1
|
Lei H, Jiang Y, Chen Z, Yao J, Ma W, Huang Y, Zhang P, Xie Z, Zhu L, Tang W. Unveiling the influence of lipidomes on inflammatory bowel disease: a bidirectional mendelian randomization study. BMC Gastroenterol 2025; 25:247. [PMID: 40217472 PMCID: PMC11992711 DOI: 10.1186/s12876-025-03858-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 04/07/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Plasma lipid homeostasis is pivotal in maintaining intestinal health. Inflammatory bowel disease (IBD), encompassing ulcerative colitis (UC) and Crohn's disease (CD) as distinct subtypes, manifests unique metabolic signatures. However, the specific roles of lipids in the pathogenesis and therapeutic targeting of IBD remain inadequately explored. This study aims to delineate the genetic influences of plasma lipids on IBD risk. METHODS We obtained genome-wide association study (GWAS) summary statistics of lipidomes and IBD (including UC and CD) from published studies to perform two-sample Mendelian randomization (MR) analyses. Outliers were removed using radial MR, followed by the application of the inverse-variance weighted (IVW) method to assess causal relationships. Sensitivity analyses were also conducted to validate the robustness of the primary results of the MR analyses. Additionally, reverse MR analyses were performed to evaluate the potential for reverse causality. RESULTS The MR analysis identified fourteen lipid species significantly associated with IBD, four with UC, and ten with CD. Phosphatidylcholine (PC; P < 0 .05) and lysophosphatidylcholine (OR = 0.83, P < 0.001) were instrumental in UC, while in CD, alongside these, cholesterol ester (OR = 0.86, P < 0.001), diacylglycerol (OR = 1.21, P = 0.004), and lysophosphatidylethanolamine (OR = 1.30, P < 0.001) also demonstrated causal links. Reverse MR analysis revealed no significant associations between IBDs and 179 lipid species. CONCLUSION This bidirectional MR study has uncovered genetic evidence of a causal relationship between lipidome and IBD, identifying potential therapeutic targets for IBD treatment. The findings suggest that elevated partial phosphatidylcholine, lysophosphatidylcholine, and cholesterol ester levels could reduce the risk of IBD, indicating a potential protective role for these lipid molecules. This study also underscores the critical role of lipidome variability in advancing our understanding of IBD's pathogenic processes and in developing targeted therapies.
Collapse
Affiliation(s)
- Hang Lei
- Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuhong Jiang
- Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zhe Chen
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jiaqi Yao
- Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Wenjun Ma
- Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Yubei District, Chongqing, 401147, China
| | - Yiqi Huang
- Department of Nephrology, Shaoxing Second Hospital, Shaoxing, 312000, Zhejiang, China
| | - Pengcheng Zhang
- Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zhijun Xie
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Lv Zhu
- Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Wenfu Tang
- Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
2
|
Zhang J, Li Z, Zhang Y, Guo YL, Zhu YR, Xia WX, Dai Y, Xia YF. Mume Fructus (Prunus mume Sieb. et Zucc.) extract accelerates colonic mucosal healing of mice with colitis induced by dextran sulfate sodium through potentiation of cPLA2-mediated lysophosphatidylcholine synthesis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 119:154985. [PMID: 37516090 DOI: 10.1016/j.phymed.2023.154985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/15/2023] [Accepted: 07/15/2023] [Indexed: 07/31/2023]
Abstract
BACKGROUND Mume Fructus (MF) is the fruit of Prunus mume Sieb. et Zucc, a plant of Rosaceae family. Previous studies demonstrated that MF was capable of ameliorating ulcerative colitis (UC) in mice, its action mechanism needs to be clarified. PURPOSE This study deciphered whether and how MF extract accelerates colonic mucosal healing, the therapeutic endpoint of UC. METHODS Biochemical, histopathological and qRT-PCR analyses were utilized to define the therapeutic efficacy of MF on dextran sulfate sodium (DSS)-induced colitis in mice. UHPLC-QTOF-MS/MS-based metabolomics technique was adopted to explore the changes of endogenous metabolites associated with UC and responses to MF intervention. qRT-PCR analysis was performed to confirm the molecular pathway in vivo. The effects of MF and lysophosphatidylcholine (LPC) on cell viability, wound healing, proliferation, and migration were examined through a series of in vitro experiments. Moreover, the effects of different subtypes of phospholipase A2 (PLA2) inhibitors on MF-treated colonic epithelial cells were detected by wound healing test and transwell assay. RESULTS Orally administered MF could alleviate colitis in mice mainly by accelerating the healing of colonic mucosa. Guided by an unbiased metabolomics screen, we identified LPC synthesis as a major modifying pathway in colitis mice after MF treatment. Notably, MF facilitated the synthesis of LPC by enhancing the expression of PLA2 in colitis mice. Mechanistically, MF and LPC accelerated wound closure by promoting cell migration. Moreover, the promotion of MF on wound healing and migration of colonic epithelial cells was blunted by a cytosolic phospholipase A2 (cPLA2) inhibitor. CONCLUSION MF can facilitate colonic mucosal healing of mice with colitis through cPLA2-mediated intestinal LPC synthesis, which may become a novel therapeutic agent of UC.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ze Li
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ying Zhang
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yi-Lei Guo
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yan-Rong Zhu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wen-Xin Xia
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yue Dai
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Yu-Feng Xia
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
3
|
Li Q, Zhang W, Zhou E, Tao Y, Wang M, Qi S, Zhao L, Tan Y, Wu L. Integrated microbiomic and metabolomic analyses reveal the mechanisms by which bee pollen and royal jelly lipid extracts ameliorate colitis in mice. Food Res Int 2023; 171:113069. [PMID: 37330827 DOI: 10.1016/j.foodres.2023.113069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/26/2023] [Accepted: 05/29/2023] [Indexed: 06/19/2023]
Abstract
Bee pollen (BP) and royal jelly (RJ) have shown therapeutic effects against colitis, but the functional components contained therein remain elusive. Here, we used an integrated microbiomic-metabolomic strategy to clarify the mechanism by which bee pollen lipid extracts (BPL) and royal jelly lipid extracts (RJL) ameliorated dextran sulfate sodium (DSS)-induced colitis in mice. Lipidomic results showed that levels of ceramide (Cer), lysophosphatidylcholine (LPC), phosphatidylcholine (PC), and phosphatidylethanolamine (PE) were significantly higher in BPL than in RJL. The anti-inflammatory efficacy of BPL surpassed that of RJL, although both BPL and RJL could attenuate DSS-induced colitis through several mechanisms: reducing the disease activity index (DAI); decreasing histopathological damage; inhibiting the expression of genes encoding proinflammatory cytokines; improving intestinal microbial community structure, and modulating host metabolism. These findings demonstrated that BPL and RJL have great potential as functional ingredients for the production of dietary supplements to prevent early colitis.
Collapse
Affiliation(s)
- Qiangqiang Li
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
| | - Wenwen Zhang
- College of Life and Health Sciences, Anhui Science and Technology University, Bengbu 233100, China
| | - Enning Zhou
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
| | - Yuxiao Tao
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
| | - Miao Wang
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
| | - Suzhen Qi
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
| | - Liuwei Zhao
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
| | - Yonggang Tan
- Oncology Center, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| | - Liming Wu
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China.
| |
Collapse
|
4
|
Wu F, Lai S, Feng H, Liu J, Fu D, Wang C, Wang C, Liu J, Li Z, Li P. Protective Effects of Protopanaxatriol Saponins on Ulcerative Colitis in Mouse Based on UPLC-Q/TOF-MS Serum and Colon Metabolomics. Molecules 2022; 27:8346. [PMID: 36500439 PMCID: PMC9738265 DOI: 10.3390/molecules27238346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/05/2022] Open
Abstract
Ulcerative colitis (UC) is a chronic, nonspecific inflammation of the bowel that mainly affects the mucosa and submucosa of the rectum and colon. Ginsenosides are the main active ingredients in ginseng and show many therapeutic effects in anti-inflammatory diseases, cancer, and nervous system regulation. Protopanaxatriol saponin (PTS) is an important part of saponins, and there is no research on its pharmacological effects on colitis. In this study, a model of ulcerative colitis in mice was induced by having mice freely drink 3.5% dextran sodium sulfate (DSS) solution, and UPLC-Q-TOF-MS-based metabolomics methods were applied to explore the therapeutic effect and protective mechanism of PTS for treating UC. The results showed that PTS could significantly prevent colon shortening and pathological damage and alleviate abnormal changes in UC mouse physiological and biochemical parameters. Moreover, PTS intervention regulated proinflammatory cytokines such as TNF-α, IL-6, and IL-1 in serum, and MPO and NO in colon. Interestingly, PTS could significantly inhibit UC mouse metabolic dysfunction by reversing abnormal changes in 29 metabolites and regulating eleven metabolic pathways. PTS has potential application in the treatment of UC and could alleviate UC in mice by affecting riboflavin metabolism, arachidonic acid metabolism, glycerophospholipid metabolism, retinol metabolism, and steroid hormone biosynthesis and by regulating pentose and glucuronate conversion, linoleic acid metabolism, phenylalanine metabolism, ether lipid metabolism, sphingolipid metabolism, and tyrosine metabolism, which points at a direction for further research and for the development of PTS as a novel natural agent.
