conclusion This study elucidates key metabolic characteristics in UC, offering insights into molecular mechanisms and biomarker discovery for the disease. Future research could focus on validating these findings and exploring their clinical applications.
Collapse
Affiliation(s)
- Meiling Liu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Siyi Guo
- Chongqing Medical University, Chongqing, China
| | - Liang Wang
- Chongqing Medical University, Chongqing, China
| |
Collapse
|
10
|
Wang S, Gong X, Xiao F, Yang Y. Recent advances in host-focused molecular tools for investigating host-gut microbiome interactions. Front Microbiol 2024; 15:1335036. [PMID: 38605718 PMCID: PMC11007152 DOI: 10.3389/fmicb.2024.1335036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 03/18/2024] [Indexed: 04/13/2024] Open
Abstract
Microbial communities in the human gut play a significant role in regulating host gene expression, influencing a variety of biological processes. To understand the molecular mechanisms underlying host-microbe interactions, tools that can dissect signaling networks are required. In this review, we discuss recent advances in molecular tools used to study this interplay, with a focus on those that explore how the microbiome regulates host gene expression. These tools include CRISPR-based whole-body genetic tools for deciphering host-specific genes involved in the interaction process, Cre-loxP based tissue/cell-specific gene editing approaches, and in vitro models of host-derived organoids. Overall, the application of these molecular tools is revolutionizing our understanding of how host-microbiome interactions contribute to health and disease, paving the way for improved therapies and interventions that target microbial influences on the host.
Collapse
Affiliation(s)
- Siyao Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, China
| | - Xu Gong
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, China
| | - Fei Xiao
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Yun Yang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, China
| |
Collapse
|
11
|
Zeng X, Li J, Wang X, Liu L, Shen S, Li N, Wang Z, Yuan Y, Yue T. Regulation of Gut Microbiota and Microbial Metabolome of Kefir Supernatant against Fusobacterium nucleatum and DSS-Coinduced Colitis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:3536-3548. [PMID: 38346349 DOI: 10.1021/acs.jafc.3c08050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
The aim of this study was to investigate the intervention effect of kefir supernatant (KS) on the initiation and progression of an ulcerative colitis (UC) murine model. We established an UC murine model by orally administrating with 109 CFUs of Fusobacterium nucleatum for 3 weeks and 3% dextran sulfate sodium (DSS) treatment in the third week. KS was used to intervene in this colitis model. Our results showed that KS supplementation ameliorated the symptoms, restrained the secretion of pro-inflammatory cytokines (TNF-α, IL-6, and IL-17F), promoted the release of anti-inflammatory cytokines (IL-4 and IL-10), and ameliorated oxidative stress. Furthermore, the increased number of goblet cells and upregulated expression of MUC2, occludin and claudin-1 indicated that the colon barrier was protected by KS. Additionally, KS supplementation mitigated gut microbiota dysbiosis in the UC murine model, leading to an increase in the abundance of Blautia and Akkermansia and a decrease in the level of Bacteroides. The altered gut microbiota also affected colon metabolism, with differential metabolites mainly associated with the biosynthesis of the l-arginine pathway. This study revealed that KS supplementation restored the community structure of gut microbiota, altered the biosynthesis of l-arginine, and thereby modulated the process of colonic inflammation.
Collapse
Affiliation(s)
- Xuejun Zeng
- College of Food Science and Technology, Northwest University, Xi'an 710069, China
- College of Food Science and Engineering, Northwest A & F University, Yangling, Xianyang 712100, China
| | - Jiahui Li
- College of Food Science and Engineering, Northwest A & F University, Yangling, Xianyang 712100, China
| | - Xin Wang
- College of Health Management, Shangluo University, Shangluo 726000, China
| | - Ling Liu
- College of Food Science and Engineering, Northwest A & F University, Yangling, Xianyang 712100, China
| | - Shiqi Shen
- College of Food Science and Engineering, Northwest A & F University, Yangling, Xianyang 712100, China
| | - Nanyang Li
- College of Food Science and Engineering, Northwest A & F University, Yangling, Xianyang 712100, China
| | - Zhouli Wang
- College of Food Science and Engineering, Northwest A & F University, Yangling, Xianyang 712100, China
| | - Yahong Yuan
- College of Food Science and Technology, Northwest University, Xi'an 710069, China
- College of Food Science and Engineering, Northwest A & F University, Yangling, Xianyang 712100, China
| | - Tianli Yue
- College of Food Science and Technology, Northwest University, Xi'an 710069, China
- College of Food Science and Engineering, Northwest A & F University, Yangling, Xianyang 712100, China
| |
Collapse
|
12
|
Chen YY, Fei F, Ding LL, Wen SY, Ren CF, Gong AH. Integrated gut microbiome and metabolome analysis reveals the inhibition effect of Lactobacillus plantarum CBT against colorectal cancer. Food Funct 2024; 15:853-865. [PMID: 38164977 DOI: 10.1039/d3fo04806c] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
The microecological stability of the gut microbiota plays a pivotal role in both preventing and treating colorectal cancer (CRC). This study investigated whether Lactobacillus plantarum CBT (LP-CBT) prevents CRC by inducing alterations in the gut microbiota composition and associated metabolites. The results showed that LP-CBT inhibited colorectal tumorigenesis in azoxymethane/dextran sulfate sodium (AOM/DSS)-treated mice by repairing the intestinal barrier function. Furthermore, LP-CBT decreased pro-inflammatory cytokines and anti-inflammatory cytokines. Importantly, LP-CBT remodeled intestinal homeostasis by increasing probiotics (Coprococcus, Mucispirillum, and Lactobacillus) and reducing harmful bacteria (Dorea, Shigella, Alistipes, Paraprevotella, Bacteroides, Sutterella, Turicibacter, Bifidobacterium, Clostridium, Allobaculum), significantly influencing arginine biosynthesis. Therefore, LP-CBT treatment regulated invertases and metabolites associated with the arginine pathway (carbamoyl phosphate, carboxymethyl proline, L-lysine, 10,11-epoxy-3-geranylgeranylindole, n-(6)-[(indol-3-yl)acetyl]-L-lysine, citrulline, N2-succinyl-L-ornithine, and (5-L-glutamyl)-L-glutamate). Furthermore, the inhibitory effect of LP-CBT on colorectal cancer was further confirmed using the MC38 subcutaneous tumor model. Collectively, these findings offer compelling evidence supporting the potential of LP-CBT as a viable preventive strategy against CRC.
Collapse
Affiliation(s)
- Yan-Yan Chen
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212003, China.
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, SAR 999078, China
- Hematological Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212003, China
| | - Fei Fei
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212003, China.
| | - Ling-Ling Ding
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212003, China.
| | - Shi-Yuan Wen
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030000, China.
| | - Cai-Fang Ren
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212003, China.
| | - Ai-Hua Gong
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212003, China.
- Hematological Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212003, China
| |
Collapse
|
13
|
Cao S, Li Y, Song R, Meng X, Fuchs M, Liang C, Kachler K, Meng X, Wen J, Schlötzer-Schrehardt U, Taudte V, Gessner A, Kunz M, Schleicher U, Zaiss MM, Kastbom A, Chen X, Schett G, Bozec A. L-arginine metabolism inhibits arthritis and inflammatory bone loss. Ann Rheum Dis 2024; 83:72-87. [PMID: 37775153 PMCID: PMC10803985 DOI: 10.1136/ard-2022-223626] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 08/29/2023] [Indexed: 10/01/2023]
Abstract
OBJECTIVES To investigate the effect of the L-arginine metabolism on arthritis and inflammation-mediated bone loss. METHODS L-arginine was applied to three arthritis models (collagen-induced arthritis, serum-induced arthritis and human TNF transgenic mice). Inflammation was assessed clinically and histologically, while bone changes were quantified by μCT and histomorphometry. In vitro, effects of L-arginine on osteoclast differentiation were analysed by RNA-seq and mass spectrometry (MS). Seahorse, Single Cell ENergetIc metabolism by profilIng Translation inHibition and transmission electron microscopy were used for detecting metabolic changes in osteoclasts. Moreover, arginine-associated metabolites were measured in the serum of rheumatoid arthritis (RA) and pre-RA patients. RESULTS L-arginine inhibited arthritis and bone loss in all three models and directly blocked TNFα-induced murine and human osteoclastogenesis. RNA-seq and MS analyses indicated that L-arginine switched glycolysis to oxidative phosphorylation in inflammatory osteoclasts leading to increased ATP production, purine metabolism and elevated inosine and hypoxanthine levels. Adenosine deaminase inhibitors blocking inosine and hypoxanthine production abolished the inhibition of L-arginine on osteoclastogenesis in vitro and in vivo. Altered arginine levels were also found in RA and pre-RA patients. CONCLUSION Our study demonstrated that L-arginine ameliorates arthritis and bone erosion through metabolic reprogramming and perturbation of purine metabolism in osteoclasts.
Collapse
Affiliation(s)
- Shan Cao
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Shanghai, Germany
- Department of Rheumatology, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yixuan Li
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Shanghai, Germany
- Department of Rheumatology, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Song
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Shanghai, Germany
- Department of Rheumatology, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xianyi Meng
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Shanghai, Germany
| | - Maximilian Fuchs
- Chair of Medical Informatics, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Chunguang Liang
- Chair of Medical Informatics, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Bioinformatics, Biocenter, University of Würzburg Am Hubland, Würzburg, Germany
| | - Katerina Kachler
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Shanghai, Germany
| | - Xinyu Meng
- Department of Rheumatology, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinming Wen
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Shanghai, Germany
- Cancer Center, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Ursula Schlötzer-Schrehardt
- Department of Ophthalmology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Verena Taudte
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Core Facility for Metabolomics, Department of Medicine, Philipps University of Marburg, Marburg, Germany
| | - Arne Gessner
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Meik Kunz
- Chair of Medical Informatics, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Ulrike Schleicher
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Mario M Zaiss
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Shanghai, Germany
| | - Alf Kastbom
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Xiaoxiang Chen
- Department of Rheumatology, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Shanghai, Germany
| | - Aline Bozec
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Shanghai, Germany
| |
Collapse
|
14
|
Pu D, Yao Y, Zhou C, Liu R, Wang Z, Liu Y, Wang D, Wang B, Wang Y, Liu Z, Zhang Z, Feng B. FMT rescues mice from DSS-induced colitis in a STING-dependent manner. Gut Microbes 2024; 16:2397879. [PMID: 39324491 PMCID: PMC11441074 DOI: 10.1080/19490976.2024.2397879] [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: 07/17/2023] [Revised: 07/28/2024] [Accepted: 08/23/2024] [Indexed: 09/27/2024] Open
Abstract
Fecal microbiota transplantation (FMT) is currently a promising therapy for inflammatory bowel disease (IBD). However, clinical studies have shown that there is an obvious individual difference in the efficacy of FMT. Therefore, it is a pressing issue to identify the factors that influence the efficacy of FMT and find ways to screen the most suitable patients for this therapy. In this work, we targeted the stimulator of interferon genes (STING), a DNA-sensing protein that regulates host-defense. By comparing the differential efficacy of FMT in mice with different expression level of STING, it is revealed that FMT therapy provides treatment for DSS-induced colitis in a STING-dependent manner. Mechanistically, FMT exerts a regulatory effect on the differentiation of intestinal Th17 cells and macrophages, splenic Th1 and Th2 cells, as well as Th1 cells of the mesenteric lymph nodes via STING, down-regulating the colonic M1/M2 and splenic Th1/Th2 cell ratios, thereby improving the imbalanced immune homeostasis in the inflamed intestine. Meanwhile, based on the 16SrDNA sequencing of mice fecal samples, STING was found to facilitate the donor strain colonization in recipients' gut, mainly Lactobacillales, thereby reshaping the gut microbiota disturbed by colitis. Consequently, we proposed that STING, as a key target of FMT therapy, is potentially a biomarker for screening the most suitable individuals for FMT to optimize treatment regimens and enhance clinical benefit.
