1
|
Li B, Chen J, Ou X, Liu X, Xu Z, Xiang X, Yang Y, Wang Q. In-depth multiomic characterization of the effects of obesity in high-fat diet-fed mice. FEBS Open Bio 2024; 14:771-792. [PMID: 38479983 PMCID: PMC11073502 DOI: 10.1002/2211-5463.13788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/08/2023] [Accepted: 03/01/2024] [Indexed: 05/07/2024] Open
Abstract
High-fat diet (HFD)-fed mice have been widely used in the clinical investigation of obesity. However, the long-term effect of HFD on gut microbiota and metabolites, plasma and liver metabolomics, colonic and liver transcriptomics remain largely unknown. In this study, 6-week-old C57BL/6J male mice fed with HFD for 14 weeks showed increased obesity-related indexes including alanine aminotransferase, aspartate aminotransferase, total cholesterol, total triglyceride, free fatty acids, lipopolysaccharides, IL-6, and TNFα. Furthermore, microbial diversity and richness were also significantly decreased. In the colon, genes involved in tryptophan metabolism, PPAR signaling pathway, cholesterol metabolism, and lipid localization and transport, were upregulated. While in the liver, MAPK signaling and unsaturated fatty acid biosynthesis were upregulated. Metabolomic analyses revealed decreased levels of glycerophospholipids and fatty acyl, but increased amino acids, coenzymes and vitamins, and organic acids in the colon, suggesting high absorption of oxidized lipids, while acyl-carnitine, lysophosphatidylcholine, lysophosphatidylethanolamine, and oxidized lipids were reduced in the liver, suggesting a more active lipid metabolism. Finally, correlation analyses revealed a positive correlation between gut microbiota and metabolites and the expression of genes associated with lipid localization, absorption, and transport in the colon, and nutrients and energy metabolism in the liver. Taken together, our results provide a comprehensive characterization of long-term HFD-induced obesity in mice.
Collapse
Affiliation(s)
- Boping Li
- Gansu Key Laboratory of Protection and Utilization for Biological Resources and Ecological Restoration in Longdong, Longdong University, Qingyang, China
- College of Medicine, Longdong University, Qingyang, China
| | - Juanjuan Chen
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Xiaobin Ou
- Gansu Key Laboratory of Protection and Utilization for Biological Resources and Ecological Restoration in Longdong, Longdong University, Qingyang, China
- College of Life Sciences and Technology, Longdong University, Qingyang, China
| | - Xiuli Liu
- Gansu Key Laboratory of Protection and Utilization for Biological Resources and Ecological Restoration in Longdong, Longdong University, Qingyang, China
- College of Life Sciences and Technology, Longdong University, Qingyang, China
| | - Zaoxu Xu
- Gansu Key Laboratory of Protection and Utilization for Biological Resources and Ecological Restoration in Longdong, Longdong University, Qingyang, China
- College of Life Sciences and Technology, Longdong University, Qingyang, China
| | - Xuesong Xiang
- Element Nutrition of National Health Commission, National Institute of Nutrition and Health, China CDC, Beijing, China
| | - Yan Yang
- Department of Endocrinology and Metabolism, Lanzhou University Second Hospital, Lanzhou, China
| | - Qi Wang
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
2
|
Wang W, Li W, Duan H, Xu C, Tian X, Li S, Tan Q, Zhang D. Mediation by DNA methylation on the association of BMI and serum uric acid in Chinese monozygotic twins. Gene 2023; 850:146957. [PMID: 36243213 DOI: 10.1016/j.gene.2022.146957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 09/06/2022] [Accepted: 10/03/2022] [Indexed: 02/13/2023]
Abstract
Obesity is an established risk factor for hyperuricemia, but the mechanisms are only partially understood. We examined whether BMI-related DNA methylation (DNAm) variation would mediate the association of BMI with serum uric acid (SUA). We first conducted an epigenome-wide association analysis (EWAS) in 64 monozygotic twin pairs to detect BMI-related DNAm variation and then evaluated the mediated effect of DNAm using mediation analysis. Ontology enrichments analysis was performed for CpGs using GREAT tool. The genes where the candidate CpG mediators mapped were validated using gene expression data. BMI was positively associated with log10 transformed SUA level (β = 0.01, P < 0.001). The association between BMI and DNAm of 138 CpGs reached P < 1 × 10-4 level. Twenty BMI-related differentially methylated regions within MAP2K2, POU4F2, AGAP2, MRGPRE, ADM5, and NKX1-1 were found. Of the 138 CpGs, 4 within VENTX (involved in cellular responses to stress pathway), SMOC2 (enable calcium ion binding), and FSCN2 (a member of fascin protein family) mediated the association between BMI and SUA, with a mediating effect of 0.002-μmol/L lower log10 transformed SUA levels and a proportion of 18.89 %-24.92 % negative mediating effect. BMI × DNAm interactions on SUA were observed for 2 CpGs within VENTX. The gene expression level of VENTX was also negatively associated with SUA level. BMI-related DNAm variation may partially mediate the association of BMI with SUA.