Collapse
Affiliation(s)
- Fulin Wu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Sihan Lai
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Hao Feng
- College of Basic Medicine Sciences, Jilin University, Changchun 130021, China
| | - Juntong Liu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Dongxing Fu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Caixia Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Cuizhu Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Jinping Liu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Zhuo Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Pingya Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| |
Collapse
|
5
|
Nyström N, Prast-Nielsen S, Correia M, Globisch D, Engstrand L, Schuppe Koistinen I, Halfvarson J. Mucosal and plasma metabolomes in new-onset paediatric inflammatory bowel disease: correlations with disease characteristics and plasma inflammation protein markers. J Crohns Colitis 2022; 17:418-432. [PMID: 36219554 PMCID: PMC10069620 DOI: 10.1093/ecco-jcc/jjac149] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS To advance the understanding of inflammatory bowel disease (IBD) pathophysiology, we compared the mucosal and plasma metabolomes between new-onset paediatric IBD patients and symptomatic non-IBD controls, and correlated plasma inflammation markers and disease characteristics with the altered metabolites. METHODS Paired colonic and ileal biopsies and plasma from 67 treatment-naïve children with incident Crohn's disease (CD; n=47), ulcerative colitis (UC; n=9), and non-IBD controls (n=11) were analysed using ultra-performance liquid chromatography-mass spectrometry (UPLC-MS/MS). Inflammatory plasma proteins (n=92) were assessed. RESULTS The metabolomes in inflamed mucosal biopsies differed between IBD patients and controls. In CD, mucosal levels of several lysophospholipids (lysophosphatidylcholines, lysophosphatidyletanolamines, lysophosphatidylinositols, and lysophosphatidylserines) were decreased, correlating with various plasma metabolites, including amino acid analogues and N-acetylated compounds. In both CD and UC, mucosal sphingolipids, including ceramide (d18:2/24:1, d18:1/24:2), lactosyl-N-palmitoyl-sphingosine (d18:1/16:0), behenoyl sphingomyelin (d18:1/22:0), lignoceroyl sphingomyelin (d18:1/24:0), and/or sphingomyelin (d18:1/24:1, d18:2/24:0) were increased, correlating with sphingolipids, bile acids, and/or N-acetylated metabolites in plasma. Among proteins associated with CD, interleukin-24 correlated with plasma metabolites, including lactosyl-N-palmitoyl sphingosine (d18:1/16:0) and phosphatidyletanolamine (18:1/18:1), haemoglobin, and faecal calprotectin. In UC, interleukin-24, interleukin-17A, and C-C motif chemokine 11 correlated with several plasma metabolites, including N-acetyltryptophan, tryptophan, glycerate, and threonate, and with the paediatric ulcerative colitis activity index, C-reactive protein, and faecal-calprotectin. CONCLUSIONS Mucosal perturbations of lysophospholipids and sphingolipids characterised the metabolome in new-onset paediatric IBD and correlated with plasma metabolites. By integrating plasma metabolomics data with inflammatory proteins and clinical data, we identified clinical and inflammatory markers associated with metabolomic signatures for IBD.
Collapse
Affiliation(s)
- Niklas Nyström
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Stefanie Prast-Nielsen
- Centre for Translational Microbiome Research (CTMR), Karolinska Institutet, Stockholm, Sweden
| | - Mario Correia
- Department of Chemistry - BMC, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Daniel Globisch
- Centre for Translational Microbiome Research (CTMR), Karolinska Institutet, Stockholm, Sweden.,Department of Chemistry - BMC, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Lars Engstrand
- Centre for Translational Microbiome Research (CTMR), Karolinska Institutet, Stockholm, Sweden
| | - Ina Schuppe Koistinen
- Centre for Translational Microbiome Research (CTMR), Karolinska Institutet, Stockholm, Sweden
| | - Jonas Halfvarson
- Department of Gastroenterology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
6
|
Utilizing Sphingomyelinase Sensitizing Liposomes in Imaging Intestinal Inflammation in Dextran Sulfate Sodium-Induced Murine Colitis. Biomedicines 2022; 10:biomedicines10020413. [PMID: 35203622 PMCID: PMC8962329 DOI: 10.3390/biomedicines10020413] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/05/2022] [Accepted: 02/06/2022] [Indexed: 02/04/2023] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by chronic inflammation in the gastrointestinal tract, resulting in severe symptoms. At the moment, the goal of medical treatments is to reduce inflammation. IBD is treated with systemic anti-inflammatory compounds, but they have serious side effects. The treatment that is most efficient and causes the fewest side effects would be the delivery of the drugs on the disease site. This study aimed to investigate the suitability of sphingomyelin (SM) containing liposomes to specifically target areas of inflammation in dextran sulfate sodium-induced murine colitis. Sphingomyelin is a substrate to the sphingomyelinase enzyme, which is only present outside cells in cell stress, like inflammation. When sphingomyelin consisting of liposomes is predisposed to the enzyme, it causes the weakening of the membrane structure. We demonstrated that SM-liposomes are efficiently taken up in intestinal macrophages, indicating their delivery potential. Furthermore, our studies showed that sphingomyelinase activity and release are increased in a dextran sulfate sodium-induced IBD mouse model. The enzyme appearance in IBD disease was also traced in intestine samples of the dextran sulfate sodium-treated mice and human tissue samples. The results from the IBD diseased animals, treated with fluorescently labeled SM-liposomes, demonstrated that the liposomes were taken up preferentially in the inflamed colon. This uptake efficiency correlated with sphingomyelinase activity.
Collapse
|
7
|
Li Y, Lu Z, Zhang L, Kirkwood CL, Kirkwood KL, Lopes-Virella MF, Huang Y. Inhibition of acid sphingomyelinase by imipramine abolishes the synergy between metabolic syndrome and periodontitis on alveolar bone loss. J Periodontal Res 2022; 57:173-185. [PMID: 34748647 PMCID: PMC8766925 DOI: 10.1111/jre.12951] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/08/2021] [Accepted: 10/26/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND OBJECTIVE Clinical studies have shown that metabolic syndrome (MetS) exacerbates periodontitis. However, the underlying mechanisms remain largely unknown. Since our animal study has shown that high-fat diet-induced MetS exacerbates lipopolysaccharide (LPS)-stimulated periodontitis in mouse model and our in vitro study showed that acid sphingomyelinase (aSMase) plays a key role in the amplification of LPS-triggered pro-inflammatory response by palmitic acid (PA) in macrophages, we tested our hypothesis that inhibitor of aSMase attenuates MetS-exacerbated periodontitis in animal model. Furthermore, to explore the potential underlying mechanisms, we tested our hypothesis that aSMase inhibitor downregulates pro-inflammatory and pro-osteoclastogenic gene expression in macrophages in vitro. MATERIAL AND METHODS We induced MetS and periodontitis in C57BL/6 mice by feeding high-fat diet (HFD) and periodontal injection of A. actinomycetemcomitans LPS, respectively, and treated mice with imipramine, a well-established inhibitor of aSMase. Micro-computed tomography (micro-CT), tartrate-resistant acid phosphatase staining, histological and pathological evaluations as well as cell cultures were performed to evaluate alveolar bone loss, osteoclast formation, periodontal inflammation and pro-inflammatory gene expression. RESULTS Analysis of metabolic parameter showed that while HFD induced MetS by increasing bodyweight, insulin resistance, cholesterol and free fatty acids, imipramine reduced free fatty acids but had no significant effects on other metabolic parameters. MicroCT showed that either MetS or periodontitis significantly reduced bone volume fraction (BVF) of maxilla and the combination of MetS and periodontitis further reduced BVF. However, imipramine increased BVF in mice with both MetS and periodontitis to a level similar to that in mice with periodontitis alone, suggesting that imipramine abolished the synergy between MetS and periodontitis on alveolar bone loss. Consistently, results showed that imipramine inhibited osteoclast formation and periodontal inflammation in mice with both MetS and periodontitis. To elucidate the mechanisms by which imipramine attenuates MetS-exacerbated periodontitis, we showed that imipramine inhibited the upregulation of pro-inflammatory cytokines and transcription factor c-FOS as well as ceramide production by LPS plus PA in macrophages. CONCLUSION This study has shown that imipramine as an inhibitor of aSMase abolishes the synergy between MetS and periodontitis on alveolar bone loss in animal model and inhibits pro-inflammatory and pro-osteoclastogenic gene expression in macrophages in vitro. This study provides the first evidence that aSMase is a potential therapeutic target for MetS-exacerbated periodontitis.