Collapse
Affiliation(s)
- Dan Pu
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yao Yao
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chuan Zhou
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruixian Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhihong Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dandan Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Binbin Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yaohe Wang
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Zhanju Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, the Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Zhe Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Baisui Feng
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
15
|
Ning L, Zhou YL, Sun H, Zhang Y, Shen C, Wang Z, Xuan B, Zhao Y, Ma Y, Yan Y, Tong T, Huang X, Hu M, Zhu X, Ding J, Zhang Y, Cui Z, Fang JY, Chen H, Hong J. Microbiome and metabolome features in inflammatory bowel disease via multi-omics integration analyses across cohorts. Nat Commun 2023; 14:7135. [PMID: 37932270 PMCID: PMC10628233 DOI: 10.1038/s41467-023-42788-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 10/20/2023] [Indexed: 11/08/2023] Open
Abstract
The perturbations of the gut microbiota and metabolites are closely associated with the progression of inflammatory bowel disease (IBD). However, inconsistent findings across studies impede a comprehensive understanding of their roles in IBD and their potential as reliable diagnostic biomarkers. To address this challenge, here we comprehensively analyze 9 metagenomic and 4 metabolomics cohorts of IBD from different populations. Through cross-cohort integrative analysis (CCIA), we identify a consistent characteristic of commensal gut microbiota. Especially, three bacteria, namely Asaccharobacter celatus, Gemmiger formicilis, and Erysipelatoclostridium ramosum, which are rarely reported in IBD. Metagenomic functional analysis reveals that essential gene of Two-component system pathway, linked to fecal calprotectin, are implicated in IBD. Metabolomics analysis shows 36 identified metabolites with significant differences, while the roles of these metabolites in IBD are still unknown. To further elucidate the relationship between gut microbiota and metabolites, we construct multi-omics biological correlation (MOBC) maps, which highlights gut microbial biotransformation deficiencies and significant alterations in aminoacyl-tRNA synthetases. Finally, we identify multi-omics biomarkers for IBD diagnosis, validated across multiple global cohorts (AUROC values ranging from 0.92 to 0.98). Our results offer valuable insights and a significant resource for developing mechanistic hypotheses on host-microbiome interactions in IBD.
Collapse
Affiliation(s)
- Lijun Ning
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Yi-Lu Zhou
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Han Sun
- Department of Gastroenterology, Xuzhou Central Hospital, Clinical School of Xuzhou Medical University, Xuzhou, China
| | - Youwei Zhang
- Department of Medical Oncology, Xuzhou Central Hospital, Clinical School of Xuzhou Medical University, Xuzhou, China
| | - Chaoqin Shen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Zhenhua Wang
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Baoqin Xuan
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Ying Zhao
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Yanru Ma
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Yuqing Yan
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Tianying Tong
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Xiaowen Huang
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Muni Hu
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Xiaoqiang Zhu
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Jinmei Ding
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Yue Zhang
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Zhe Cui
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Jing-Yuan Fang
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Haoyan Chen
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine. 145 Middle Shandong Road, Shanghai, 200001, China.
| | - Jie Hong
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine. 145 Middle Shandong Road, Shanghai, 200001, China.
| |
Collapse
|
16
|
Zhang Q, Song Q, Liu S, Xu Y, Gao D, Lu P, Liu Y, Zhao G, Wu L, Zhao C, Yang J. Integrated transcriptomic and metabolomic analysis reveals the metabolic programming of GM-CSF- and M-CSF- differentiated mouse macrophages. Front Immunol 2023; 14:1230772. [PMID: 37818352 PMCID: PMC10560851 DOI: 10.3389/fimmu.2023.1230772] [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: 05/29/2023] [Accepted: 09/11/2023] [Indexed: 10/12/2023] Open
Abstract
Macrophages play a critical role in the inflammatory response and tumor development. Macrophages are primarily divided into pro-inflammatory M1-like and anti-inflammatory M2-like macrophages based on their activation status and functions. In vitro macrophage models could be derived from mouse bone marrow cells stimulated with two types of differentiation factors: GM-CSF (GM-BMDMs) and M-CSF (M-BMDMs), to represent M1- and M2-like macrophages, respectively. Since macrophage differentiation requires coordinated metabolic reprogramming and transcriptional rewiring in order to fulfill their distinct roles, we combined both transcriptome and metabolome analysis, coupled with experimental validation, to gain insight into the metabolic status of GM- and M-BMDMs. The data revealed higher levels of the tricarboxylic acid cycle (TCA cycle), oxidative phosphorylation (OXPHOS), fatty acid oxidation (FAO), and urea and ornithine production from arginine in GM-BMDMs, and a preference for glycolysis, fatty acid storage, bile acid metabolism, and citrulline and nitric oxide (NO) production from arginine in M-BMDMs. Correlation analysis with the proteomic data showed high consistency in the mRNA and protein levels of metabolic genes. Similar results were also obtained when compared to RNA-seq data of human monocyte derived macrophages from the GEO database. Furthermore, canonical macrophage functions such as inflammatory response and phagocytosis were tightly associated with the representative metabolic pathways. In the current study, we identified the core metabolites, metabolic genes, and functional terms of the two distinct mouse macrophage populations. We also distinguished the metabolic influences of the differentiation factors GM-CSF and M-CSF, and wish to provide valuable information for in vitro macrophage studies.
Collapse
Affiliation(s)
- Qianyue Zhang
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Qiaoling Song
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Innovation Platform of Marine Drug Screening and Evaluation, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Shan Liu
- Innovation Platform of Marine Drug Screening and Evaluation, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Yuting Xu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Danling Gao
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Peizhe Lu
- Department of Neuroscience, University of Michigan, Ann Arbor, MI, United States
| | - Yuantao Liu
- Department of Endocrinology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Guanghui Zhao
- Medical Laboratory Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Oncology Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Lihong Wu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Chenyang Zhao
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Jinbo Yang
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Innovation Platform of Marine Drug Screening and Evaluation, Qingdao Marine Science and Technology Center, Qingdao, China
| |
Collapse
|
17
|
Yu X, Mao C, Zong S, Khan A, Wang W, Yun H, Zhang P, Shigaki T, Fang Y, Han H, Li X. Transcriptome analysis reveals self-redox mineralization mechanism of azo dyes and novel decolorizing hydrolases in Aspergillus tabacinus LZ-M. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 325:121459. [PMID: 36934962 DOI: 10.1016/j.envpol.2023.121459] [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: 01/04/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 06/18/2023]
Abstract
Bio-degradation is the most affordable method of azo dye decontamination, while its drawbacks such as aromatic amines accumulation and low degradation efficiency must be overcome. In this study, a novel mechanism of azo dye degradation by a fungus was discovered. At a concentration of 400 mg/L, the decolorization efficiency of Acid Red 73 (AR73) by Aspergillus tabacinus LZ-M was 90.28%. Metabolite analysis and transcriptome sequencing analysis revealed a self-redox process of AR73 degradation, where the electrons generated in carbon oxidation were transferred to the reduction of -C-N = and -NN. The metabolites, 2-hydroxynaphthalene and N-phenylnitrous amide were mineralized into CO2 through catechol pathway and a glycolytic process. Furthermore, the mineralization ratio of dye was computed to be 31.8% by the carbon balance and electron balance. By using comparative transcriptome, a novel decoloring enzyme Ord95 was discovered in unknown genes through gene cloning. It hydrolyzed AR73 into 2-hydroxynaphthalene and N-phenylnitrous amide, containing a glutathione S-transferase domain with three arginines as key active sites. Here the new mechanism of azo dye degradation was discovered with identification of a novel enzyme in Aspergillus tabacinus LZ-M.
Collapse
Affiliation(s)
- Xuan Yu
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Tianshui South Road #222, Lanzhou, 730000, Gansu, China
| | - Chunlan Mao
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Tianshui South Road #222, Lanzhou, 730000, Gansu, China
| | - Simin Zong
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Tianshui South Road #222, Lanzhou, 730000, Gansu, China
| | - Aman Khan
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Tianshui South Road #222, Lanzhou, 730000, Gansu, China
| | - Wenxue Wang
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Tianshui South Road #222, Lanzhou, 730000, Gansu, China
| | - Hui Yun
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Tianshui South Road #222, Lanzhou, 730000, Gansu, China
| | - Peng Zhang
- Key Laboratory for Resources Utilization Technoloy of Unconventional Water of Gansu Province, Gansu Academy of Membrane Science and Technology, Duanjiatanlu #1272, Lanzhou, 730020, Gansu, China
| | - Toshiro Shigaki
- Laboratory of Plant Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Yitian Fang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Huawen Han
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Tianshui South Road #222, Lanzhou, 730000, Gansu, China
| | - Xiangkai Li
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Tianshui South Road #222, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
18
|
Monaghan KL, Zheng W, Akhter H, Wang L, Ammer AG, Li P, Lin JX, Hu G, Leonard WJ, Wan ECK. Tetramerization of STAT5 regulates monocyte differentiation and the dextran sulfate sodium-induced colitis in mice. Front Immunol 2023; 14:1117828. [PMID: 37153611 PMCID: PMC10157487 DOI: 10.3389/fimmu.2023.1117828] [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: 12/06/2022] [Accepted: 04/11/2023] [Indexed: 05/09/2023] Open
Abstract
In response to external stimuli during immune responses, monocytes can have multifaceted roles such as pathogen clearance and tissue repair. However, aberrant control of monocyte activation can result in chronic inflammation and subsequent tissue damage. Granulocyte-macrophage colony-stimulating factor (GM-CSF) induces monocyte differentiation into a heterogenous population of monocyte-derived dendritic cells (moDCs) and macrophages. However, the downstream molecular signals that dictate the differentiation of monocytes under pathological conditions is incompletely understood. We report here that the GM-CSF-induced STAT5 tetramerization is a critical determinate of monocyte fate and function. Monocytes require STAT5 tetramers to differentiate into moDCs. Conversely, the absence of STAT5 tetramers results in a switch to a functionally distinct monocyte-derived macrophage population. In the dextran sulfate sodium (DSS) model of colitis, STAT5 tetramer-deficient monocytes exacerbate disease severity. Mechanistically, GM-CSF signaling in STAT5 tetramer-deficient monocytes results in the overexpression of arginase I and a reduction in nitric oxide synthesis following stimulation with lipopolysaccharide. Correspondingly, the inhibition of arginase I activity and sustained supplementation of nitric oxide ameliorates the worsened colitis in STAT5 tetramer-deficient mice. This study suggests that STAT5 tetramers protect against severe intestinal inflammation through the regulation of arginine metabolism.
Collapse
Affiliation(s)
- Kelly L. Monaghan
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
| | - Wen Zheng
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
| | - Halima Akhter
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Department of Computer Science and Electrical Engineering, West Virginia University, Morgantown, WV, United States
| | - Lei Wang
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
| | - Amanda G. Ammer
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Microscope Imaging Facility, West Virginia University, Morgantown, WV, United States
| | - Peng Li
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jian-Xin Lin
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Gangqing Hu
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Bioinformatics Core, West Virginia University, Morgantown, WV, United States
| | - Warren J. Leonard
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Edwin C. K. Wan
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Department of Neuroscience, West Virginia University, Morgantown, WV, United States
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
19
|
Grüner N, Ortlepp AL, Mattner J. Pivotal Role of Intestinal Microbiota and Intraluminal Metabolites for the Maintenance of Gut-Bone Physiology. Int J Mol Sci 2023; 24:ijms24065161. [PMID: 36982235 PMCID: PMC10048911 DOI: 10.3390/ijms24065161] [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: 01/29/2023] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/30/2023] Open
Abstract
Intestinal microbiota, and their mutual interactions with host tissues, are pivotal for the maintenance of organ physiology. Indeed, intraluminal signals influence adjacent and even distal tissues. Consequently, disruptions in the composition or functions of microbiota and subsequent altered host-microbiota interactions disturb the homeostasis of multiple organ systems, including the bone. Thus, gut microbiota can influence bone mass and physiology, as well as postnatal skeletal evolution. Alterations in nutrient or electrolyte absorption, metabolism, or immune functions, due to the translocation of microbial antigens or metabolites across intestinal barriers, affect bone tissues, as well. Intestinal microbiota can directly and indirectly alter bone density and bone remodeling. Intestinal dysbiosis and a subsequently disturbed gut-bone axis are characteristic for patients with inflammatory bowel disease (IBD) who suffer from various intestinal symptoms and multiple bone-related complications, such as arthritis or osteoporosis. Immune cells affecting the joints are presumably even primed in the gut. Furthermore, intestinal dysbiosis impairs hormone metabolism and electrolyte balance. On the other hand, less is known about the impact of bone metabolism on gut physiology. In this review, we summarized current knowledge of gut microbiota, metabolites and microbiota-primed immune cells in IBD and bone-related complications.