Collapse
Affiliation(s)
- Weijing Wang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, Shandong, China.
| | - Weilong Li
- Population Research Unit, Faculty of Social Sciences, University of Helsinki, Finland.
| | - Haiping Duan
- Qingdao Municipal Center for Disease Control and Prevention/Qingdao Institute of Preventive Medicine, Qingdao, Shandong, China
| | - Chunsheng Xu
- Qingdao Municipal Center for Disease Control and Prevention/Qingdao Institute of Preventive Medicine, Qingdao, Shandong, China
| | - Xiaocao Tian
- Qingdao Municipal Center for Disease Control and Prevention/Qingdao Institute of Preventive Medicine, Qingdao, Shandong, China
| | - Shuxia Li
- Epidemiology and Biostatistics, Department of Public Health, University of Southern Denmark, Odense, Denmark.
| | - Qihua Tan
- Epidemiology and Biostatistics, Department of Public Health, University of Southern Denmark, Odense, Denmark.
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
3
|
Wang YX, Lin C, Cui LJ, Deng TZ, Li QM, Chen FY, Miao XP. Mechanism of M2 macrophage-derived extracellular vesicles carrying lncRNA MEG3 in inflammatory responses in ulcerative colitis. Bioengineered 2021; 12:12722-12739. [PMID: 34895044 PMCID: PMC8810016 DOI: 10.1080/21655979.2021.2010368] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/19/2021] [Accepted: 11/20/2021] [Indexed: 12/20/2022] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory disease of the colon. M2 macrophages possess certain anti-inflammation activity. Accordingly, the current study set out to investigate the potential mechanism of M2 macrophage-derived extracellular vesicles (M2-EVs) in UC inflammation. Firstly, mouse peritoneal macrophages were induced to M2 phenotype, and M2-EVs were isolated. , the murine model of UC was established, and the length and weight of the colon, disease activity index (DAI), apoptosis, and inflammatory response of UC mice were measured. Young adult mouse colon (YAMC) cells were induced with the help of lipopolysaccharide. LncRNA maternally expressed 3 (LncRNA MEG3), miR-20b-5p, and cAMP responsive element binding protein 1 (CREB1) expression patterns were detected in UC models. In addition, we analyzed the binding relationship among MEG3, miR-20b-5p, and CREB1. UC mice presented with shortened colon length, lightened weight, increased DAI score, enhanced apoptosis, and significant inflammatory cell infiltration, while M2-EVs reversed these trends. In vitro, M2-EVs increased UC cell viability and reduced inflammation. Mechanistic experimentation revealed that M2-EVs transferred MEG3 into YAMC cells to up-regulate MEG3 expression and promote CREB1 transcription by competitively binding to miR-20b-5p. Moreover, up-regulation of MEG3 in M2-EVs enhanced the protective effect of M2-EVs on UC cells, while over-expression of miR-20b-5p attenuated the aforementioned protective effect of M2-EVs on UC mice and cells. Collectively, our findings revealed that M2-EVs carrying MEG3 enhanced UC cell viability and reduced inflammatory responses via the miR-20b-5p/CREB1 axis, thus alleviating UC inflammation.