Collapse
Affiliation(s)
- Yanchun Li
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, College of Medicine, Medical University of South Carolina, Charleston, SC 29425
| | - Zhongyang Lu
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, College of Medicine, Medical University of South Carolina, Charleston, SC 29425
| | - Lixia Zhang
- Department of Oral Biology, School of Dental Medicine, University at Buffalo
| | - Cameron L. Kirkwood
- Department of Oral Biology, School of Dental Medicine, University at Buffalo
| | - Keith L. Kirkwood
- Department of Oral Biology, School of Dental Medicine, University at Buffalo,,Department of Head and Neck/Plastic and Reconstructive Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14214
| | - Maria F. Lopes-Virella
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, College of Medicine, Medical University of South Carolina, Charleston, SC 29425,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401
| | - Yan Huang
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, College of Medicine, Medical University of South Carolina, Charleston, SC 29425,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401,Correspondence to Yan Huang, M.D., Ph.D., Ralph H. Johnson Veterans Affairs Medical Center, and Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, 114 Doughty St. Charleston, SC29403, Tel: (843) 789-6824; Fax: (843) 876-5133;
| |
Collapse
|
8
|
Semler A, Hammad S, Lopes-Virella MF, Klein RL, Huang Y. Deoxysphingolipids Upregulate MMP-1, Downregulate TIMP-1, and Induce Cytotoxicity in Human Schwann Cells. Neuromolecular Med 2021; 24:352-362. [PMID: 34853975 DOI: 10.1007/s12017-021-08698-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 11/18/2021] [Indexed: 11/29/2022]
Abstract
Sphingolipids are a heterogeneous class of lipids and essential components of the plasma membrane and plasma lipoproteins. Studies have shown that plasma deoxysphingolipid (DSL), a newly identified sphingolipid class, is increased in diabetic patients and associated with diabetic neuropathy. However, it remains unknown if there is a causal relationship between plasma DSL increase and diabetic neuropathy. Since matrix metalloproteinases (MMPs) play an important role in diabetic neuropathy by degrading extracellular matrix in the peripheral nervous system, we investigated the effect of DSLs on the expression of MMPs and tissue inhibitor of metalloproteinase (TIMPs), and cytotoxicity in human Schwann cells. We quantified protein secretion, gene expression, and collagenase activity, and performed cytotoxicity assays. Results showed that DSLs upregulated MMP-1, downregulated TIMP-1, and induced cytotoxicity in Schwann cells. Furthermore, we quantified DSLs in VLDL, LDL, HDL2, and HDL3 isolated from type 2 diabetes mellitus (T2DM) patients with or without neuropathy. Interestingly, lipidomic analysis showed that only HDL2 isolated from T2DM patients with neuropathy contains significantly higher level of DSLs than that isolated from T2DM patients without neuropathy. Additionally, results showed that HDL2 isolated from T2DM patients with neuropathy was more potent than that isolated from T2DM patients without neuropathy in upregulating MMP-1, downregulating TIMP-1, and stimulating collagenase activity in Schwann cell. Taken together, this study demonstrated for the first time a potential causal relationship between DSLs and diabetic neuropathy and that DSL-containing HDL2 from T2DM patients with neuropathy was more potent than that from T2DM patients without neuropathy in stimulating collagenase activity.
Collapse
Affiliation(s)
- Andrea Semler
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Samar Hammad
- Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina, Charleston, USA
| | - Maria F Lopes-Virella
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, 29401, USA.,Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Richard L Klein
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, 29401, USA.,Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Yan Huang
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, 29401, USA. .,Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
9
|
Fat of the Gut: Epithelial Phospholipids in Inflammatory Bowel Diseases. Int J Mol Sci 2021; 22:ijms222111682. [PMID: 34769112 PMCID: PMC8584226 DOI: 10.3390/ijms222111682] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/20/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022] Open
Abstract
Inflammatory bowel diseases (IBD) comprise a distinct set of clinical symptoms resulting from chronic inflammation within the gastrointestinal (GI) tract. Despite the significant progress in understanding the etiology and development of treatment strategies, IBD remain incurable for thousands of patients. Metabolic deregulation is indicative of IBD, including substantial shifts in lipid metabolism. Recent data showed that changes in some phospholipids are very common in IBD patients. For instance, phosphatidylcholine (PC)/phosphatidylethanolamine (PE) and lysophosphatidylcholine (LPC)/PC ratios are associated with the severity of the inflammatory process. Composition of phospholipids also changes upon IBD towards an increase in arachidonic acid and a decrease in linoleic and a-linolenic acid levels. Moreover, an increase in certain phospholipid metabolites, such as lysophosphatidylcholine, sphingosine-1-phosphate and ceramide, can result in enhanced intestinal inflammation, malignancy, apoptosis or necroptosis. Because some phospholipids are associated with pathogenesis of IBD, they may provide a basis for new strategies to treat IBD. Current attempts are aimed at controlling phospholipid and fatty acid levels through the diet or via pharmacological manipulation of lipid metabolism.
Collapse
|
10
|
Jiang S, Shen X, Xuan S, Yang B, Ruan Q, Cui H, Zhao Z, Jin J. Serum and colon metabolomics study reveals the anti-ulcerative colitis effect of Croton crassifolius Geisel. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 87:153570. [PMID: 34062350 DOI: 10.1016/j.phymed.2021.153570] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/28/2021] [Accepted: 04/12/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Croton crassifolius Geisel (CCG, also known as Ji-Gu-Xiang in Traditional Chinese Medicine), is traditionally prescribed for the therapy of rheumatic arthritis and gastrointestinal ulcer. However, the effect of CCG on ulcerative colitis (UC) has not been investigated. PURPOSE To explore the therapeutic potential and underlying mechanism of CCG extract against UC by colonic and serum metabolomics. METHODS In order to standardize the CCG extract, UPLC-QTOF-MS was used for quantitative and qualitative analysis of the representative terpenoids. C57BL/6J mice were divided into control, Dextran Sulfate Sodium (DSS), mesalazine (100 mg•kg-1), CCG extract (150 and 600 mg•kg-1) groups. The mice were provided 3% DSS dissolved in distilled water ad libitum for 7 days except control group. Weight change, disease activity index (DAI), colon lengths and expression of inflammatory mediators iNOS and COX-2 in colonic tissue were determined. Serum and colon metabolomics using UPLC-QTOF-MS technology coupled with multivariate data analysis were performed to reveal the underlying mechanism. RESULTS Thirty-five terpenoids in CCG were identified by fingerprint, in which ten representative terpenes were quantified. CCG could relieve the weight loss, the degree of bloody stool and ulcer of colon, as well as significantly lowering the expression level of iNOS and COX-2. Metabolomics analysis showed that 25 biomarkers were obviously interfered by CCG treatment and 16 of them were highly correlated with the efficacy of CCG. The analysis of metabolic pathway showed that the anti-UC effect of CCG was associated with the regulation on linoleic acid metabolism, sphingolipid metabolism, α-linolenic acid metabolism, and glycerophospholipids metabolism. CONCLUSIONS The oral administration of CCG significantly alleviated DSS-induced UC symptoms by reducing inflammation and rectifying the metabolic disorder. CCG may provide a new strategy for the management of UC.
Collapse
Affiliation(s)
- Shiqin Jiang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China; School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou China
| | - Xiuting Shen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou China
| | - Shenxin Xuan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou China
| | - Bao Yang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou China
| | - Qingfeng Ruan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou China
| | - Hui Cui
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou China
| | - Zhongxiang Zhao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou China.
| | - Jing Jin
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
11
|
Liu K, Jia B, Zhou L, Xing L, Wu L, Li Y, Lu J, Zhang L, Guan S. Ultraperformance Liquid Chromatography Coupled with Quadrupole Time-of-Flight Mass Spectrometry-Based Metabolomics and Lipidomics Identify Biomarkers for Efficacy Evaluation of Mesalazine in a Dextran Sulfate Sodium-Induced Ulcerative Colitis Mouse Model. J Proteome Res 2020; 20:1371-1381. [PMID: 33356298 DOI: 10.1021/acs.jproteome.0c00757] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This study aims to identify biomarkers for evaluating the therapeutic efficacy of mesalazine on ulcerative colitis by metabolomics and lipidomics. A dextran sulfate sodium-induced mouse model was used. The disease status was assessed by a disease activity index, the TNF-α level of colon was measured by an enzyme-linked immunosorbent assay, and the pathological changes of colon tissue was examined by hematoxylin-eosin staining. Serum metabolomics and lipidomics analysis based on ultraperformance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry were applied to decipher the metabolic profile changes. Multivariate analysis was applied to differentiate the metabolites of controls, models, and mesalazine-treated mice. By the receiver operating characteristic (ROC) analysis, 40 differential metabolites with an area under curve (AUC) >0.80 were screened out between control and model groups. Among them, four potential biomarkers (palmitoyl glucuronide, isobutyrylglycine, PC (20:3 (5Z, 8Z, 11Z)/15:0) and L-arginine) had a signficantly reversed level of peak areas in the mesalazine group, and three of them were closely correlated with mesalazine efficacy by linear regression analysis. Furthermore, metabolic pathway analysis revealed several dysregulated pathways in colitis mice, including glycerophospholipid metabolism, pyrimidine metabolism, linoleic acid metabolism, arginine biosynthesis, etc. This study indicates that serum metabolomics is a useful approach that can noninvasively evaluate the therapeutic effect and provide unique insights into the underlying mechanism of mesalazine.