Collapse
Affiliation(s)
- Niklas Grüner
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Anna Lisa Ortlepp
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jochen Mattner
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Medical Immunology Campus Erlangen, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
20
|
Ji Z, Ma L. Controlling taxa abundance improves metatranscriptomics differential analysis. BMC Microbiol 2023; 23:60. [PMID: 36882742 PMCID: PMC9990291 DOI: 10.1186/s12866-023-02799-9] [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: 05/09/2022] [Accepted: 02/17/2023] [Indexed: 03/09/2023] Open
Abstract
BACKGROUND A common task in analyzing metatranscriptomics data is to identify microbial metabolic pathways with differential RNA abundances across multiple sample groups. With information from paired metagenomics data, some differential methods control for either DNA or taxa abundances to address their strong correlation with RNA abundance. However, it remains unknown if both factors need to be controlled for simultaneously. RESULTS We discovered that when either DNA or taxa abundance is controlled for, RNA abundance still has a strong partial correlation with the other factor. In both simulation studies and a real data analysis, we demonstrated that controlling for both DNA and taxa abundances leads to superior performance compared to only controlling for one factor. CONCLUSIONS To fully address the confounding effects in analyzing metatranscriptomics data, both DNA and taxa abundances need to be controlled for in the differential analysis.
Collapse
Affiliation(s)
- Zhicheng Ji
- Department of Biostatistics and Bioinformatics, Duke University, Durham, USA.
| | - Li Ma
- Department of Biostatistics and Bioinformatics, Duke University, Durham, USA. .,Department of Statistical Science, Duke University, Durham, USA.
| |
Collapse
|
21
|
Brugaletta G, Zampiga M, Laghi L, Indio V, Oliveri C, De Cesare A, Sirri F. Feeding broiler chickens with arginine above recommended levels: effects on growth performance, metabolism, and intestinal microbiota. J Anim Sci Biotechnol 2023; 14:33. [PMID: 36864475 PMCID: PMC9983211 DOI: 10.1186/s40104-023-00839-y] [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: 09/30/2022] [Accepted: 01/10/2023] [Indexed: 03/04/2023] Open
Abstract
BACKGROUND Arginine is an essential amino acid for chickens and feeding diets with arginine beyond the recommended levels has been shown to influence the growth performance of broiler chickens in a positive way. Nonetheless, further research is required to understand how arginine supplementation above the widely adopted dosages affects metabolism and intestinal health of broilers. Therefore, this study was designed to assess the effects of arginine supplementation (i.e., total arginine to total lysine ratio of 1.20 instead of 1.06-1.08 recommended by the breeding company) on growth performance of broiler chickens and to explore its impacts on the hepatic and blood metabolic profiles, as well as on the intestinal microbiota. For this purpose, 630 one-day-old male Ross 308 broiler chicks were assigned to 2 treatments (7 replicates each) fed a control diet or a crystalline L-arginine-supplemented diet for 49 d. RESULTS Compared to control birds, those supplemented with arginine performed significantly better exhibiting greater final body weight at D49 (3778 vs. 3937 g; P < 0.001), higher growth rate (76.15 vs. 79.46 g of body weight gained daily; P < 0.001), and lower cumulative feed conversion ratio (1.808 vs. 1.732; P < 0.05). Plasma concentrations of arginine, betaine, histidine, and creatine were greater in supplemented birds than in their control counterparts, as were those of creatine, leucine and other essential amino acids at the hepatic level. In contrast, leucine concentration was lower in the caecal content of supplemented birds. Reduced alpha diversity and relative abundance of Firmicutes and Proteobacteria (specifically Escherichia coli), as well as increased abundance of Bacteroidetes and Lactobacillus salivarius were found in the caecal content of supplemented birds. CONCLUSIONS The improvement in growth performance corroborates the advantages of supplementing arginine in broiler nutrition. It can be hypothesized that the performance enhancement found in this study is associated with the increased availability of arginine, betaine, histidine, and creatine in plasma and the liver, as well as to the ability of extra dietary arginine to potentially ameliorate intestinal conditions and microbiota of supplemented birds. However, the latter promising property, along with other research questions raised by this study, deserve further investigations.
Collapse
Affiliation(s)
- Giorgio Brugaletta
- grid.6292.f0000 0004 1757 1758Department of Agricultural and Food Sciences, Alma Mater Studiorum – University of Bologna, Ozzano Dell’Emilia, 40064 Bologna, Italy
| | - Marco Zampiga
- grid.6292.f0000 0004 1757 1758Department of Agricultural and Food Sciences, Alma Mater Studiorum – University of Bologna, Ozzano Dell’Emilia, 40064 Bologna, Italy
| | - Luca Laghi
- grid.6292.f0000 0004 1757 1758Department of Agricultural and Food Sciences, Alma Mater Studiorum – University of Bologna, Ozzano Dell’Emilia, 40064 Bologna, Italy
| | - Valentina Indio
- grid.6292.f0000 0004 1757 1758Department of Veterinary Medical Sciences, Alma Mater Studiorum – University of Bologna, Ozzano Dell’Emilia, 40064 Bologna, Italy
| | - Chiara Oliveri
- grid.6292.f0000 0004 1757 1758Department of Physics and Astronomy, Alma Mater Studiorum – University of Bologna, 40127 Bologna, Italy
| | - Alessandra De Cesare
- grid.6292.f0000 0004 1757 1758Department of Veterinary Medical Sciences, Alma Mater Studiorum – University of Bologna, Ozzano Dell’Emilia, 40064 Bologna, Italy
| | - Federico Sirri
- Department of Agricultural and Food Sciences, Alma Mater Studiorum - University of Bologna, Ozzano Dell'Emilia, 40064, Bologna, Italy.
| |
Collapse
|
22
|
Du C, Quan S, Zhao Y, Nan X, Chen R, Tang X, Xiong B. Bovine milk-derived extracellular vesicles prevent gut inflammation by regulating lipid and amino acid metabolism. Food Funct 2023; 14:2212-2222. [PMID: 36757176 DOI: 10.1039/d2fo03975c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Inflammatory bowel disease (IBD) is a global health problem in which metabolite alteration plays an important pathogenic role. Bovine milk-derived extracellular vesicles (mEVs) have been shown to regulate nutrient metabolism in healthy animal models. This study investigated the effect of oral mEVs on metabolite changes in DSS-induced murine colitis. We performed metabolomic profiling on plasma samples and measured the concentrations of lipids and amino acids in both fecal samples and colonic tissues. Plasma metabolome analysis found that mEVs significantly upregulated 148 metabolite levels and downregulated 44 metabolite concentrations (VIP > 1, and p < 0.05). In the fecal samples, mEVs significantly increased the contents of acetate and butyrate and decreased the levels of tridecanoic acid (C13:0), methyl cis-10-pentadecenoate (C15:1) and cis-11-eicosenoic acid (C20:1). Moreover, the concentrations of eicosadienoic acid (C20:2), eicosapentaenoic acid (C20:5), and docosahexaenoic acid (C22:6) were decreased in colonic tissues with mEV supplementation. In addition, compared with the DSS group, mEVs significantly increased the content of L-arginine, decreased the level of L-valine in the fecal samples, and also decreased the levels of L-serine and L-glutamate in the colonic tissues. Collectively, our findings demonstrated that mEVs could recover the metabolic abnormalities caused by inflammation and provided novel insights into mEVs as a potential modulator for metabolites to prevent and treat IBD.
Collapse
Affiliation(s)
- Chunmei Du
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Suyu Quan
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Yiguang Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Xuemei Nan
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Ruipeng Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Xiangfang Tang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Benhai Xiong
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
23
|
Role of the Intestinal Microbiota in the Genesis of Major Depression and the Response to Antidepressant Drug Therapy: A Narrative Review. Biomedicines 2023; 11:biomedicines11020550. [PMID: 36831086 PMCID: PMC9953611 DOI: 10.3390/biomedicines11020550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
A major depressive disorder is a serious mental illness characterized by a pervasive low mood that negatively concerns personal life, work life, or education, affecting millions of people worldwide. To date, due to the complexity of the disease, the most common and effective treatments consist of a multi-therapy approach, including psychological, social, and pharmacological support with antidepressant drugs. In general, antidepressants are effective in correcting chemical imbalances of neurotransmitters in the brain, but recent evidence has underlined the pivotal role of gut microbiota (GM) also in the regulation of their pharmacokinetics/pharmacodynamics, through indirect or direct mechanisms. The study of these complex interactions between GM and drugs is currently under the spotlight, and it has been recently named "pharmacomicrobiomics". Hence, the purpose of this review is to summarize the contribution of GM and its metabolites in depression, as well as their role in the metabolism and activity of antidepressant drugs, in order to pave the way for the personalized administration of antidepressant therapies.
Collapse
|
24
|
Yoon JH, Do JS, Velankanni P, Lee CG, Kwon HK. Gut Microbial Metabolites on Host Immune Responses in Health and Disease. Immune Netw 2023; 23:e6. [PMID: 36911800 PMCID: PMC9995988 DOI: 10.4110/in.2023.23.e6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/12/2023] [Accepted: 02/13/2023] [Indexed: 03/07/2023] Open
Abstract
Intestinal microorganisms interact with various immune cells and are involved in gut homeostasis and immune regulation. Although many studies have discussed the roles of the microorganisms themselves, interest in the effector function of their metabolites is increasing. The metabolic processes of these molecules provide important clues to the existence and function of gut microbes. The interrelationship between metabolites and T lymphocytes in particular plays a significant role in adaptive immune functions. Our current review focuses on 3 groups of metabolites: short-chain fatty acids, bile acids metabolites, and polyamines. We collated the findings of several studies on the transformation and production of these metabolites by gut microbes and explained their immunological roles. Specifically, we summarized the reports on changes in mucosal immune homeostasis represented by the Tregs and Th17 cells balance. The relationship between specific metabolites and diseases was also analyzed through latest studies. Thus, this review highlights microbial metabolites as the hidden treasure having potential diagnostic markers and therapeutic targets through a comprehensive understanding of the gut-immune interaction.
Collapse
Affiliation(s)
- Jong-Hwi Yoon
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Jun-Soo Do
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Priyanka Velankanni
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Korea
| | - Choong-Gu Lee
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Korea
- Division of Bio-Medical Science and Technology, Korea Institute of Science and Technology (KIST) School, University of Science and Technology, Seoul 02792, Korea
| | - Ho-Keun Kwon
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Korea
- Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
25
|
Brandt A, Baumann A, Hernández-Arriaga A, Jung F, Nier A, Staltner R, Rajcic D, Schmeer C, Witte OW, Wessner B, Franzke B, Wagner KH, Camarinha-Silva A, Bergheim I. Impairments of intestinal arginine and NO metabolisms trigger aging-associated intestinal barrier dysfunction and 'inflammaging'. Redox Biol 2022; 58:102528. [PMID: 36356464 PMCID: PMC9649383 DOI: 10.1016/j.redox.2022.102528] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/28/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Aging is considered a state of low grade inflammation, occurring in the absence of any overt infection often referred to as 'inflammaging'. Maintaining intestinal homeostasis may be a target to extend a healthier status in older adults. Here, we report that even in healthy older men low grade bacterial endotoxemia is prevalent. In addition, employing multiple mouse models, we also show that while intestinal microbiota composition changes significantly during aging, fecal microbiota transplantation to old mice does not protect against aging-associated intestinal barrier dysfunction in small intestine. Rather, intestinal NO homeostasis and arginine metabolism mediated through arginase and NO synthesis is altered in small intestine of aging mice. Treatment with the arginase inhibitor norNOHA prevented aging-associated intestinal barrier dysfunction, low grade endotoxemia and delayed the onset of senescence in peripheral tissue e.g., liver. Intestinal arginine and NO metabolisms could be a target in the prevention of aging-associated intestinal barrier dysfunction and subsequently decline and 'inflammaging'.