Collapse
Affiliation(s)
- Yu-Xuan Wang
- Department of Gastroenterology, Hainan General Hospital, Haikou, P.R. China
| | - Cheng Lin
- Department of Gastroenterology, Hainan General Hospital, Haikou, P.R. China
| | - Lu-Jia Cui
- Department of Gastroenterology, Hainan General Hospital, Haikou, P.R. China
| | - Tao-Zhi Deng
- Department of Gastroenterology, Hainan General Hospital, Haikou, P.R. China
| | - Qiu-Min Li
- Department of Gastroenterology, Hainan General Hospital, Haikou, P.R. China
| | - Feng-Ying Chen
- Department of Gastroenterology, Hainan General Hospital, Haikou, P.R. China
| | - Xin-Pu Miao
- Department of Gastroenterology, Hainan General Hospital, Haikou, P.R. China
| |
Collapse
|
4
|
Yin KJ, Ren JN, Li X, Fan G, Zhao L, Li Z, Zhang LL, Xie DY, Pan SY, Yuan F. Effect and mechanism of high-fat diet on the preference for sweeteners on mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2021; 101:1844-1853. [PMID: 32901966 DOI: 10.1002/jsfa.10798] [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: 01/08/2020] [Revised: 05/31/2020] [Accepted: 09/09/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Male Kunming mice were divided into a normal diet group (control group) and a high-fat diet group (HF group) (185 g·kg-1 protein, 600 g·kg-1 fat and 205 g·kg-1 carbohydrate). After 8 weeks' feeding, behavioral indicators and biochemical indicators in serum were determined. The double-bottle preference experiment was used to study the preferences of mice for five sweeteners. The monoamine neurotransmitter content, gene expression related to dopamine (DA), and opioid receptors were also determined. RESULTS The body weight of the mice in the HF group increased significantly (P < 0.05) after 36 days compared with the control group. The feed intake of the HF group increased sharply in the first 12 days, and then it became basically unchanged. The preference of the HF group for all of the five sweeteners was highly significantly lower (P < 0.01) than that of the control group. Depression-related behavior was observed in the HF group mice. The triglyceride (TG), total cholesterol (TC), and low-density lipoprotein cholesterol (LDLC) content in the HF group were very much higher (P < 0.01) than those of the control group. The gene expression related to DA and opioid receptor in the HF group was significantly lower than that of the control group, except for preproenkephalin (PENK). CONCLUSIONS In summary, this study suggested that a long-term high-fat diet could result in a decrease in the preference for sweeteners and could result in a state of reward hypofunction in mice. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Kai-Jing Yin
- College of Food Science and Technology, Huazhong Agricultural University, Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan, China
| | - Jing-Nan Ren
- College of Food Science and Technology, Huazhong Agricultural University, Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan, China
| | - Xiao Li
- College of Food Science and Technology, Huazhong Agricultural University, Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan, China
| | - Gang Fan
- College of Food Science and Technology, Huazhong Agricultural University, Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan, China
| | - Lei Zhao
- Food and Agriculture Standardization Institute, China National Institute of Standardization, Beijing, China
| | - Zhi Li
- College of Food Science and Technology, Huazhong Agricultural University, Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan, China
| | - Lu-Lu Zhang
- College of Food Science and Technology, Huazhong Agricultural University, Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan, China
| | - Ding-Yuan Xie
- College of Food Science and Technology, Huazhong Agricultural University, Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan, China
| | - Si-Yi Pan
- College of Food Science and Technology, Huazhong Agricultural University, Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan, China
| | - Fang Yuan
- College of Food Science and Technology, Huazhong Agricultural University, Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan, China
| |
Collapse
|
5
|
Shen Z, Zheng K, Zou J, Gu P, Xing J, Zhang L, Zhu L, Shen H. Chinese herbal extract granules combined with 5-aminosalicylic acid for patients with moderately active ulcerative colitis: study protocol for a multicenter randomized double-blind placebo-controlled trial. Trials 2021; 22:55. [PMID: 33441157 PMCID: PMC7805063 DOI: 10.1186/s13063-020-05012-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 12/29/2020] [Indexed: 12/30/2022] Open
Abstract
Background Ulcerative colitis (UC) is an intestinal inflammatory disease characterized by inflammation of the colonic mucosa. With unknown pathogenesis, it has become a chronic lifetime disorder worldwide. In patients with moderately active UC, several therapies (e.g., aminosalicylates, corticosteroids, immunosuppressants, and biologics) are recommended for induction (or maintenance) of remission. Given the side effects and disease burden, it is difficult for most patients to achieve ideal treatment goals in clinical practice. Chinese herbal medicine (CHM), as a complementary therapy, has been widely used in the management of UC in China. Qing-Chang-Hua-Shi granule (QCHS) is a classical Chinese herbal formula. Our preliminary study suggested that the QCHS decoction has a significant effect on patients with moderately active UC. However, its effectiveness and safety has not been evaluated convincingly. Therefore, we designed this protocol to investigate the efficacy of QCHS granule for moderately active UC. Methods This is a multicenter, randomized, double-blind, placebo-controlled, superiority trial. A total of 120 patients with moderately active UC will be recruited from 10 hospitals in China. Each eligible participant will be randomly assigned to receive QCHS granule or placebo for 12 weeks. Both groups will be given basic treatment with mesalazine (4 g/day). The primary outcomes are the clinical response (remission) rate. The secondary outcomes are health-related quality of life, endoscopic response rate, mucosal healing rate, and inflammatory markers (e.g., fecal calprotectin and CRP). The whole study period will last 36 weeks, including 24 weeks follow-up time. According to the intention-to-treat principle, variables will be assessed at 2, 4, 6, 8, 10, and 12 weeks after study commencement. Discussion This is the first randomized controlled clinical study protocol regarding Chinese herbal extract granules in the management of moderately active UC. We aim to investigate the superiority of QCHS granules over placebo in terms of induction of remission. If the trial shows significant benefits of QCHS granules, it will help clinical practitioners, UC patients, and policymakers make more informed choices in the decision-making. Trial registration Chinese Clinical Trial Registry ChiCTR-IOR-14005554. Registered on 27 November 2014. Supplementary Information The online version contains supplementary material available at 10.1186/s13063-020-05012-8.