Collapse
Affiliation(s)
- Kun Liu
- MOE Joint International Research Laboratory of Synthetic Biology and Medicine, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P.R. China
| | - Bingjie Jia
- MOE Joint International Research Laboratory of Synthetic Biology and Medicine, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P.R. China
| | - Liping Zhou
- Evaluation and Monitoring Center of Occupational Health, Guangzhou Twelfth People's Hospital, Guangzhou 510620, P.R. China
| | - Lei Xing
- MOE Joint International Research Laboratory of Synthetic Biology and Medicine, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P.R. China
| | - Lvying Wu
- MOE Joint International Research Laboratory of Synthetic Biology and Medicine, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P.R. China
| | - Yuanyuan Li
- MOE Joint International Research Laboratory of Synthetic Biology and Medicine, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P.R. China
| | - Jungang Lu
- Agilent Technologies (China) Co., Ltd., Guangzhou 510613, P.R. China
| | - Lei Zhang
- MOE Joint International Research Laboratory of Synthetic Biology and Medicine, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P.R. China
| | - Su Guan
- MOE Joint International Research Laboratory of Synthetic Biology and Medicine, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P.R. China
| |
Collapse
|
12
|
Alhouayek M, Ameraoui H, Muccioli GG. Bioactive lipids in inflammatory bowel diseases - From pathophysiological alterations to therapeutic opportunities. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1866:158854. [PMID: 33157277 DOI: 10.1016/j.bbalip.2020.158854] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/16/2020] [Accepted: 10/27/2020] [Indexed: 12/12/2022]
Abstract
Inflammatory bowel diseases (IBDs), such as Crohn's disease and ulcerative colitis, are lifelong diseases that remain challenging to treat. IBDs are characterized by alterations in intestinal barrier function and dysregulation of the innate and adaptive immunity. An increasing number of lipids are found to be important regulators of inflammation and immunity as well as gut physiology. Therefore, the study of lipid mediators in IBDs is expected to improve our understanding of disease pathogenesis and lead to novel therapeutic opportunities. Here, through selected examples - such as fatty acids, specialized proresolving mediators, lysophospholipids, endocannabinoids, and oxysterols - we discuss how lipid signaling is involved in IBD physiopathology and how modulating lipid signaling pathways could affect IBDs.
Collapse
Affiliation(s)
- Mireille Alhouayek
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, 1200 Bruxelles, Belgium.
| | - Hafsa Ameraoui
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, 1200 Bruxelles, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, 1200 Bruxelles, Belgium.
| |
Collapse
|
13
|
Branco-de-Almeida LS, Franco GCN, Castro ML, Vieira MS, Galvão-Moreira LV, Cortelli SC, Anbinder AL, Kawai T, Rosalen PL. Protective effects of desipramine on alveolar bone in experimental periodontitis. J Periodontol 2020; 91:1694-1703. [PMID: 32294250 DOI: 10.1002/jper.19-0569] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 02/21/2020] [Accepted: 02/23/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Desipramine is a tricyclic antidepressant with immune-modulatory activity, whose effects on ligature-induced periodontitis are yet to be investigated. Hence, its actions on alveolar bone resorption, gingival collagen content and key inflammatory mediators were herewith analyzed. METHODS A total of 60 male Wistar rats were randomly assigned into three groups: 1) control: rats without ligature treated with vehicle (saline); 2) ligature: rats with ligature-induced periodontitis treated with vehicle; 3) ligature + desipramine: rats with ligature-induced periodontitis treated with desipramine (20 mg/kg/d in vehicle). Mandibles and gingival tissues were collected 3 or 15 days after ligature insertion (or no ligature insertion for controls) and treatments. Alveolar bone resorption and gingival collagen fibers were histologically analyzed using either HE or picrosirius red staining. Gingival mRNA expressions of interleukin (IL)-1β, inducible nitric oxide synthase (iNOS), cyclooxygenase (COX)-2, matrix metalloproteinase (MMP)-9 and tissue inhibitor of metalloproteinase (TIMP)-1 were obtained through reverse transcription polymerase chain reaction. MMP-9 activity was analyzed by zymography. RESULTS Alveolar bone loss was significantly reduced in the ligature + desipramine group (P < 0.05), whereas gingival collagen degradation was like the ligature group (P > 0.05). Desipramine administration downregulated mRNA expressions of IL-1β, iNOS, COX-2, and TIMP-1 when compared to vehicle alone in the ligature group (P < 0.05). MMP-9 expression and MMP-9/TIMP-1 ratio were similar among rats with ligature-induced periodontitis (P > 0.05); however, MMP-9 activity was lower in the group treated with desipramine (P < 0.05). CONCLUSION Desipramine administration reduced alveolar bone loss as histologically observed, and modulated key bone remodeling and inflammatory mediators in rats with ligature-induced periodontitis.
Collapse
Affiliation(s)
| | - Gilson C N Franco
- Department of General Biology, State University of Ponta Grossa, Ponta Grossa, Paraná, Brazil
| | | | - Mayana S Vieira
- Post Graduate Program in Dentistry, Universidade CEUMA, São Luís, Maranhão, Brazil
| | | | - Sheila C Cortelli
- Nucleus of Periodontal Research, University of Taubaté, Taubaté, São Paulo, Brazil
| | - Ana L Anbinder
- Department of Bioscience and Oral Diagnosis, São Paulo State University, São José dos Campos, São Paulo, Brazil
| | - Toshihisa Kawai
- College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, Florida, USA
| | - Pedro L Rosalen
- Biological Sciences Graduate Program, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| |
Collapse
|
14
|
Liu F, Wang TTY, Tang Q, Xue C, Li RW, Wu VCH. Malvidin 3-Glucoside Modulated Gut Microbial Dysbiosis and Global Metabolome Disrupted in a Murine Colitis Model Induced by Dextran Sulfate Sodium. Mol Nutr Food Res 2019; 63:e1900455. [PMID: 31444937 DOI: 10.1002/mnfr.201900455] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/10/2019] [Indexed: 12/25/2022]
Abstract
SCOPE This study aims to elucidate the mechanisms of the anthocyanin malvidin 3-glucoside (MV) in alleviating gut dysbiosis using a murine colitis model induced by dextran sulfate sodium (DSS). METHODS AND RESULTS The effect of MV on the structure and function of the colon microbiome and microbial metabolism is evaluated using 16S rRNA gene sequencing, global metabolomics, and a network algorithm based on the random-matrix theory. MV ingestion improved histopathological scores and increased IL10 expression in the colon mucosa of colitis mice. While DSS has a profound effect on the gut microbiome and significantly decreases both microbial richness and evenness, MV further reduces evenness but promotes microbial interactions and restores the Firmicutes/Bacteroidetes ratio repressed by DSS. Moreover, MV reduces the abundance of pathogenic bacteria, such as Ruminococcus gnavus, in colitis mice and has a strong modulatory effect on microbial co-occurrence patterns and gut metabolites. In addition, MV reverses several key inflammatory mediators, including sphingolipid metabolites, from elevated levels in DSS colitis mice. As a bioactive ingredient, MV exerts its effect on the gut microbiome in a mechanism that differs from the whole blueberry. CONCLUSION MV ingestion ameliorates intestinal inflammation by modulating colon epithelium integrity, gut microbiome, and key inflammatory mediators.
Collapse
Affiliation(s)
- Fang Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, China.,Produce Safety and Microbiology Research Unit, United States Department of Agriculture, Agriculture Research Service, Albany, CA, 94706, USA
| | - Thomas T Y Wang
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, United States Department of Agriculture, Agriculture Research Service, Beltsville, MD, 20705, USA
| | - Qingjuan Tang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, China
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, China
| | - Robert W Li
- Animal Genomics and Improvement Laboratory, United States Department of Agriculture, Agriculture Research Service, Beltsville, MD, 20705, USA
| | - Vivian C H Wu
- Produce Safety and Microbiology Research Unit, United States Department of Agriculture, Agriculture Research Service, Albany, CA, 94706, USA
| |
Collapse
|
15
|
Suzuki S, Tanaka A, Nakamura H, Murayama T. Knockout of Ceramide Kinase Aggravates Pathological and Lethal Responses in Mice with Experimental Colitis. Biol Pharm Bull 2018; 41:797-805. [DOI: 10.1248/bpb.b18-00051] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Satomi Suzuki
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University
| | - Ai Tanaka
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University
| | - Hiroyuki Nakamura
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University
| | - Toshihiko Murayama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University
| |
Collapse
|
16
|
Ohnishi T, Hashizume C, Taniguchi M, Furumoto H, Han J, Gao R, Kinami S, Kosaka T, Okazaki T. Sphingomyelin synthase 2 deficiency inhibits the induction of murine colitis-associated colon cancer. FASEB J 2017; 31:3816-3830. [PMID: 28522594 DOI: 10.1096/fj.201601225rr] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 04/24/2017] [Indexed: 12/13/2022]
Abstract
Sphingomyelin synthase 2 (SMS2) is the synthetic enzyme of sphingomyelin (SM), which regulates membrane fluidity and microdomain structure. SMS2 plays a role in LPS-induced lung injury and inflammation; however, its role in inflammation-mediated tumorigenesis is unclear. We investigated the effect of SMS2 deficiency on dextran sodium sulfate (DSS)-induced murine colitis and found inhibition of DSS-induced inflammation in SMS2-deficient (SMS2-/-) mice. DSS treatment induced a significant increase in ceramide levels, with a decrease of SM levels in SMS2-/- colon tissue, and demonstrated attenuation of the elevation of both inflammation-related gene expression and proinflammatory cytokines and chemokines, leukocyte infiltration, and MAPK and signal transducer and activator of transcription 3 activation. After undergoing transplantation of wild-type bone marrow, SMS2-/- mice also exhibited inhibition of DSS-induced inflammation in the colon, which suggested that SMS2 deficiency in bone marrow-derived immune cells was not involved in the inhibition of colitis. Finally, in an azoxymethane/DSS-induced cancer model, SMS2 deficiency significantly decreased tumor incidence in the colon. Our results demonstrate that SMS2 deficiency inhibits DSS-induced colitis and subsequent colitis-associated colon cancer via inhibition of colon epithelial cell-mediated inflammation; therefore, inhibition of SMS2 may be a potential therapeutic target for human colitis and colorectal cancer.-Ohnishi, T., Hashizume, C., Taniguchi, M., Furumoto, H., Han, J., Gao, R., Kinami, S., Kosaka, T., Okazaki, T. Sphingomyelin synthase 2 deficiency inhibits the induction of murine colitis-associated colon cancer.