Collapse
Affiliation(s)
- Annette Brandt
- Department of Nutritional Sciences, R.F. Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Anja Baumann
- Department of Nutritional Sciences, R.F. Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | | | - Finn Jung
- Department of Nutritional Sciences, R.F. Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Anika Nier
- Department of Nutritional Sciences, R.F. Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Raphaela Staltner
- Department of Nutritional Sciences, R.F. Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Dragana Rajcic
- Department of Nutritional Sciences, R.F. Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Christian Schmeer
- Hans-Berger Department of Neurology, University Hospital Jena, Jena, Germany
| | - Otto W Witte
- Hans-Berger Department of Neurology, University Hospital Jena, Jena, Germany
| | - Barbara Wessner
- Centre for Sport Science and University Sports, University of Vienna, Vienna, Austria
| | - Bernhard Franzke
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | - Karl-Heinz Wagner
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | | | - Ina Bergheim
- Department of Nutritional Sciences, R.F. Molecular Nutritional Science, University of Vienna, Vienna, Austria.
| |
Collapse
|
26
|
Adolph TE, Meyer M, Schwärzler J, Mayr L, Grabherr F, Tilg H. The metabolic nature of inflammatory bowel diseases. Nat Rev Gastroenterol Hepatol 2022; 19:753-767. [PMID: 35906289 DOI: 10.1038/s41575-022-00658-y] [Citation(s) in RCA: 118] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/23/2022] [Indexed: 02/06/2023]
Abstract
Crohn's disease and ulcerative colitis, phenotypically comprising a spectrum of inflammatory bowel diseases (IBDs), spread globally during the westernization of lifestyle and dietary habits over the past few decades. Here, we review experimental and clinical evidence for the metabolic nature of gut inflammation in IBD and delineate distinct parallels to the inflammatory state in metabolic diseases. Experimental evidence indicates that excessive intake of specific macronutrients in a Western diet fuels an inflammatory response in the gut by exploiting sensors of innate immunity and perturbation of gut microbial metabolism. Genetic IBD risk partly affects metabolism and stress signalling of innate immunity, and immunometabolism controls susceptibility to gut inflammation. Epidemiological and clinical studies indicate that specific nutrients in the Western diet pose a risk for the development of IBD and a poor disease course. Translational studies in IBD indicate perturbation of energy metabolism in immune cells and perturbation of gut microbial metabolism, which can be shaped by diet. In turn, dietary restriction by exclusive enteral nutrition induces remission in patients with IBD. Collectively, these studies support a metabolic underpinning of gut inflammation in IBD as described for metabolic inflammation in obesity and related disorders.
Collapse
Affiliation(s)
- Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Moritz Meyer
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Julian Schwärzler
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Lisa Mayr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Felix Grabherr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
27
|
Yu QQ, Zhang H, Zhao S, Xie D, Zhao H, Chen W, Pang M, Han B, Jiang P. Systematic evaluation of irinotecan-induced intestinal mucositis based on metabolomics analysis. Front Pharmacol 2022; 13:958882. [PMID: 36188576 PMCID: PMC9520243 DOI: 10.3389/fphar.2022.958882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Chemotherapy-induced intestinal mucositis (CIM) is a major dose-limiting side effect of chemotherapy, especially in regimens containing irinotecan (CPT-11). Several studies on the pathologic mechanisms of CIM focused on both the genomics and molecular pathways triggered by chemotherapy. However, systematic evaluation of metabolomic analysis in irinotecan-induced intestinal mucositis (IIM) has not been investigated. This study aimed to comprehensively analyze metabolite changes in main tissues of IIM mouse models. Male ICR mice were assigned to two groups: the model group (n = 11) treated with CPT-11 (20 mg/kg daily; i.p.) and the control group (n= 11) with solvent for 9 days. Gas chromatography-mass spectrometry (GC-MS) was used to investigate the metabolic alterations in the serum, intestinal, colonic, hepatic, and splenic samples of mice between two groups by multivariate statistical analyses, including GC–MS data processing, pattern recognition analysis, and pathway analysis. Forty-six metabolites, including hydrocarbons, amino acids, lipids, benzenoids, hydroxy acids, and amines, had significant changes in levels in tissues and sera of IIM mouse models. The most important pathways related to the identified metabolites were the glycerolipid metabolism in the colon and aminoacyl-tRNA biosynthesis; glycine, serine, and threonine metabolism; and glyoxylate and dicarboxylate metabolism in the liver. Our study firstly provided a comprehensive and systematic view of metabolic alterations of IIM using GC-MS analysis. The characterizations of metabolic changes could offer profound and theoretical insight into exploring new biomarkers for diagnosis and treatment of IIM.
Collapse
Affiliation(s)
- Qing-Qing Yu
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao, China
- Jining First People’s Hospital, Jining Medical College, Jining, China
| | - Heng Zhang
- Department of Laboratory, Shandong Daizhuang Hospital, Jining, China
| | - Shiyuan Zhao
- Jining First People’s Hospital, Jining Medical College, Jining, China
| | - Dadi Xie
- Department of Endocrine, Tengzhou Central People’s Hospital, Tengzhou, China
| | - Haibo Zhao
- Jining First People’s Hospital, Jining Medical College, Jining, China
| | - Weidong Chen
- Jining First People’s Hospital, Jining Medical College, Jining, China
| | - Min Pang
- MNR Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, China
| | - Baoqin Han
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
- *Correspondence: Baoqin Han, ; Pei Jiang,
| | - Pei Jiang
- Jining First People’s Hospital, Jining Medical College, Jining, China
- *Correspondence: Baoqin Han, ; Pei Jiang,
| |
Collapse
|
28
|
Li JY, Guo YC, Zhou HF, Yue TT, Wang FX, Sun F, Wang WZ. Arginine metabolism regulates the pathogenesis of inflammatory bowel disease. Nutr Rev 2022; 81:578-586. [PMID: 36040377 PMCID: PMC10086623 DOI: 10.1093/nutrit/nuac070] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Abstract
The pathogenesis of inflammatory bowel disease (IBD) is related to genetic susceptibility, enteric dysbiosis, and uncontrolled, chronic inflammatory responses that lead to colonic tissue damage and impaired intestinal absorption. As a consequence, patients with IBD are prone to nutrition deficits after each episode of disease resurgence. Nutritional supplementation, especially for protein components, is often implemented during the remission phase of IBD. Notably, ingested nutrients could affect the progression of IBD and the prognostic outcome of patients; therefore, they should be cautiously evaluated prior to being used for IBD intervention. Arginine (Arg) is a semi-essential amino acid required for protein synthesis and intimately associated with gut pathophysiology. To help optimize arginine-based nutritional intervention strategies, the present work summarizes that during the process of IBD, patients manifest colonic Arg deficiency and the turbulence of Arg metabolic pathways. The roles of Arg–nitric oxide (catalyzed by inducible nitric oxide synthase) and Arg–urea (catalyzed by arginases) pathways in IBD are debatable; the Arg–polyamine and Arg–creatine pathways are mainly protective. Overall, supplementation with Arg is a promising therapeutic strategy for IBD; however, the dosage of Arg may need to be carefully tailored for different individuals at different disease stages. Additionally, the combination of Arg supplementation with inhibitors of Arg metabolic pathways as well as other treatment options is worthy of further exploration.
Collapse
Affiliation(s)
- Jun-Yi Li
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital , Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan-Chao Guo
- Huazhong University of Science and Technology Department of Clinical Nutrition, Tongji Medical College, , Wuhan, China
| | - Hai-Feng Zhou
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital , Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tian-Tian Yue
- Huazhong University of Science and Technology Department of Clinical Nutrition, Tongji Medical College, , Wuhan, China
| | - Fa-Xi Wang
- Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology , Wuhan, China
| | - Fei Sun
- Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology , Wuhan, China
| | - Wen-Zhu Wang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital , Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
29
|
Tung TH, Chen YC, Lin YT, Huang SY. N-3 PUFA Ameliorates the Gut Microbiota, Bile Acid Profiles, and Neuropsychiatric Behaviours in a Rat Model of Geriatric Depression. Biomedicines 2022; 10:biomedicines10071594. [PMID: 35884899 PMCID: PMC9313093 DOI: 10.3390/biomedicines10071594] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/27/2022] [Accepted: 07/03/2022] [Indexed: 11/27/2022] Open
Abstract
The brain−gut−microbiome (BGM) axis affects host bioinformation. N-3 polyunsaturated fatty acids (PUFAs) alleviate cognitive impairment and depression in older adults. This study investigated altered microbiota−bile acid signalling as a potential mechanism linking fish oil-induced gut changes in microbiota to alleviate psychological symptoms. Sprague Dawley rats were fed a fish oil diet and administered D-galactose combined with chronic unpredictable mild stress (CUMS) to simulate geriatric depression. The cognitive function, psychological symptoms, microbiota compositions, and faecal bile acid profiles of the rats were assessed thereafter. A correlation analysis was conducted to determine whether the fish oil-induced alteration of the rats’ microbiota and bile acid profiles affected the rats’ behaviour. D-galactose and CUMS resulted in lower concentrations of Firmicutes, significantly altered bile acid profiles, and abnormal neurobehaviours. Fish oil intake alleviated the rats’ emotional symptoms and increased the abundance of Bacteroidetes, Prevotellaceae, Marinifilaceae, and Bacteroidesuniformis. It also elevated the concentrations of primary bile acids and taurine-conjugated bile acids in the rats’ faeces. The rats’ taurine-conjugated bile acid levels were significantly correlated with their behavioural outcomes. In short, fish oil intake may alleviate psychological symptoms by altering the microbial metabolites involved in the BGM axis, especially in the conjugation of bile acids.
Collapse
Affiliation(s)
- Te-Hsuan Tung
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110301, Taiwan; (T.-H.T.); (Y.-C.C.)
| | - Yang-Ching Chen
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110301, Taiwan; (T.-H.T.); (Y.-C.C.)
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 110301, Taiwan;
- Department of Family Medicine, Taipei Medical University Hospital, Taipei 110301, Taiwan
- Department of Family Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Ya-Tin Lin
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 110301, Taiwan;
| | - Shih-Yi Huang
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110301, Taiwan; (T.-H.T.); (Y.-C.C.)