Collapse
Affiliation(s)
- Toshio Ohnishi
- Division of General and Digestive Surgery, Department of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Chieko Hashizume
- Division of Hematology/Immunology, Department of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Makoto Taniguchi
- Department of Life Science, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| | - Hidehiro Furumoto
- Division of Hematology/Immunology, Department of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Jia Han
- Division of Medical Oncology, Department of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Rongfen Gao
- Division of Hematology/Immunology, Department of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Shinichi Kinami
- Division of General and Digestive Surgery, Department of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Takeo Kosaka
- Division of General and Digestive Surgery, Department of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Toshiro Okazaki
- Division of Hematology/Immunology, Department of Medicine, Kanazawa Medical University, Uchinada, Japan;
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| |
Collapse
|
17
|
Wang R, Gu X, Dai W, Ye J, Lu F, Chai Y, Fan G, Gonzalez FJ, Duan G, Qi Y. A lipidomics investigation into the intervention of celastrol in experimental colitis. MOLECULAR BIOSYSTEMS 2017; 12:1436-44. [PMID: 27021137 DOI: 10.1039/c5mb00864f] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Celastrol is well known for its anti-inflammatory and anti-cancer effects. In this study, the efficacy of celastrol against dextran sulfate sodium (DSS)-induced inflammatory bowel disease (IBD) in mice was established and the mechanism was investigated using lipidomics. Celastrol treatment significantly alleviated DSS-induced colitis in mice, as revealed by the body weight, colon length, scores of rectal bleeding and diarrhea, serum TNF-α level, and histological analysis results. Lipidomics analysis based on UPLC/MS revealed characteristic changes in the metabolic profiles of the colitis mice, with altered levels of lipid markers associated with IBD, including LPC18 : 0, LPC18 : 1, LPC18 : 2, sphingomyelin (SM), and increased LPC18 : 0/LPC18 : 1 and LPC18 : 0/LPC18 : 2 ratios. For the celastrol-treated colitis mice, however, levels of the above lipid markers were restored, together with recovered saturated LPC/unsaturated LPC ratios. Accordingly, using GC-MS analysis, increased stearic acid (C18 : 0)/oleic acid (C18 : 1) and stearic acid (C18 : 0)/linoleic acid (C18 : 2) ratios were observed in colitis mice, which were later recovered after celastrol treatment. Quantitative real-time PCR analysis revealed that the liver expression of stearoyl-coenzyme A desaturase 1 (SCD1), the key enzyme controlling the desaturation of saturated fatty acid, was dramatically inhibited in IBD mice, and was obviously recovered after celastrol treatment. These results suggest that the increased saturated LPC/unsaturated LPC (and saturated fatty acid/unsaturated fatty acid) ratios associated with SCD1 down-regulation could be regarded as biomarkers of colitis, and celastrol alleviates DSS-induced colitis partially via up-regulation of SCD1, restoring the altered balance between stearic acid- and oleic acid-derived lipid species, which plays an important role in alleviating colitis. In all, this study provided the scientific basis for further development of celastrol in treating IBD.
Collapse
Affiliation(s)
- Renping Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| | - Xueqin Gu
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| | - Weiquan Dai
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| | - Jun Ye
- Shanghai Zhabei Institute for Food and Drug Control, Shanghai 200436, China
| | - Feng Lu
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| | - Yifeng Chai
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| | - Guorong Fan
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gengli Duan
- Department of Pharmaceutical Analysis, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Yunpeng Qi
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai 200433, China. and Department of Pharmaceutical Analysis, School of Pharmacy, Fudan University, Shanghai 201203, China.
| |
Collapse
|
18
|
Sphingolipids as Mediators in the Crosstalk between Microbiota and Intestinal Cells: Implications for Inflammatory Bowel Disease. Mediators Inflamm 2016; 2016:9890141. [PMID: 27656050 PMCID: PMC5021499 DOI: 10.1155/2016/9890141] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/10/2016] [Accepted: 07/14/2016] [Indexed: 12/21/2022] Open
Abstract
Inflammatory bowel disease (IBD) describes different illnesses characterized by chronic inflammation of the gastrointestinal tract. Although the pathogenic mechanisms leading to IBD are poorly understood, immune system disturbances likely underlie its development. Sphingolipids (SLs) have been identified as important players and promising therapeutic targets to control inflammation in IBD. Interestingly, it seems that microorganisms of the normal gut microbiota and probiotics are involved in sphingolipid function. However, there is a great need to investigate the role of SLs as intermediates in the crosstalk between intestinal immunity and microorganisms. This review focuses on recent investigations that describe some mechanisms involved in the regulation of cytokine profiles by SLs. We also describe the importance of gut microbiota in providing signaling molecules that favor the communication between resident bacteria and intestinal cells. This, in turn, modulates the immune response in the bowel and likely in other peripheral organs. The potential of SLs and gut microbiota as targets or therapeutic agents for IBD is also discussed.
Collapse
|
19
|
Frasch SC, McNamee EN, Kominsky D, Jedlicka P, Jakubzick C, Zemski Berry K, Mack M, Furuta GT, Lee JJ, Henson PM, Colgan SP, Bratton DL. G2A Signaling Dampens Colitic Inflammation via Production of IFN-γ. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:1425-34. [PMID: 27402702 PMCID: PMC4975950 DOI: 10.4049/jimmunol.1600264] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 06/06/2016] [Indexed: 12/19/2022]
Abstract
Proinflammatory consequences have been described for lysophosphatidylcholine, a lipid product of cellular injury, signaling via the G protein-coupled receptor G2A on myeloid and lymphoid inflammatory cells. This prompted the hypothesis that genetic deletion of G2A would limit intestinal inflammation in a mouse model of colitis induced by dextran sodium sulfate. Surprisingly, G2A(-/-) mice exhibited significantly worsened colitis compared with wild-type mice, as demonstrated by disease activity, colon shortening, histology, and elevated IL-6 and IL-5 in colon tissues. Investigation of inflammatory cells recruited to inflamed G2A(-/-) colons showed significantly more TNF-α(+) and Ly6C(hi)MHCII(-) proinflammatory monocytes and eosinophils than in wild-type colons. Both monocytes and eosinophils were pathogenic as their depletion abolished the excess inflammation in G2A(-/-) mice. G2A(-/-) mice also had less IFN-γ in inflamed colon tissues than wild-type mice. Fewer CD4(+) lymphocytes were recruited to inflamed G2A(-/-) colons, and fewer colonic lymphocytes produced IFN-γ upon ex vivo stimulation. Administration of IFN-γ to G2A(-/-) mice during dextran sodium sulfate exposure abolished the excess colitic inflammation and reduced colonic IL-5 and eosinophil numbers to levels seen in wild-type mice. Furthermore, IFN-γ reduced the numbers of TNF-α(+) monocyte and enhanced their maturation from Ly6C(hi)MHCII(-) to Ly6C(int)MHCII(+) Taken together, the data suggest that G2A signaling serves to dampen intestinal inflammation via the production of IFN-γ, which, in turn, enhances monocyte maturation to a less inflammatory program and ultimately reduces eosinophil-induced injury of colonic tissues.
Collapse
Affiliation(s)
| | - Eóin N McNamee
- Mucosal Inflammation Program, Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| | - Douglas Kominsky
- Mucosal Inflammation Program, Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| | - Paul Jedlicka
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| | - Claudia Jakubzick
- Department of Pediatrics, National Jewish Health, Denver, CO 80206; Department of Immunology and Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80206
| | - Karin Zemski Berry
- Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045
| | - Matthias Mack
- Department of Internal Medicine, University of Regensburg, 93042 Regensburg, Germany
| | - Glenn T Furuta
- Digestive Health Institute, Children's Hospital Colorado, Aurora, CO 80045; Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045; and
| | - James J Lee
- Division of Pulmonary Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic in Arizona, Scottsdale, AZ 85259
| | - Peter M Henson
- Department of Pediatrics, National Jewish Health, Denver, CO 80206
| | - Sean P Colgan
- Mucosal Inflammation Program, Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| | - Donna L Bratton
- Department of Pediatrics, National Jewish Health, Denver, CO 80206
| |
Collapse
|
20
|
Wang K, Xu R, Snider AJ, Schrandt J, Li Y, Bialkowska AB, Li M, Zhou J, Hannun YA, Obeid LM, Yang VW, Mao C. Alkaline ceramidase 3 deficiency aggravates colitis and colitis-associated tumorigenesis in mice by hyperactivating the innate immune system. Cell Death Dis 2016; 7:e2124. [PMID: 26938296 PMCID: PMC4823937 DOI: 10.1038/cddis.2016.36] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 01/13/2016] [Accepted: 01/19/2016] [Indexed: 12/16/2022]
Abstract
Increasing studies suggest that ceramides differing in acyl chain length and/or degree of unsaturation have distinct roles in mediating biological responses. However, still much remains unclear about regulation and role of distinct ceramide species in the immune response. Here, we demonstrate that alkaline ceramidase 3 (Acer3) mediates the immune response by regulating the levels of C18:1-ceramide in cells of the innate immune system and that Acer3 deficiency aggravates colitis in a murine model by augmenting the expression of pro-inflammatory cytokines in myeloid and colonic epithelial cells (CECs). According to the NCBI Gene Expression Omnibus (GEO) database, ACER3 is downregulated in immune cells in response to lipopolysaccharides (LPS), a potent inducer of the innate immune response. Consistent with these data, we demonstrated that LPS downregulated both Acer3 mRNA levels and its enzymatic activity while elevating C(18:1)-ceramide, a substrate of Acer3, in murine immune cells or CECs. Knocking out Acer3 enhanced the elevation of C(18:1)-ceramide and the expression of pro-inflammatory cytokines in immune cells and CECs in response to LPS challenge. Similar to Acer3 knockout, treatment with C(18:1)-ceramide, but not C18:0-ceramide, potentiated LPS-induced expression of pro-inflammatory cytokines in immune cells. In the mouse model of dextran sulfate sodium-induced colitis, Acer3 deficiency augmented colitis-associated elevation of colonic C(18:1)-ceramide and pro-inflammatory cytokines. Acer3 deficiency aggravated diarrhea, rectal bleeding, weight loss and mortality. Pathological analyses revealed that Acer3 deficiency augmented colonic shortening, immune cell infiltration, colonic epithelial damage and systemic inflammation. Acer3 deficiency also aggravated colonic dysplasia in a mouse model of colitis-associated colorectal cancer. Taken together, these results suggest that Acer3 has an important anti-inflammatory role by suppressing cellular or tissue C(18:1)-ceramide, a potent pro-inflammatory bioactive lipid and that dysregulation of ACER3 and C(18:1)-ceramide may contribute to the pathogenesis of inflammatory diseases including cancer.