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 110301, Taiwan;
- Nutrition Research Center, Taipei Medical University Hospital, Taipei 110301, Taiwan
- Correspondence: ; Tel.: +886-2-27361661 (ext. 6543); Fax: +886-2-27361661
| |
Collapse
|
30
|
Liu Y, Yang M, Tang L, Wang F, Huang S, Liu S, Lei Y, Wang S, Xie Z, Wang W, Zhao X, Tang B, Yang S. TLR4 regulates RORγt + regulatory T-cell responses and susceptibility to colon inflammation through interaction with Akkermansia muciniphila. MICROBIOME 2022; 10:98. [PMID: 35761415 PMCID: PMC9235089 DOI: 10.1186/s40168-022-01296-x] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/25/2022] [Indexed: 05/05/2023]
Abstract
BACKGROUND Well-balanced interactions between gut microbiota and the immune system are essential to prevent chronic intestinal inflammation, as observed in inflammatory bowel diseases (IBD). Toll-like receptor 4 (TLR4) functions as a sensor mediating the crosstalk between the intestinal commensal microbiome and host immunity, but the influence of TLR4 on the shaping of intestinal microbiota and immune responses during colon inflammation remains poorly characterized. We investigated whether the different susceptibilities to colitis between wild-type (WT) and TLR4-/- mice were gut microbiota-dependent and aimed to identify the potential immunity modulation mechanism. METHODS We performed antibiotic depletion of the microbiota, cohousing experiments, and faecal microbiota transplantation (FMT) in WT and TLR4-/- mice to assess the influence of TLR4 on intestinal microbial ecology. 16S rRNA sequencing was performed to dissect microbial discrepancies, and dysbiosis-associated immune perturbation was investigated by flow cytometry. Akkermansia muciniphila (A. muciniphila)-mediated immune modulation was confirmed through the T-cell transfer colitis model and bone marrow chimaera construction. RESULTS TLR4-/- mice experienced enhanced susceptibility to DSS-induced colitis. 16S rRNA sequencing showed notable discrepancy in the gut microbiota between WT and TLR4-/- mice. In particular, A. muciniphila contributed most to distinguishing the two groups. The T-cell transfer colitis model and bone marrow transplantation (BMT) consistently demonstrated that A. muciniphila ameliorated colitis by upregulating RORγt+ Treg cell-mediated immune responses. Mucosal biopsies from human manifested parallel outcomes with colon tissue from WT mice, as evidenced by the positive correlation between TLR4 expression and intestinal A. muciniphila colonization during homeostasis. CONCLUSIONS Our results demonstrate a novel protective role of TLR4 against intestinal inflammation, wherein it can modulate A. muciniphila-associated immune responses. These findings provide a new perspective on host-commensal symbiosis, which may be beneficial for developing potential therapeutic strategies. Video abstract.
Collapse
Affiliation(s)
- Yaojiang Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Third Military Medical University, 400037, Chongqing, China
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Min Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Third Military Medical University, 400037, Chongqing, China
| | - Li Tang
- Department of Gastroenterology, The Second Affiliated Hospital of Third Military Medical University, 400037, Chongqing, China
| | - Fengchao Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, College of Preventive Medicine, Third Military Medical University, Chongqing, 400037, China
| | - Shengjie Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Shuang Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Third Military Medical University, 400037, Chongqing, China
| | - Yuanyuan Lei
- Department of Gastroenterology, The Second Affiliated Hospital of Third Military Medical University, 400037, Chongqing, China
| | - Sumin Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Third Military Medical University, 400037, Chongqing, China
| | - Zhuo Xie
- Department of Gastroenterology, The Second Affiliated Hospital of Third Military Medical University, 400037, Chongqing, China
| | - Wei Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Third Military Medical University, 400037, Chongqing, China
| | - Xiaoyan Zhao
- Department of Gastroenterology, The Second Affiliated Hospital of Third Military Medical University, 400037, Chongqing, China.
| | - Bo Tang
- Department of Gastroenterology, The Second Affiliated Hospital of Third Military Medical University, 400037, Chongqing, China.
| | - Shiming Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Third Military Medical University, 400037, Chongqing, China.
| |
Collapse
|
31
|
Liu Y, Tian X, Daniel RC, Okeugo B, Armbrister SA, Luo M, Taylor CM, Wu G, Rhoads JM. Impact of probiotic Limosilactobacillus reuteri DSM 17938 on amino acid metabolism in the healthy newborn mouse. Amino Acids 2022; 54:1383-1401. [PMID: 35536363 DOI: 10.1007/s00726-022-03165-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 04/19/2022] [Indexed: 12/15/2022]
Abstract
We studied the effect of feeding a single probiotic Limosilactobacillus reuteri DSM 17938 (LR 17938) on the luminal and plasma levels of amino acids and their derivatives in the suckling newborn mouse, using gas chromatography and high-performance liquid chromatography. We found that LR 17938 increased the relative abundance of many amino acids and their derivatives in stool, while it simultaneously significantly reduced the plasma levels of three amino acids (serine, citrulline, and taurine). Many peptides and dipeptides were increased in stool and plasma, notably gamma-glutamyl derivatives of amino acids, following ingestion of the LR 17938. Gamma-glutamyl transformation of amino acids facilitates their absorption. LR 17938 significantly upregulated N-acetylated amino acids, the levels of which could be useful biomarkers in plasma and warrant further investigation. Specific fecal microbiota were associated with higher levels of fecal amino acids and their derivatives. Changes in luminal and circulating levels of amino acid derivatives, polyamines, and tryptophan metabolites may be mechanistically related to probiotic efficacy.
Collapse
Affiliation(s)
- Yuying Liu
- Division of Gastroenterology, Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.140A, Houston, TX, 77030, USA.
| | - Xiangjun Tian
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rhea C Daniel
- Division of Gastroenterology, Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.140A, Houston, TX, 77030, USA
| | - Beanna Okeugo
- Division of Gastroenterology, Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.140A, Houston, TX, 77030, USA
| | - Shabba A Armbrister
- Division of Gastroenterology, Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.140A, Houston, TX, 77030, USA
| | - Meng Luo
- Department of Microbiology, Immunology and Parasitology, Louisiana State University School of Medicine, New Orleans, LA, USA
| | - Christopher M Taylor
- Department of Microbiology, Immunology and Parasitology, Louisiana State University School of Medicine, New Orleans, LA, USA
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - J Marc Rhoads
- Division of Gastroenterology, Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.140A, Houston, TX, 77030, USA
| |
Collapse
|
32
|
A potent HNF4α agonist reveals that HNF4α controls genes important in inflammatory bowel disease and Paneth cells. PLoS One 2022; 17:e0266066. [PMID: 35385524 PMCID: PMC8985954 DOI: 10.1371/journal.pone.0266066] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 03/11/2022] [Indexed: 11/19/2022] Open
Abstract
HNF4α has been implicated in IBD through a number of genome-wide association studies. Recently, we developed potent HNF4α agonists, including N-trans caffeoyltyramine (NCT). NCT was identified by structural similarity to previously the previously identified but weak HNF4α agonists alverine and benfluorex. Here, we administered NCT to mice fed a high fat diet, with the goal of studying the role of HNF4α in obesity-related diseases. Intestines from NCT-treated mice were examined by RNA-seq to determine the role of HNF4α in that organ. Surprisingly, the major classes of genes altered by HNF4α were involved in IBD and Paneth cell biology. Multiple genes downregulated in IBD were induced by NCT. Paneth cells identified by lysozyme expression were reduced in high fat fed mice. NCT reversed the effect of high fat diet on Paneth cells, with multiple markers being induced, including a number of defensins, which are critical for Paneth cell function and intestinal barrier integrity. NCT upregulated genes that play important role in IBD and that are downregulated in that disease. It reversed the loss of Paneth cell markers that occurred in high fat diet fed mice. These data suggest that HNF4α could be a therapeutic target for IBD and that the agonists that we have identified could be candidate therapeutics.
Collapse
|
33
|
Li M, Yang L, Mu C, Sun Y, Gu Y, Chen D, Liu T, Cao H. Gut microbial metabolome in inflammatory bowel disease: From association to therapeutic perspectives. Comput Struct Biotechnol J 2022; 20:2402-2414. [PMID: 35664229 PMCID: PMC9125655 DOI: 10.1016/j.csbj.2022.03.038] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 03/26/2022] [Accepted: 03/31/2022] [Indexed: 12/11/2022] Open
Abstract
Inflammatory bowel disease (IBD), comprising Crohn's disease (CD) and ulcerative colitis (UC), is a set of clinically chronic, relapsing gastrointestinal inflammatory disease and lacks of an absolute cure. Although the precise etiology is unknown, developments in high-throughput microbial genomic sequencing significantly illuminate the changes in the intestinal microbial structure and functions in patients with IBD. The application of microbial metabolomics suggests that the microbiota can influence IBD pathogenesis by producing metabolites, which are implicated as crucial mediators of host-microbial crosstalk. This review aims to elaborate the current knowledge of perturbations of the microbiome-metabolome interface in IBD with description of altered composition and metabolite profiles of gut microbiota. We emphasized and elaborated recent findings of several potentially protective metabolite classes in IBD, including fatty acids, amino acids and derivatives and bile acids. This article will facilitate a deeper understanding of the new therapeutic approach for IBD by applying metabolome-based adjunctive treatment.
Collapse
Key Words
- AMPs, Antimicrobial peptides
- BAs, Bile acids
- BC, Bray Curtis
- CD, Crohn’s disease
- CDI, Clostridioides difficile infection
- DC, Diversion colitis
- DCA, Deoxycholic acid
- DSS, Dextran sulfate sodium
- FAs, Fatty acid
- FMT, Fecal microbiota transplantation
- FODMAP, Fermentable oligosaccharide, disaccharide, monosaccharide, and polyol
- GC–MS, Gas chromatography-mass spectrometry
- Gut microbiota
- HDAC, Histone deacetylase
- IBD, Inflammatory bowel disease
- Inflammatory bowel diseases
- LC-MS, Liquid chromatography-mass spectrometry
- LCA, Lithocholic acid
- LCFAs, Long-chain fatty acids
- MCFAs, Medium-chain fatty acids
- MD, Mediterranean diet
- MS, Mass spectrometry
- Metabolite
- Metabolomics
- Metagenomics
- Microbial therapeutics
- NMR, Nuclear magnetic resonance
- PBAs, Primary bile acids
- SBAs, Secondary bile acids
- SCD, Special carbohydrate diet
- SCFAs, Short-chain fatty acids
- TNBS, 2,4,6-trinitro-benzene sulfonic acid
- UC, Ulcerative colitis
- UDCA, Ursodeoxycholic acid
- UPLC-MS, ultraperformance liquid chromatography coupled to mass spectrometry
- UU, Unweighted UniFrac
- WMS, Whole-metagenome shotgun
Collapse
Affiliation(s)
| | | | | | - Yue Sun
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Yu Gu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Danfeng Chen
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Tianyu Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| |
Collapse
|
34
|
Shen R, Liu D, Wang X, Guo Z, Sun H, Song Y, Wang D. DNA Damage and Activation of cGAS/STING Pathway Induce Tumor Microenvironment Remodeling. Front Cell Dev Biol 2022; 9:828657. [PMID: 35265630 PMCID: PMC8900217 DOI: 10.3389/fcell.2021.828657] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 12/27/2021] [Indexed: 12/11/2022] Open
Abstract
DNA damage occurs throughout tumorigenesis and development. The immunogenicity of DNA makes it an immune stimulatory molecule that initiates strong inflammatory responses. The cGAS/STING pathway has been investigated as a critical receptor in both exogenous and endogenous DNA sensing to activate the innate immune response. Growing lines of evidence have indicated that activation of the cGAS/STING pathway is critical in antitumor immunity. Recent studies have demonstrated the outstanding advancement of this pathway in tumor-combined immunotherapy; accordingly, increased studies focus on exploration of STING pathway agonists and analogues. However, current studies propose the potential use of the cGAS/STING pathway in tumor initiation and metastasis. Here, we review the molecular mechanisms and activation of the cGAS/STING pathway, and the relationship between DNA damage and this pathway, particularly highlighting the remodeling of immune contexture in tumor environment (TME) triggered by cascade inflammatory signals. A detailed understanding of TME reprogramming initiated by this pathway may pave the way for the development of new therapeutic strategies and rational clinical application.