Collapse
Affiliation(s)
- K Wang
- Department of Medicine, State University of New York at Stony Brook University, Stony Brook, NY, USA
- Stony Brook Cancer Center, Stony Brook, NY, USA
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - R Xu
- Department of Medicine, State University of New York at Stony Brook University, Stony Brook, NY, USA
- Stony Brook Cancer Center, Stony Brook, NY, USA
| | - A J Snider
- Department of Medicine, State University of New York at Stony Brook University, Stony Brook, NY, USA
- Stony Brook Cancer Center, Stony Brook, NY, USA
- Northport Veterans Affairs Medical Center, Northport, NY, USA
| | - J Schrandt
- Department of Medicine, State University of New York at Stony Brook University, Stony Brook, NY, USA
| | - Y Li
- Department of Medicine, State University of New York at Stony Brook University, Stony Brook, NY, USA
- Stony Brook Cancer Center, Stony Brook, NY, USA
| | - A B Bialkowska
- Department of Medicine, State University of New York at Stony Brook University, Stony Brook, NY, USA
| | - M Li
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - J Zhou
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Y A Hannun
- Department of Medicine, State University of New York at Stony Brook University, Stony Brook, NY, USA
- Stony Brook Cancer Center, Stony Brook, NY, USA
| | - L M Obeid
- Department of Medicine, State University of New York at Stony Brook University, Stony Brook, NY, USA
- Stony Brook Cancer Center, Stony Brook, NY, USA
- Northport Veterans Affairs Medical Center, Northport, NY, USA
| | - V W Yang
- Department of Medicine, State University of New York at Stony Brook University, Stony Brook, NY, USA
| | - C Mao
- Department of Medicine, State University of New York at Stony Brook University, Stony Brook, NY, USA
- Stony Brook Cancer Center, Stony Brook, NY, USA
| |
Collapse
|
21
|
Abstract
Acid sphingomyelinase (ASM), a lipid hydrolase enzyme, has the potential to modulate various cellular activation responses via the generation of ceramide and by interaction with cellular receptors. We have hypothesized that ASM modulates CD4+ T-cell receptor activation and impacts immune responses. We first observed interactions of ASM with the intracellular domains of both CD3 and CD28. ASM further mediates T-cell proliferation after anti-CD3/CD28 antibody stimulation and alters CD4+ T-cell activation signals by generating ceramide. We noted that various pharmacological inhibitors of ASM or knockdown of ASM using small hairpin RNA inhibit CD3/CD28-mediated CD4+ T-cell proliferation and activation. Furthermore, such blockade of ASM bioactivity by biochemical inhibitors and/or molecular-targeted knockdown of ASM broadly abrogate T-helper cell responses. In conclusion, we detail immune, pivotal roles of ASM in adaptive immune T-cell responses, and propose that these pathways might provide novel targets for the therapy of autoimmune and inflammatory diseases.
Collapse
|
22
|
Bai A, Moss A, Kokkotou E, Usheva A, Sun X, Cheifetz A, Zheng Y, Longhi MS, Gao W, Wu Y, Robson SC. CD39 and CD161 modulate Th17 responses in Crohn's disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:3366-3377. [PMID: 25172498 PMCID: PMC4170017 DOI: 10.4049/jimmunol.1400346] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CD39 (ENTPD1) is expressed by subsets of pathogenic human CD4(+) T cells, such as Th17 cells. These Th17 cells are considered important in intestinal inflammation, such as seen in Crohn's disease (CD). Recently, CD161 (NKR-P1A) was shown to be a phenotypic marker of human Th17 cells. In this study, we report that coexpression of CD161 and CD39 not only identifies these cells but also promotes Th17 generation. We note that human CD4(+)CD39(+)CD161(+) T cells can be induced under stimulatory conditions that promote Th17 in vitro. Furthermore, CD4(+)CD39(+)CD161(+) cells purified from blood and intestinal tissues, from both healthy controls and patients with CD, are of the Th17 phenotype and exhibit proinflammatory functions. CD39 is coexpressed with CD161, and this association augments acid sphingomyelinase (ASM) activity upon stimulation of CD4(+) T cells. These pathways regulate mammalian target of rapamycin and STAT3 signaling to drive the Th17 phenotype. Inhibition of ASM activity by pharmacological blockers or knockdown of ASM abrogates STAT3 signaling, thereby limiting IL-17 production in CD4(+) T cells obtained from both controls and patients with active CD. Increased levels of CD39(+)CD161(+) CD4(+) T cells in blood or lamina propria are noted in patients with CD, and levels directly correlate with clinical disease activity. Hence, coexpression of CD39 and CD161 by CD4(+) T cells might serve as a biomarker to monitor Th17 responsiveness. Collectively, CD39 and CD161 modulate human Th17 responses in CD through alterations in purinergic nucleotide-mediated responses and ASM catalytic bioactivity, respectively.
Collapse
Affiliation(s)
- Aiping Bai
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard University, Boston, MA 02215
| | - Alan Moss
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard University, Boston, MA 02215
| | - Efi Kokkotou
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard University, Boston, MA 02215
| | - Anny Usheva
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard University, Boston, MA 02215
| | - Xiaofeng Sun
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard University, Boston, MA 02215
| | - Adam Cheifetz
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard University, Boston, MA 02215
| | - Yi Zheng
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard University, Boston, MA 02215; and
| | - Maria Serena Longhi
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard University, Boston, MA 02215
| | - Wenda Gao
- Antagen Institute for Biomedical Research, Boston, MA 02118
| | - Yan Wu
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard University, Boston, MA 02215
| | - Simon C Robson
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard University, Boston, MA 02215;
| |
Collapse
|
23
|
Beckmann N, Sharma D, Gulbins E, Becker KA, Edelmann B. Inhibition of acid sphingomyelinase by tricyclic antidepressants and analogons. Front Physiol 2014; 5:331. [PMID: 25228885 PMCID: PMC4151525 DOI: 10.3389/fphys.2014.00331] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 08/12/2014] [Indexed: 11/13/2022] Open
Abstract
Amitriptyline, a tricyclic antidepressant, has been used in the clinic to treat a number of disorders, in particular major depression and neuropathic pain. In the 1970s the ability of tricyclic antidepressants to inhibit acid sphingomyelinase (ASM) was discovered. The enzyme ASM catalyzes the hydrolysis of sphingomyelin to ceramide. ASM and ceramide were shown to play a crucial role in a wide range of diseases, including cancer, cystic fibrosis, diabetes, Alzheimer's disease, and major depression, as well as viral (e.g., measles virus) and bacterial (e.g., Staphylococcus aureus, Pseudomonas aeruginosa) infections. Ceramide molecules may act in these diseases by the alteration of membrane biophysics, the self-association of ceramide molecules within the cell membrane and the ultimate formation of larger ceramide-enriched membrane domains/platforms. These domains were shown to serve the clustering of certain receptors such as CD95 and may also act in the above named diseases. The potential to block the generation of ceramide by inhibiting the ASM has opened up new therapeutic approaches for the treatment of these conditions. Since amitriptyline is one of the longest used clinical drugs and side effects are well studied, it could potentially become a cheap and easily accessible medication for patients suffering from these diseases. In this review, we aim to provide an overview of current in vitro and in vivo studies and clinical trials utilizing amitriptyline to inhibit ASM and contemplate possible future applications of the drug.
Collapse
Affiliation(s)
- Nadine Beckmann
- Department of Molecular Biology, Institute of Molecular Biology, University of Duisburg-Essen Essen, Germany
| | - Deepa Sharma
- Department of Molecular Biology, Institute of Molecular Biology, University of Duisburg-Essen Essen, Germany
| | - Erich Gulbins
- Department of Molecular Biology, Institute of Molecular Biology, University of Duisburg-Essen Essen, Germany
| | - Katrin Anne Becker
- Department of Molecular Biology, Institute of Molecular Biology, University of Duisburg-Essen Essen, Germany
| | - Bärbel Edelmann
- Department of Molecular Biology, Institute of Molecular Biology, University of Duisburg-Essen Essen, Germany
| |
Collapse
|
24
|
Qi Y, Jiang C, Tanaka N, Krausz KW, Brocker CN, Fang ZZ, Bredell BX, Shah YM, Gonzalez FJ. PPARα-dependent exacerbation of experimental colitis by the hypolipidemic drug fenofibrate. Am J Physiol Gastrointest Liver Physiol 2014; 307:G564-G573. [PMID: 25035112 PMCID: PMC4154119 DOI: 10.1152/ajpgi.00153.2014] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 07/14/2014] [Indexed: 02/07/2023]
Abstract
Fibrates, such as fenofibrate, are peroxisome proliferator-activated receptor-α (PPARα) agonists and have been used for several decades as hypolipidemic agents in the clinic. However, contradictory observations exist on the role of fibrates in host response to acute inflammation, with unclear mechanisms. The role of PPARα in colitis was assessed using fenofibrate and Ppara-null mice. Wild-type or Ppara-null mice were subjected to acute colitis under three distinct protocols, dextran sulfate sodium, trinitrobenzenesulfonic acid, and Salmonella Typhi. Serum and colon lipidomics were analyzed to characterize the metabolic profiles by ultra-performance liquid chromatography-coupled with electrospray ionization quadrupole time-of-flight mass spectrometry. Messenger RNAs of PPARα target genes and genes involved in inflammation were determined by qunatitative PCR analysis. Fenofibrate treatment exacerbated inflammation and tissue injury in acute colitis, and this was dependent on PPARα activation. Lipidomics analysis revealed that bioactive sphingolipids, including sphingomyelins (SM) and ceramides, were significantly increased in the colitis group compared with the control group; this was further potentiated following fenofibrate treatment. In the colon, fenofibrate did not reduce the markedly increased expression of mRNA encoding TNFα found in the acute colitis model, while it decreased hydrolysis and increased synthesis of SM, upregulated RIPK3-dependent necrosis, and elevated mitochondrial fatty acid β-oxidation, which were possibly related to the exacerbated colitis.
Collapse
Affiliation(s)
- Yunpeng Qi
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China; Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Changtao Jiang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Naoki Tanaka
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Chad N Brocker
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Zhong-Ze Fang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Bryce X Bredell
- Department of Molecular and Integrative Physiology, Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; and
| |
Collapse
|
25
|
Tibboel J, Keijzer R, Reiss I, de Jongste JC, Post M. Intravenous and intratracheal mesenchymal stromal cell injection in a mouse model of pulmonary emphysema. COPD 2014; 11:310-8. [PMID: 24295402 PMCID: PMC4046870 DOI: 10.3109/15412555.2013.854322] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The aim of this study was to characterize the evolution of lung function and -structure in elastase-induced emphysema in adult mice and the effect of mesenchymal stromal cell (MSC) administration on these parameters. Adult mice were treated with intratracheal (4.8 units/100 g bodyweight) elastase to induce emphysema. MSCs were administered intratracheally or intravenously, before or after elastase injection. Lung function measurements, histological and morphometric analysis of lung tissue were performed at 3 weeks, 5 and 10 months after elastase and at 19, 20 and 21 days following MSC administration. Elastase-treated mice showed increased dynamic compliance and total lung capacity, and reduced tissue-specific elastance and forced expiratory flows at 3 weeks after elastase, which persisted during 10 months follow-up. Histology showed heterogeneous alveolar destruction which also persisted during long-term follow-up. Jugular vein injection of MSCs before elastase inhibited deterioration of lung function but had no effects on histology. Intratracheal MSC treatment did not modify lung function or histology. In conclusion, elastase-treated mice displayed persistent characteristics of pulmonary emphysema. Jugular vein injection of MSCs prior to elastase reduced deterioration of lung function. Intratracheal MSC treatment had no effect on lung function or histology.
Collapse
Affiliation(s)
- Jeroen Tibboel
- Department of Physiology and Experimental Medicine, Hospital for Sick Children,Toronto,Canada
- Department of Pediatrics, Erasmus University Medical Center –Sophia Children’s Hospital,Rotterdam,the Netherlands
| | - Richard Keijzer
- Department of Pediatric General Surgery, Manitoba Institute of Child Health,Winnipeg,Canada
| | - Irwin Reiss
- Department of Pediatrics, Erasmus University Medical Center –Sophia Children’s Hospital,Rotterdam,the Netherlands
| | - Johan C. de Jongste
- Department of Pediatrics, Erasmus University Medical Center –Sophia Children’s Hospital,Rotterdam,the Netherlands
| | - Martin Post
- Department of Physiology and Experimental Medicine, Hospital for Sick Children,Toronto,Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto,Toronto,Canada
| |
Collapse
|
26
|
Tibboel J, Reiss I, de Jongste JC, Post M. Sphingolipids in lung growth and repair. Chest 2014; 145:120-128. [PMID: 24394822 DOI: 10.1378/chest.13-0967] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Sphingolipids comprise a class of bioactive lipids that are involved in a variety of pathophysiologic processes, including cell death and survival. Ceramide and sphingosine-1-phosphate (S1P) form the center of sphingolipid metabolism and determine proapoptotic and antiapoptotic balance. Findings in animal models suggest a possible pathophysiologic role of ceramide and S1P in COPD, cystic fibrosis, and asthma. Sphingolipid research is now focusing on the role of ceramides during lung inflammation and its regulation by sphingomyelinases. Recently, sphingolipids have been shown to play a role in the pathogenesis of bronchopulmonary dysplasia (BPD). Ceramide upregulation was linked with vascular endothelial growth factor suppression and decreased surfactant protein B levels, pathways important for the development of BPD. In a murine model of BPD, intervention with an S1P analog had a favorable effect on histologic abnormalities and ceramide levels. Ceramides and S1P also regulate endothelial permeability through cortical actin cytoskeletal rearrangement, which is relevant for the pathogenesis of ARDS. On the basis of these observations, the feasibility of pharmacologic intervention in the sphingolipid pathway to influence disease development and progression is presently explored, with promising early results. The prospect of new strategies to prevent and repair lung disease by interfering with sphingolipid metabolism is exciting and could potentially reduce morbidity and mortality in patients with severe lung disorders.
Collapse
Affiliation(s)
- Jeroen Tibboel
- Department of Physiology and Experimental Medicine, The Hospital for Sick Children, Toronto, ON, Canada; Department of Pediatrics, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Irwin Reiss
- Department of Pediatrics, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Johan C de Jongste
- Department of Pediatrics, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Martin Post
- Department of Physiology and Experimental Medicine, The Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
27
|
Leucht K, Fischbeck A, Caj M, Liebisch G, Hartlieb E, Benes P, Fried M, Humpf HU, Rogler G, Hausmann M. Sphingomyelin and phosphatidylcholine contrarily affect the induction of apoptosis in intestinal epithelial cells. Mol Nutr Food Res 2013; 58:782-98. [PMID: 24142587 DOI: 10.1002/mnfr.201300369] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 08/14/2013] [Accepted: 08/20/2013] [Indexed: 12/22/2022]
Abstract
SCOPE The major alimentary sources for the plasma membrane lipid sphingomyelin (SM) are dairy products, eggs, and meat. We recently reported that the SM metabolite ceramide induces cathepsin D mediated apoptosis in murine intestinal epithelial cells (IECs) and increases inflammation in acute colitis. We investigated the impact of SM and phosphatidylcholine on apoptosis in human IECs and point out BH3-interacting death agonist (BID) as link between cathepsin D and apoptosis. METHODS AND RESULTS HT-29 and isolated human IECs were stimulated with SM or phosphatidylcholine. SM treatment resulted in increased apoptosis. Phosphatidylcholine showed contrary effects. Western revealed higher amounts of cathepsin D and BID activation upon lipid stimulation. Western blotting revealed BID activation through SM in both an induced and a spontaneous mouse model of colitis. CONCLUSION Dietary phospholipids may induce or abolish apoptosis in IECs and seem to play a role in the pathogenesis of inflammatory bowel diseases. This nutritional factor might be considered when evaluating the pathogenesis of inflammatory bowel diseases. Effects of SMase- and SM treatment on inflammation in dextran sulfate sodium induced animal models of colitis and in vitro experiments are discussed as controversial. Variable sources of SM, feeding techniques, and mouse strains might play a role.
Collapse
Affiliation(s)
- Katharina Leucht
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University Hospital Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Lee YH, Kim SH, Kim Y, Lim Y, Ha K, Shin SY. Inhibitory effect of the antidepressant imipramine on NF-κB-dependent CXCL1 expression in TNFα-exposed astrocytes. Int Immunopharmacol 2012; 12:547-55. [PMID: 22326584 DOI: 10.1016/j.intimp.2012.01.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Revised: 01/10/2012] [Accepted: 01/25/2012] [Indexed: 11/19/2022]
Abstract
Neuroinflammation is associated with the pathophysiology of various neurodegenerative diseases. Emerging evidence indicates that imipramine, a tricyclic antidepressant commonly used in depressive disorders, exhibits neuroprotective activity partly through anti-inflammatory effects. However, the molecular mechanisms underlying imipramine-mediated anti-inflammatory response are poorly understood. In this study, rat primary cultured astrocytes were used to elucidate the effect of the imipramine on TNFα-induced inflammatory responses. The results clearly demonstrated that imipramine reduced TNFα-induced CXCL1 expression through suppression of NF-κB-dependent CXCL1 promoter activity in primary astrocytes. In addition, we found that imipramine suppressed TNFα-induced phosphorylation of inhibitor of κBα (IκBα) and p65/RelA nuclear factor-κB (NF-κB), as well as the nuclear translocation of p65/RelA in primary cultured astrocytes. Chemotaxis assay demonstrated that astrocyte-derived CXCL1 contributed to migration of BV2 microglial cells toward astrocytes. This response was significantly blocked by treatment of astrocytes with imipramine or NF-κB inhibitor BAY11-7082. This study indicates that the antidepressant imipramine inhibits TNFα-induced CXCL1 expression via down-regulation of NF-κB signaling pathway in astrocytes and suggests that imipramine has a potential as an anti-inflammatory drug.