Collapse
Affiliation(s)
- Rong Shen
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Disheng Liu
- The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaoning Wang
- School of Medicine, Shandong University, Jinan, China
| | - Zhao Guo
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Haonan Sun
- The First Hospital of Lanzhou University, Lanzhou, China
| | - Yanfeng Song
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Degui Wang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| |
Collapse
|
35
|
Mecocci S, Ottaviani A, Razzuoli E, Fiorani P, Pietrucci D, De Ciucis CG, Dei Giudici S, Franzoni G, Chillemi G, Cappelli K. Cow Milk Extracellular Vesicle Effects on an In Vitro Model of Intestinal Inflammation. Biomedicines 2022; 10:biomedicines10030570. [PMID: 35327370 PMCID: PMC8945533 DOI: 10.3390/biomedicines10030570] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/19/2022] [Accepted: 02/27/2022] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) are lipid bilayer nano-dimensional spherical structures and act mainly as signaling mediators between cells, in particular modulating immunity and inflammation. Milk-derived EVs (mEVs) can have immunomodulatory and anti-inflammatory effects, and milk is one of the most promising food sources of EVs. In this context, this study aimed to evaluate bovine mEVs anti-inflammatory and immunomodulating effects on an in vitro co-culture (Caco-2 and THP-1) model of intestinal inflammation through gene expression evaluation with RT-qPCR and cytokine release through ELISA. After establishing a pro-inflammatory environment due to IFN-γ and LPS stimuli, CXCL8, IL1B, TNFA, IL12A, IL23A, TGFB1, NOS2, and MMP9 were significantly up-regulated in inflamed Caco-2 compared to the basal co-culture. Moreover, IL-17, IL-1β, IL-6, TNF-α release was increased in supernatants of THP-1. The mEV administration partially restored initial conditions with an effective anti-inflammatory activity. Indeed, a decrease in gene expression and protein production of most of the tested cytokines was detected, together with a significant gene expression decrease in MMP9 and the up-regulation of MUC2 and TJP1. These results showed a fundamental capability of mEVs to modulate inflammation and their potential beneficial effect on the intestinal mucosa.
Collapse
Affiliation(s)
- Samanta Mecocci
- Department of Veterinary Medicine, University of Perugia, 06123 Perugia, Italy;
- Sports Horse Research Center (CRCS), University of Perugia, 06123 Perugia, Italy
| | - Alessio Ottaviani
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Elisabetta Razzuoli
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Piazza Borgo Pila 39-24, 16129 Genova, Italy;
- Correspondence: (E.R.); (G.C.); (K.C.); Tel.: +39-010-542274 (E.R.); +39-0761-357429 (G.C.); +39-075-5857722 (K.C.)
| | - Paola Fiorani
- Institute of Translational Pharmacology, National Research Council, CNR, 00133 Rome, Italy;
| | - Daniele Pietrucci
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, IBIOM, National Research Council, CNR, 70126 Bari, Italy;
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy
| | - Chiara Grazia De Ciucis
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Piazza Borgo Pila 39-24, 16129 Genova, Italy;
| | - Silvia Dei Giudici
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (S.D.G.); (G.F.)
| | - Giulia Franzoni
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (S.D.G.); (G.F.)
| | - Giovanni Chillemi
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, IBIOM, National Research Council, CNR, 70126 Bari, Italy;
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy
- Correspondence: (E.R.); (G.C.); (K.C.); Tel.: +39-010-542274 (E.R.); +39-0761-357429 (G.C.); +39-075-5857722 (K.C.)
| | - Katia Cappelli
- Department of Veterinary Medicine, University of Perugia, 06123 Perugia, Italy;
- Sports Horse Research Center (CRCS), University of Perugia, 06123 Perugia, Italy
- Correspondence: (E.R.); (G.C.); (K.C.); Tel.: +39-010-542274 (E.R.); +39-0761-357429 (G.C.); +39-075-5857722 (K.C.)
| |
Collapse
|
36
|
Wei W, Zhang Y, Li R, Cao Y, Yan X, Ma Y, Zhang Y, Yang M, Zhang M. Oral Delivery of Pterostilbene by L-Arginine-Mediated “Nano-Bomb” Carrier for the Treatment of Ulcerative Colitis. Int J Nanomedicine 2022; 17:603-616. [PMID: 35177902 PMCID: PMC8843770 DOI: 10.2147/ijn.s347506] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/25/2022] [Indexed: 12/23/2022] Open
Affiliation(s)
- Wei Wei
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Xi’an No.1 Hospital, Shaanxi Institute of Ophthalmology, Shaanxi Key Laboratory of Ophthalmology, Clinical Research Center for Ophthalmology Diseases of Shaanxi Province, First Affiliated Hospital of Northwestern University, Xi’an, Shaanxi, People’s Republic of China
| | - Yujie Zhang
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Runqing Li
- Department of Radiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Yameng Cao
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Xiangji Yan
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Yana Ma
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Yuanyuan Zhang
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Mei Yang
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Correspondence: Mei Yang; Mingzhen Zhang, Email ;
| | - Mingzhen Zhang
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| |
Collapse
|
37
|
Yu D, Du J, Pu X, Zheng L, Chen S, Wang N, Li J, Chen S, Pan S, Shen B. The Gut Microbiome and Metabolites Are Altered and Interrelated in Patients With Rheumatoid Arthritis. Front Cell Infect Microbiol 2022; 11:763507. [PMID: 35145919 PMCID: PMC8821809 DOI: 10.3389/fcimb.2021.763507] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 12/28/2021] [Indexed: 01/04/2023] Open
Abstract
The relationship among the gut microbiome, global fecal metabolites and rheumatoid arthritis (RA) has not been systematically evaluated. In this study, we performed 16S rDNA sequencing and liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based nontargeted metabolomic profiling on feces of 26 untreated RA patients and 26 healthy controls. Twenty-six genera and forty-one MS2-identified metabolites were significantly altered in the RA patients. Klebsiella, Escherichia, Eisenbergiella and Flavobacterium were more abundant in the RA patients, while Fusicatenibacter, Megamonas and Enterococcus were more abundant in the healthy controls. Function prediction analysis demonstrated that the biosynthesis pathways of amino acids, such as L-arginine and aromatic amino acids, were depleted in the RA group. In the metabolome results, fecal metabolites including glycerophospholipids (PC(18:3(9Z,12Z,15Z)/16:1(9Z)), lysoPE 19:1, lysoPE 18:0, lysoPC(18:0/0:0)), sphingolipids (Cer(d18:0/16:0), Cer(d18:0/12:0), Cer(d18:0/14:0)), kynurenic acid, xanthurenic acid and 3-hydroxyanthranilic acid were remarkably altered between the RA patients and healthy controls. Dysregulation of pathways, such as tryptophan metabolism, alpha-linolenic acid metabolism and glycerophospholipid metabolism, may contribute to the development of RA. Additionally, we revealed that the gut microbiome and metabolites were interrelated in the RA patients, while Escherichia was the core genus. By depicting the overall landscape of the intestinal microbiome and metabolome in RA patients, our study could provide possible novel research directions regarding RA pathogenesis and targeted therapy.
Collapse
Affiliation(s)
- Die Yu
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Juping Du
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Xia Pu
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Liyuan Zheng
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Shuaishuai Chen
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Na Wang
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Jun Li
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Shiyong Chen
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Shaobiao Pan
- Department of Rheumatology and Immunology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
- *Correspondence: Shaobiao Pan, ; Bo Shen,
| | - Bo Shen
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
- *Correspondence: Shaobiao Pan, ; Bo Shen,
| |
Collapse
|
38
|
Doré E, Joly-Beauparlant C, Morozumi S, Mathieu A, Lévesque T, Allaeys I, Duchez AC, Cloutier N, Leclercq M, Bodein A, Payré C, Martin C, Petit-Paitel A, Gelb MH, Rangachari M, Murakami M, Davidovic L, Flamand N, Arita M, Lambeau G, Droit A, Boilard E. The interaction of secreted phospholipase A2-IIA with the microbiota alters its lipidome and promotes inflammation. JCI Insight 2022; 7:152638. [PMID: 35076027 PMCID: PMC8855825 DOI: 10.1172/jci.insight.152638] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 12/01/2021] [Indexed: 12/13/2022] Open
Abstract
Secreted phospholipase A2-IIA (sPLA2-IIA) hydrolyzes phospholipids to liberate lysophospholipids and fatty acids. Given its poor activity toward eukaryotic cell membranes, its role in the generation of proinflammatory lipid mediators is unclear. Conversely, sPLA2-IIA efficiently hydrolyzes bacterial membranes. Here, we show that sPLA2-IIA affects the immune system by acting on the intestinal microbial flora. Using mice overexpressing transgene-driven human sPLA2-IIA, we found that the intestinal microbiota was critical for both induction of an immune phenotype and promotion of inflammatory arthritis. The expression of sPLA2-IIA led to alterations of the intestinal microbiota composition, but housing in a more stringent pathogen-free facility revealed that its expression could affect the immune system in the absence of changes to the composition of this flora. In contrast, untargeted lipidomic analysis focusing on bacteria-derived lipid mediators revealed that sPLA2-IIA could profoundly alter the fecal lipidome. The data suggest that a singular protein, sPLA2-IIA, produces systemic effects on the immune system through its activity on the microbiota and its lipidome.