Collapse
Affiliation(s)
- Young Han Lee
- Department of Biomedical Science and Technology, Research Center for Transcription Control, SMART Institute of Advanced Biomedical Science, Konkuk University, Seoul 143-701, Republic of Korea
| | | | | | | | | | | |
Collapse
|
29
|
Shores DR, Binion DG, Freeman BA, Baker PR. New insights into the role of fatty acids in the pathogenesis and resolution of inflammatory bowel disease. Inflamm Bowel Dis 2011; 17:2192-204. [PMID: 21910181 PMCID: PMC4100336 DOI: 10.1002/ibd.21560] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Accepted: 10/05/2010] [Indexed: 12/12/2022]
Abstract
Dietary and endogenously modified lipids modulate inflammation by functioning as intra- and intercellular signaling molecules. Proinflammatory lipid mediators such as the eicosanoids compete against the signaling actions of newly discovered modified fatty acids that act to resolve inflammation. In inflammatory bowel disease, multiple aberrancies in lipid metabolism have been discovered, which shed further light on the pathogenesis of intestinal inflammation. Mechanisms by which lipids modulate inflammation, abnormalities of lipid metabolism in the setting of inflammatory bowel disease, and potential therapeutic application of lipid derivatives in this setting are discussed.
Collapse
Affiliation(s)
- Darla R. Shores
- Division of Pediatric Gastroenterology, Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - David G. Binion
- Division of Gastroenterology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Bruce A. Freeman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Paul R.S. Baker
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
30
|
Kolter T. A view on sphingolipids and disease. Chem Phys Lipids 2011; 164:590-606. [PMID: 21570958 DOI: 10.1016/j.chemphyslip.2011.04.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 04/26/2011] [Accepted: 04/28/2011] [Indexed: 12/27/2022]
Abstract
Sphingolipid and glycosphingolipid levels and expression of sphingolipid metabolizing enzymes are altered in a variety of diseases or in response to drug treatment. Inherited defects of enzymes and other proteins required for the lysosomal degradation of these lipids lead to human sphingolipidoses. Also genetic defects that affect sphingolipid biosynthesis are known. Although the molecular details are often far from clear, (glyco)sphingolipids have been implicated to play a role in atherosclerosis, insulin resistance, cancer, and infections by pathogens. More general aspects of selected diseases are discussed.
Collapse
Affiliation(s)
- Thomas Kolter
- LiMES-Laboratory of Lipid Biochemistry, Kekulé-Institut für Organische Chemie und Biochemie der Universität, Bonn, Germany.
| |
Collapse
|
31
|
Isik F, Tunali Akbay T, Yarat A, Genc Z, Pisiriciler R, Caliskan-Ak E, Cetinel S, Altıntas A, Sener G. Protective effects of black cumin (Nigella sativa) oil on TNBS-induced experimental colitis in rats. Dig Dis Sci 2011; 56:721-30. [PMID: 20658190 DOI: 10.1007/s10620-010-1333-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Accepted: 06/18/2010] [Indexed: 12/31/2022]
Abstract
BACKGROUND The pathogenesis and treatment of ulcerative colitis remain poorly understood. The aim of the present study is to investigate the effects of black cumin (Nigella sativa) oil on rats with colitis. METHODS Experimental colitis was induced with 1 mL trinitrobenzene sulfonic acid (TNBS) in 40% ethanol by intracolonic administration with 8-cm-long cannula under ether anesthesia to rats in colitis group and colitis + black cumin oil group. Rats in the control group were given saline at the same volume by intracolonic administration. Black cumin oil (BCO, Origo "100% natural Black Cumin Seed Oil," Turkey) was given to colitis + black cumin oil group by oral administration during 3 days, 5 min after colitis induction. Saline was given to control and colitis groups at the same volume by oral administration. At the end of the experiment, macroscopic lesions were scored and the degree of oxidant damage was evaluated by colonic total protein, sialic acid, malondialdehyde, and glutathione levels, collagen content, and tissue factor, superoxide dismutase, and myeloperoxidase activities. Tissues were also examined by histological and cytological analysis. Proinflammatory cytokines [tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1β, and IL-6], lactate dehydrogenase activity, and triglyceride and cholesterol levels were analyzed in blood samples. RESULTS We found that black cumin oil decreased the proinflammatory cytokines, lactate dehydrogenase, triglyceride, and cholesterol, which were increased in colitis. CONCLUSIONS BCO, by preventing inflammatory status in the blood, partly protected colonic tissue against experimental ulcerative colitis.
Collapse
Affiliation(s)
- F Isik
- Faculty of Dentistry, Department of Biochemistry, Marmara University, Nisantası, 34365, Istanbul, Turkey.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Mazzei JC, Zhou H, Brayfield BP, Hontecillas R, Bassaganya-Riera J, Schmelz EM. Suppression of intestinal inflammation and inflammation-driven colon cancer in mice by dietary sphingomyelin: importance of peroxisome proliferator-activated receptor γ expression. J Nutr Biochem 2011; 22:1160-71. [PMID: 21295961 DOI: 10.1016/j.jnutbio.2010.09.017] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Revised: 09/17/2010] [Accepted: 09/27/2010] [Indexed: 12/23/2022]
Abstract
Inflammation of the gastrointestinal tract increases the risk of developing colon cancer especially in younger adults. Dietary compounds are not only associated with the etiology of inflammation and colon cancer but also in their prevention. Sphingolipid metabolites have been shown to play a role in the initiation and perpetuation of inflammatory responses. In the present study, we investigated the suppression of dextran sodium sulfate-induced colitis and azoxymethane-induced colon cancer by dietary sphingomyelin (SM) in mice that lack functional peroxisome proliferator-activated receptor γ (PPAR-γ) in intestinal epithelial and immune cells. Dietary SM decreased disease activity and colonic inflammatory lesions in mice of both genotypes but more efficiently in mice expressing PPAR-γ. The increased survival and suppression of tumor formation in the SM-fed mice appeared to be independent of PPAR-γ expression in immune and epithelial cells. Using a real-time polymerase chain reaction array, we detected an up-regulation in genes involved in Th1 (interferon γ) and Th17 (interleukin [IL]-17 and IL-23) responses despite the reduced inflammation scores. However, the genes involved in Th2 (IL-4, IL-13 and IL-13ra2) and Treg (IL-10rb) anti-inflammatory responses were up-regulated in a PPAR-γ-dependent manner. In line with the PPAR-γ dependency of our in vivo findings, treatment of RAW macrophages with sphingosine increased the PPAR-γ reporter activity. In conclusion, dietary SM modulated inflammatory responses at the early stages of the disease by activating PPAR-γ, but its anticarcinogenic effects followed a PPAR-γ-independent pattern.
Collapse
Affiliation(s)
- Joseph C Mazzei
- Department of Human Nutrition, Foods and Exercise, Virginia Bioinformatics Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | | | | | | | | | | |
Collapse
|
33
|
Brulet M, Seyer A, Edelman A, Brunelle A, Fritsch J, Ollero M, Laprévote O. Lipid mapping of colonic mucosa by cluster TOF-SIMS imaging and multivariate analysis in cftr knockout mice. J Lipid Res 2010; 51:3034-45. [PMID: 20616379 DOI: 10.1194/jlr.m008870] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The cftr knockout mouse model of cystic fibrosis (CF) shows intestinal obstruction; malabsorption and inflammation; and a fatty acid imbalance in intestinal mucosa. We performed a lipid mapping of colon sections from CF and control (WT) mice by cluster time of flight secondary-ion mass spectrometry (TOF-SIMS) imaging to localize lipid alterations. Data were processed either manually or by multivariate statistical methods. TOF-SIMS analysis showed a particular localization for cholesteryl sulfate at the epithelial border, C16:1 fatty acid in Lieberkühn glands, and C18:0 fatty acid in lamina propria and submucosa. Significant increases in vitamin E (vE) and C16:0 fatty acid in the epithelial border of CF colon were detected. Principal component analysis (PCA) and partitioning clustering allowed us to characterize different structural regions of colonic mucosa according to variations in C14:0, C16:0, C16:1, C18:0, C18:1, C18:2, C20:3, C20:4, and C22:6 fatty acids; phosphatidylethanolamine, phosphatidylcholine, and phosphatidylinositol glycerolipids; cholesterol; vitamin E; and cholesteryl sulfate. PCA on spectra from Lieberkühn glands led to separation of CF and WT individuals. This study shows for the first time the spatial distribution of lipids in colonic mucosa and suggests TOF-SIMS plus multivariate analyses as a powerful tool to investigate disease-related tissue spatial lipid signatures.
Collapse
Affiliation(s)
- Marc Brulet
- Centre de Recherche de Gif, Institut de Chimie des Substances Naturelles, CNRS, Gif-sur-Yvette, France
| | | | | | | | | | | | | |
Collapse
|