Collapse
Affiliation(s)
- Etienne Doré
- CHU de Québec-Université Laval Research Center, Department of Microbiology, Infectiology and Immunology, Quebec City, Quebec, Canada
- ARThrite Research Center, University Laval, Quebec City, Quebec, Canada
| | - Charles Joly-Beauparlant
- CHU de Québec-Université Laval Research Center, Endocrinology and Nephrology Axis, Quebec City, Quebec, Canada
| | - Satoshi Morozumi
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
| | - Alban Mathieu
- CHU de Québec-Université Laval Research Center, Endocrinology and Nephrology Axis, Quebec City, Quebec, Canada
| | - Tania Lévesque
- CHU de Québec-Université Laval Research Center, Department of Microbiology, Infectiology and Immunology, Quebec City, Quebec, Canada
- ARThrite Research Center, University Laval, Quebec City, Quebec, Canada
| | - Isabelle Allaeys
- CHU de Québec-Université Laval Research Center, Department of Microbiology, Infectiology and Immunology, Quebec City, Quebec, Canada
- ARThrite Research Center, University Laval, Quebec City, Quebec, Canada
| | - Anne-Claire Duchez
- CHU de Québec-Université Laval Research Center, Department of Microbiology, Infectiology and Immunology, Quebec City, Quebec, Canada
| | - Nathalie Cloutier
- CHU de Québec-Université Laval Research Center, Department of Microbiology, Infectiology and Immunology, Quebec City, Quebec, Canada
| | - Mickaël Leclercq
- CHU de Québec-Université Laval Research Center, Endocrinology and Nephrology Axis, Quebec City, Quebec, Canada
| | - Antoine Bodein
- CHU de Québec-Université Laval Research Center, Endocrinology and Nephrology Axis, Quebec City, Quebec, Canada
| | - Christine Payré
- Côte d’Azur University, The French National Centre for Scientific Research, Institute of Molecular and Cellular Pharmacology, UMR7275, Valbonne Sophia Antipolis, France
| | - Cyril Martin
- The Research Center of the University Institute of Cardiology and Pneumology of Quebec, Quebec City, Quebec, Canada
| | - Agnes Petit-Paitel
- Côte d’Azur University, The French National Centre for Scientific Research, Institute of Molecular and Cellular Pharmacology, UMR7275, Valbonne Sophia Antipolis, France
| | - Michael H. Gelb
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| | - Manu Rangachari
- CHU de Québec-Université Laval Research Center, Neurosciences Axis, Quebec City, Quebec, Canada
| | - Makoto Murakami
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Laetitia Davidovic
- Côte d’Azur University, The French National Centre for Scientific Research, Institute of Molecular and Cellular Pharmacology, UMR7275, Valbonne Sophia Antipolis, France
| | - Nicolas Flamand
- ARThrite Research Center, University Laval, Quebec City, Quebec, Canada
- The Research Center of the University Institute of Cardiology and Pneumology of Quebec, Quebec City, Quebec, Canada
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
- Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama-City University, Yokohama, Japan
| | - Gérard Lambeau
- Côte d’Azur University, The French National Centre for Scientific Research, Institute of Molecular and Cellular Pharmacology, UMR7275, Valbonne Sophia Antipolis, France
| | - Arnaud Droit
- CHU de Québec-Université Laval Research Center, Endocrinology and Nephrology Axis, Quebec City, Quebec, Canada
| | - Eric Boilard
- CHU de Québec-Université Laval Research Center, Department of Microbiology, Infectiology and Immunology, Quebec City, Quebec, Canada
- ARThrite Research Center, University Laval, Quebec City, Quebec, Canada
| |
Collapse
|
39
|
Baumann A, Rajcic D, Brandt A, Sánchez V, Jung F, Staltner R, Nier A, Trauner M, Staufer K, Bergheim I. Alterations of nitric oxide homeostasis as trigger of intestinal barrier dysfunction in non-alcoholic fatty liver disease. J Cell Mol Med 2022; 26:1206-1218. [PMID: 35029027 PMCID: PMC8831936 DOI: 10.1111/jcmm.17175] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 02/07/2023] Open
Abstract
Changes in intestinal nitric oxide metabolism are discussed to contribute for the development of intestinal barrier dysfunction in non‐alcoholic fatty liver disease (NAFLD). To induce steatosis, female C57BL/6J mice were pair‐fed with a liquid control diet (C) or a fat‐, fructose‐ and cholesterol‐rich diet (FFC) for 8 weeks. Mice received the diets ± 2.49 g L‐arginine/kg bw/day for additional 5 weeks. Furthermore, mice fed C or FFC ± L‐arginine/kg bw/day for 8 weeks were concomitantly treated with the arginase inhibitor Nω‐hydroxy‐nor‐L‐arginine (nor‐NOHA, 0.01 g/kg bw). Liver damage, intestinal barrier function, nitric oxide levels and arginase activity in small intestine were assessed. Also, arginase activity was measured in serum from 13 patients with steatosis (NAFL) and 14 controls. The development of steatosis with beginning inflammation was associated with impaired intestinal barrier function, increased nitric oxide levels and a loss of arginase activity in small intestine in mice. L‐arginine supplementation abolished the latter along with an improvement of intestinal barrier dysfunction; nor‐NOHA attenuated these effects. In patients with NAFL, arginase activity in serum was significantly lower than in healthy controls. Our data suggest that increased formation of nitric oxide and a loss of intestinal arginase activity is critical in NAFLD‐associated intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Anja Baumann
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Dragana Rajcic
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Annette Brandt
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Victor Sánchez
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Finn Jung
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Raphaela Staltner
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Anika Nier
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Michael Trauner
- Division of Gastroenterology & Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Katharina Staufer
- Division of Gastroenterology & Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria.,Department of Surgery Division of Transplantation, Medical University of Vienna, Vienna, Austria
| | - Ina Bergheim
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| |
Collapse
|
40
|
Kim YJ, Lee JY, Lee JJ, Jeon SM, Silwal P, Kim IS, Kim HJ, Park CR, Chung C, Han JE, Choi JW, Tak EJ, Yoo JH, Jeong SW, Kim DY, Ketphan W, Kim SY, Jhun BW, Whang J, Kim JM, Eoh H, Bae JW, Jo EK. Arginine-mediated gut microbiome remodeling promotes host pulmonary immune defense against nontuberculous mycobacterial infection. Gut Microbes 2022; 14:2073132. [PMID: 35579969 PMCID: PMC9116420 DOI: 10.1080/19490976.2022.2073132] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/08/2022] [Accepted: 04/18/2022] [Indexed: 02/06/2023] Open
Abstract
Nontuberculous mycobacterial pulmonary diseases (NTM-PDs) are emerging as global health threats with issues of antibiotic resistance. Accumulating evidence suggests that the gut-lung axis may provide novel candidates for host-directed therapeutics against various infectious diseases. However, little is known about the gut-lung axis in the context of host protective immunity to identify new therapeutics for NTM-PDs. This study was performed to identify gut microbes and metabolites capable of conferring pulmonary immunity to NTM-PDs. Using metabolomics analysis of sera from NTM-PD patients and mouse models, we showed that the levels of l-arginine were decreased in sera from NTM-PD patients and NTM-infected mice. Oral administration of l-arginine significantly enhanced pulmonary antimicrobial activities with the expansion of IFN-γ-producing effector T cells and a shift to microbicidal (M1) macrophages in the lungs of NTM-PD model mice. Mice that received fecal microbiota transplants from l-arginine-treated mice showed increased protective host defense in the lungs against NTM-PD, whereas l-arginine-induced pulmonary host defense was attenuated in mice treated with antibiotics. Using 16S rRNA sequencing, we further showed that l-arginine administration resulted in enrichment of the gut microbiota composition with Bifidobacterium species. Notably, oral treatment with either Bifidobacterium pseudolongum or inosine enhanced antimicrobial pulmonary immune defense against NTM infection, even with multidrug-resistant clinical NTM strains. Our findings indicate that l-arginine-induced gut microbiota remodeling with enrichment of B. pseudolongum boosts pulmonary immune defense against NTM infection by driving the protective gut-lung axis in vivo.
Collapse
Affiliation(s)
- Young Jae Kim
- Department of Microbiology, Chungnam National University School of MedicineDaejeon, Korea
- Infection Control Convergence Research Center, Chungnam National University School of MedicineDaejeon, Korea
- Department of Medical Science, Chungnam National University School of MedicineDaejeon, Korea
- Brain Korea 21 FOUR Project for Medical Science, Chungnam National University School of MedicineDaejeon, Korea
| | - June-Young Lee
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, Korea
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Korea
| | - Jae Jin Lee
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern CaliforniaCA, USA
| | - Sang Min Jeon
- Department of Microbiology, Chungnam National University School of MedicineDaejeon, Korea
- Infection Control Convergence Research Center, Chungnam National University School of MedicineDaejeon, Korea
- Department of Medical Science, Chungnam National University School of MedicineDaejeon, Korea
- Brain Korea 21 FOUR Project for Medical Science, Chungnam National University School of MedicineDaejeon, Korea
| | - Prashanta Silwal
- Department of Microbiology, Chungnam National University School of MedicineDaejeon, Korea
- Infection Control Convergence Research Center, Chungnam National University School of MedicineDaejeon, Korea
| | - In Soo Kim
- Department of Microbiology, Chungnam National University School of MedicineDaejeon, Korea
- Infection Control Convergence Research Center, Chungnam National University School of MedicineDaejeon, Korea
- Department of Medical Science, Chungnam National University School of MedicineDaejeon, Korea
| | - Hyeon Ji Kim
- Department of Microbiology, Chungnam National University School of MedicineDaejeon, Korea
- Infection Control Convergence Research Center, Chungnam National University School of MedicineDaejeon, Korea
- Department of Medical Science, Chungnam National University School of MedicineDaejeon, Korea
| | - Cho Rong Park
- Department of Microbiology, Chungnam National University School of MedicineDaejeon, Korea
- Infection Control Convergence Research Center, Chungnam National University School of MedicineDaejeon, Korea
- Department of Medical Science, Chungnam National University School of MedicineDaejeon, Korea
| | - Chaeuk Chung
- Infection Control Convergence Research Center, Chungnam National University School of MedicineDaejeon, Korea
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Chungnam National University School of MedicineDaejeon, Korea
| | - Jeong Eun Han
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, Korea
| | - Jee-Won Choi
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, Korea
| | - Euon Jung Tak
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, Korea
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Korea
| | - Ji-Ho Yoo
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Korea
| | - Su-Won Jeong
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, Korea
| | - Do-Yeon Kim
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, Korea
| | - Warisa Ketphan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern CaliforniaCA, USA
| | - Su-Young Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of MedicineSeoul, South Korea
| | - Byung Woo Jhun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of MedicineSeoul, South Korea
| | - Jake Whang
- Korea Mycobacterium Resource Center (KMRC) & Basic Research Section, The Korean Institute of Tuberculosis (KIT), Cheongju, Korea
| | - Jin-Man Kim
- Department of Pathology, Chungnam National University School of Medicine, Daejeon, Korea
| | - Hyungjin Eoh
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern CaliforniaCA, USA
| | - Jin-Woo Bae
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, Korea
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of MedicineDaejeon, Korea
- Infection Control Convergence Research Center, Chungnam National University School of MedicineDaejeon, Korea
- Department of Medical Science, Chungnam National University School of MedicineDaejeon, Korea
| |
Collapse
|
41
|
Boby N, Cao X, Ransom A, Pace BT, Mabee C, Shroyer MN, Das A, Didier PJ, Srivastav SK, Porter E, Sha Q, Pahar B. Identification, Characterization, and Transcriptional Reprogramming of Epithelial Stem Cells and Intestinal Enteroids in Simian Immunodeficiency Virus Infected Rhesus Macaques. Front Immunol 2021; 12:769990. [PMID: 34887863 PMCID: PMC8650114 DOI: 10.3389/fimmu.2021.769990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/02/2021] [Indexed: 12/18/2022] Open
Abstract
Epithelial cell injury and impaired epithelial regeneration are considered key features in HIV pathogenesis and contribute to HIV-induced generalized immune activation. Understanding the molecular mechanisms underlying the disrupted epithelial regeneration might provide an alternative approach for the treatment of HIV-mediated enteropathy and immune activation. We have observed a significant increased presence of α defensin5+ (HD5) Paneth cells and proliferating Ki67+ epithelial cells as well as decreased expression of E-cadherin expression in epithelial cells during SIV infection. SIV infection did not significantly influence the frequency of LGR5+ stem cells, but the frequency of HD5+ cells was significantly higher compared to uninfected controls in jejunum. Our global transcriptomics analysis of enteroids provided novel information about highly significant changes in several important pathways like metabolic, TCA cycle, and oxidative phosphorylation, where the majority of the differentially expressed genes were downregulated in enteroids grown from chronically SIV-infected macaques compared to the SIV-uninfected controls. Despite the lack of significant reduction in LGR5+ stem cell population, the dysregulation of several intestinal stem cell niche factors including Notch, mTOR, AMPK and Wnt pathways as well as persistence of inflammatory cytokines and chemokines and loss of epithelial barrier function in enteroids further supports that SIV infection impacts on epithelial cell proliferation and intestinal homeostasis.
Collapse
Affiliation(s)
- Nongthombam Boby
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Xuewei Cao
- Department of Mathematical Sciences, Michigan Technological University, Houghton, MI, United States
| | - Alyssa Ransom
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Barcley T Pace
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Christopher Mabee
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Monica N Shroyer
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, United States
| | - Arpita Das
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, United States
| | - Peter J Didier
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Sudesh K Srivastav
- Department of Biostatistics, Tulane University, New Orleans, LA, United States
| | - Edith Porter
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
| | - Qiuying Sha
- Department of Mathematical Sciences, Michigan Technological University, Houghton, MI, United States
| | - Bapi Pahar
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States.,Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States.,Department of Tropical Medicine, Tulane School of Public Health and Tropical Medicine, New Orleans, LA, United States
| |
Collapse
|
42
|
Citrulline, Biomarker of Enterocyte Functional Mass and Dietary Supplement. Metabolism, Transport, and Current Evidence for Clinical Use. Nutrients 2021; 13:nu13082794. [PMID: 34444954 PMCID: PMC8398474 DOI: 10.3390/nu13082794] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 12/25/2022] Open
Abstract
L-Citrulline is a non-essential but still important amino acid that is released from enterocytes. Because plasma levels are reduced in case of impaired intestinal function, it has become a biomarker to monitor intestinal integrity. Moreover, oxidative stress induces protein citrullination, and antibodies against anti-citrullinated proteins are useful to monitor rheumatoid diseases. Citrullinated histones, however, may even predict a worse outcome in cancer patients. Supplementation of citrulline is better tolerated compared to arginine and might be useful to slightly improve muscle strength or protein balance. The following article shall provide an overview of L-citrulline properties and functions, as well as the current evidence for its use as a biomarker or as a therapeutic supplement.
Collapse
|
43
|
Chen S, Duan Y, Wu Y, Yang D, An J. A Novel Integrated Metabolism-Immunity Gene Expression Model Predicts the Prognosis of Lung Adenocarcinoma Patients. Front Pharmacol 2021; 12:728368. [PMID: 34393804 PMCID: PMC8361602 DOI: 10.3389/fphar.2021.728368] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Although multiple metabolic pathways are involved in the initiation, progression, and therapy of lung adenocarcinoma (LUAD), the tumor microenvironment (TME) for immune cell infiltration that is regulated by metabolic enzymes has not yet been characterized. Methods: 517 LUAD samples and 59 non-tumor samples were obtained from The Cancer Genome Atlas (TCGA) database as the training cohort. Kaplan-Meier analysis and Univariate Cox analysis were applied to screen the candidate metabolic enzymes for their role in relation to survival rate in LUAD patients. A prognostic metabolic enzyme signature, termed the metabolic gene risk score (MGRS), was established based on multivariate Cox proportional hazards regression analysis and was verified in an independent test cohort, GSE31210. In addition, we analyzed the immune cell infiltration characteristics in patients grouped by their Risk Score. Furthermore, the prognostic value of these four enzymes was verified in another independent cohort by immunohistochemistry and an optimized model of the metabolic-immune protein risk score (MIPRS) was constructed. Results: The MGRS model comprising 4 genes (TYMS, NME4, LDHA, and SMOX) was developed to classify patients into high-risk and low-risk groups. Patients with a high-risk score had a poor prognosis and exhibited activated carbon and nucleotide metabolism, both of which were associated with changes to TME immune cell infiltration characteristics. In addition, the optimized MIPRS model showed more accurate predictive power in prognosis of LUAD. Conclusion: Our study revealed an integrated metabolic enzyme signature as a reliable prognostic tool to accurately predict the prognosis of LUAD.
Collapse
Affiliation(s)
- Songming Chen
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Xiangya Hospital, Central South University, Changsha, China.,Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China
| | - Yumei Duan
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Yanhao Wu
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Desong Yang
- Thoracic Surgery Department II, Hunan Cancer Hospital & the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jian An
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China.,National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China.,Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, China.,Hunan Provincial Clinical Research Center for Respiratory Diseases, Changsha, China
| |
Collapse
|
44
|
CD101 as an indicator molecule for pathological changes at the interface of host-microbiota interactions. Int J Med Microbiol 2021; 311:151497. [PMID: 33773220 DOI: 10.1016/j.ijmm.2021.151497] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 02/22/2021] [Accepted: 03/16/2021] [Indexed: 11/21/2022] Open
Abstract
Intestinal microbiota signal to local and distant tissues in the body. Thus, they also regulate biochemical, metabolic and immunological processes in the gut and in the pancreas. Vice versa, eating habits or the immune system of the host shape the intraluminal microbiota. Disruptions of these versatile host-microbiota interactions underlie the pathogenesis of complex immune-mediated disorders such as inflammatory bowel disease (IBD) or type 1 diabetes (T1D). Consequently, dysbiosis and increased intestinal permeability associated with both disorders change the biology of underlying tissues, as evidenced, for example, by an altered expression of surface markers such as CD101 on immune cells located at these dynamic host-microbiota interfaces. CD101, a heavily glycosylated member of the immunoglobulin superfamiliy, is predominantly detected on myeloid cells, intraepithelial lymphocytes (IELs) and regulatory T cells (Tregs) in the gut. The expression of CD101 on both myeloid cells and T lymphocytes protects from experimental enterocolitis and T1D. The improved outcome of both diseases is associated with an anti-inflammatory cytokine profile and a reduced expansion of T cells. However, distinct bacteria suppress the expression of CD101 on myeloid cells, similar as does inflammation on T cells. Thus, the reduced CD101 expression in T1D and IBD patients might be a consequence of an altered composition of the intestinal microbiota, enhanced bacterial translocation and a subsequent primining of local and systemic inflammatory immune responses.
Collapse
|
45
|
Crovesy L, El-Bacha T, Rosado EL. Modulation of the gut microbiota by probiotics and symbiotics is associated with changes in serum metabolite profile related to a decrease in inflammation and overall benefits to metabolic health: a double-blind randomized controlled clinical trial in women with obesity. Food Funct 2021; 12:2161-2170. [PMID: 33565558 DOI: 10.1039/d0fo02748k] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Modulation of the gut microbiota may help in treating obesity by improving host metabolic health. We aimed to evaluate the effects of probiotics or symbiotics on body weight and serum metabolite profile in women with obesity. A double-blind, parallel, randomized, controlled clinical trial was conducted with 32 adult women with body mass index ranging from 30 to 34.9 kg m-2. Volunteers followed a low-energy diet and were subjected to 8 weeks intervention: probiotic group (PG - Bifidobacterium lactis UBBLa-70, n = 10), symbiotic group (SG - Bifidobacterium lactis UBBLa-70 and fructooligosaccharide, n = 11), or control group (CG - placebo, n = 11). Analyses of anthropometric variables, gut microbiota and serum metabolites by 1H nuclear magnetic resonance (NMR) were performed at baseline and after the intervention. Multivariate statistics showed that all groups presented a decrease in glycerol and increase in arginine, glutamine and 2-oxoisovalerate. Therefore, a low-energy diet per se promoted changes in the metabolite profile related to decreased inflammation and positive effects on body weight. SG presented unique changes in metabolites (increase in pyruvate and alanine and decrease in citrate and BCAA). Negative correlations between arginine and glutamine with fat mass were observed in the SG. PG presented a decrease in 1H NMR lipid signals and negative correlation between Verrucomicrobia and Firmicutes with (CH2)n lipids. Both probiotics and symbiotics promoted changes in metabolites related to improved metabolic health. Specific metabolite changes following symbiotic intervention might suggest some advantage in providing Bifidobacterium lactis in combination with fructooligosaccharide in a low-energy diet, rather than probiotics or diet alone. Clinical trial: NCT02505854.
Collapse
Affiliation(s)
- Louise Crovesy
- Department of Nutrition and Dietetics (DND), Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro - RJ, Brazil.
| | - Tatiana El-Bacha
- Lebiome - Núcleo de estudos com bioativos, Mitocôndria e metabolismo da placenta, Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro - RJ, Brazil
| | - Eliane Lopes Rosado
- Department of Nutrition and Dietetics (DND), Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro - RJ, Brazil.
| |
Collapse
|
46
|
Associations of serum short-chain fatty acids with circulating immune cells and serum biomarkers in patients with multiple sclerosis. Sci Rep 2021; 11:5244. [PMID: 33664396 PMCID: PMC7933417 DOI: 10.1038/s41598-021-84881-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/22/2021] [Indexed: 02/06/2023] Open
Abstract
Altered composition of gut bacteria and changes to the production of their bioactive metabolites, the short-chain fatty acids (SCFAs), have been implicated in the development of multiple sclerosis (MS). However, the immunomodulatory actions of SCFAs and intermediaries in their ability to influence MS pathogenesis are uncertain. In this study, levels of serum SCFAs were correlated with immune cell abundance and phenotype as well as with other relevant serum factors in blood samples taken at first presentation of Clinically Isolated Syndrome (CIS; an early form of MS) or MS and compared to healthy controls. There was a small but significant reduction in propionate levels in the serum of patients with CIS or MS compared with healthy controls. The frequencies of circulating T follicular regulatory cells and T follicular helper cells were significantly positively correlated with serum levels of propionate. Levels of butyrate associated positively with frequencies of IL-10-producing B-cells and negatively with frequencies of class-switched memory B-cells. TNF production by polyclonally-activated B-cells correlated negatively with acetate levels. Levels of serum SCFAs associated with changes in circulating immune cells and biomarkers implicated in the development of MS.
Collapse
|
47
|
Bile Acids and Microbiota: Multifaceted and Versatile Regulators of the Liver-Gut Axis. Int J Mol Sci 2021; 22:ijms22031397. [PMID: 33573273 PMCID: PMC7866539 DOI: 10.3390/ijms22031397] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/25/2021] [Accepted: 01/28/2021] [Indexed: 12/12/2022] Open
Abstract
After their synthesis from cholesterol in hepatic tissues, bile acids (BAs) are secreted into the intestinal lumen. Most BAs are subsequently re-absorbed in the terminal ileum and are transported back for recycling to the liver. Some of them, however, reach the colon and change their physicochemical properties upon modification by gut bacteria, and vice versa, BAs also shape the composition and function of the intestinal microbiota. This mutual interplay of both BAs and gut microbiota regulates many physiological processes, including the lipid, carbohydrate and energy metabolism of the host. Emerging evidence also implies an important role of this enterohepatic BA circuit in shaping mucosal colonization resistance as well as local and distant immune responses, tissue physiology and carcinogenesis. Subsequently, disrupted interactions of gut bacteria and BAs are associated with many disorders as diverse as Clostridioides difficile or Salmonella Typhimurium infection, inflammatory bowel disease, type 1 diabetes, asthma, metabolic syndrome, obesity, Parkinson’s disease, schizophrenia and epilepsy. As we cannot address all of these interesting underlying pathophysiologic mechanisms here, we summarize the current knowledge about the physiologic and pathogenic interplay of local site microbiota and the enterohepatic BA metabolism using a few selected examples of liver and gut diseases.
Collapse
|
48
|
Citrulline supplementation attenuates the development of non-alcoholic steatohepatitis in female mice through mechanisms involving intestinal arginase. Redox Biol 2021; 41:101879. [PMID: 33550112 PMCID: PMC7868995 DOI: 10.1016/j.redox.2021.101879] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/22/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is by now the most prevalent liver disease worldwide. The non-proteogenic amino acid l-citrulline (L-Cit) has been shown to protect mice from the development of NAFLD. Here, we aimed to further assess if L-Cit also attenuates the progression of a pre-existing diet-induced NAFLD and to determine molecular mechanisms involved. Female C57BL/6J mice were either fed a liquid fat-, fructose- and cholesterol-rich diet (FFC) or control diet (C) for 8 weeks to induce early stages of NASH followed by 5 more weeks with either FFC-feeding +/- 2.5 g L-Cit/kg bw or C-feeding. In addition, female C57BL/6J mice were either pair-fed a FFC +/- 2.5 g L-Cit/kg bw +/- 0.01 g/kg bw i.p. N(ω)-hydroxy-nor-l-arginine (NOHA) or C diet for 8 weeks. The protective effects of supplementing L-Cit on the progression of a pre-existing NAFLD were associated with an attenuation of 1) the increased translocation of bacterial endotoxin and 2) the loss of tight junction proteins as well as 3) arginase activity in small intestinal tissue, while no marked changes in intestinal microbiota composition were prevalent in small intestine. Treatment of mice with the arginase inhibitor NOHA abolished the protective effects of L-Cit on diet-induced NAFLD. Our results suggest that the protective effects of L-Cit on the development and progression of NAFLD are related to alterations of intestinal arginase activity and intestinal permeability. l-citrulline diminished progression of non-alcoholic fatty liver disease (NAFLD). l-citrulline protects from fructose-induced small intestinal barrier dysfunction. NASH development is associated with a loss of arginase activity in small intestine. l-citrulline improves intestinal arginase activity in diet-induced NAFLD. Arginase inhibitor attenuates effects of l-citrulline on NAFLD development.
Collapse
|
49
|
Castellano F, Molinier-Frenkel V. Control of T-Cell Activation and Signaling by Amino-Acid Catabolizing Enzymes. Front Cell Dev Biol 2020; 8:613416. [PMID: 33392202 PMCID: PMC7773816 DOI: 10.3389/fcell.2020.613416] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
Amino acids are essential for protein synthesis, epigenetic modification through the methylation of histones, and the maintenance of a controlled balance of oxidoreduction via the production of glutathione and are precursors of certain neurotransmitters. T lymphocytes are particularly sensitive to fluctuations in amino acid levels. During evolution, the production of amino-acid catabolizing enzymes by mainly antigen-presenting cells has become a physiological mechanism to control T-cell activation and polarization. The action of these enzymes interferes with TCR and co-stimulation signaling, allowing tuning of the T-cell response. These capacities can be altered in certain pathological conditions, with relevant consequences for the development of disease.
Collapse
Affiliation(s)
- Flavia Castellano
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France.,AP-HP, Hopital Henri Mondor, Departement Immunologie-Hématologie, Creteil, France
| | - Valérie Molinier-Frenkel
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France.,AP-HP, Hopital Henri Mondor, Departement Immunologie-Hématologie, Creteil, France
| |
Collapse
|