Collapse
Affiliation(s)
- Zhaofeng Shen
- Institute of Digestive Diseases, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,School of Public Health, Nanjing Medical University, Nanjing, China
| | - Kai Zheng
- Department of Gastroenterology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Jiandong Zou
- Institute of Digestive Diseases, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Peiqing Gu
- Department of Gastroenterology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Jing Xing
- Department of Gastroenterology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Lu Zhang
- Department of Gastroenterology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Lei Zhu
- Department of Gastroenterology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.
| | - Hong Shen
- Department of Gastroenterology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.
| |
Collapse
|
6
|
A Comparison of Gene Expression Changes in the Blood of Individuals Consuming Diets Supplemented with Olives, Nuts or Long-Chain Omega-3 Fatty Acids. Nutrients 2020; 12:nu12123765. [PMID: 33302351 PMCID: PMC7762614 DOI: 10.3390/nu12123765] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022] Open
Abstract
Background: The Mediterranean diet, which is rich in olive oil, nuts, and fish, is considered healthy and may reduce the risk of chronic diseases. Methods: Here, we compared the transcriptome from the blood of subjects with diets supplemented with olives, nuts, or long-chain omega-3 fatty acids and identified the genes differentially expressed. The dietary genes obtained were subjected to network analysis to determine the main pathways, as well as the transcription factors and microRNA interaction networks to elucidate their regulation. Finally, a gene-associated disease interaction network was performed. Results: We identified several genes whose expression is altered after the intake of components of the Mediterranean diets compared to controls. These genes were associated with infection and inflammation. Transcription factors and miRNAs were identified as potential regulators of the dietary genes. Interestingly, caspase 1 and sialophorin are differentially expressed in the opposite direction after the intake of supplements compared to Alzheimer’s disease patients. In addition, ten transcription factors were identified that regulated gene expression in supplemented diets, mild cognitive impairment, and Alzheimer’s disease. Conclusions: We identified genes whose expression is altered after the intake of the supplements as well as the transcription factors and miRNAs involved in their regulation. These genes are associated with schizophrenia, neoplasms, and rheumatic arthritis, suggesting that the Mediterranean diet may be beneficial in reducing these diseases. In addition, the results suggest that the Mediterranean diet may also be beneficial in reducing the risk of dementia.
Collapse
|
7
|
Chang ML, Yang Z, Yang SS. Roles of Adipokines in Digestive Diseases: Markers of Inflammation, Metabolic Alteration and Disease Progression. Int J Mol Sci 2020; 21:E8308. [PMID: 33167521 PMCID: PMC7663948 DOI: 10.3390/ijms21218308] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/30/2020] [Accepted: 11/01/2020] [Indexed: 12/13/2022] Open
Abstract
Adipose tissue is a highly dynamic endocrine tissue and constitutes a central node in the interorgan crosstalk network through adipokines, which cause pleiotropic effects, including the modulation of angiogenesis, metabolism, and inflammation. Specifically, digestive cancers grow anatomically near adipose tissue. During their interaction with cancer cells, adipocytes are reprogrammed into cancer-associated adipocytes and secrete adipokines to affect tumor cells. Moreover, the liver is the central metabolic hub. Adipose tissue and the liver cooperatively regulate whole-body energy homeostasis via adipokines. Obesity, the excessive accumulation of adipose tissue due to hyperplasia and hypertrophy, is currently considered a global epidemic and is related to low-grade systemic inflammation characterized by altered adipokine regulation. Obesity-related digestive diseases, including gastroesophageal reflux disease, Barrett's esophagus, esophageal cancer, colon polyps and cancer, non-alcoholic fatty liver disease, viral hepatitis-related diseases, cholelithiasis, gallbladder cancer, cholangiocarcinoma, pancreatic cancer, and diabetes, might cause specific alterations in adipokine profiles. These patterns and associated bases potentially contribute to the identification of prognostic biomarkers and therapeutic approaches for the associated digestive diseases. This review highlights important findings about altered adipokine profiles relevant to digestive diseases, including hepatic, pancreatic, gastrointestinal, and biliary tract diseases, with a perspective on clinical implications and mechanistic explorations.
Collapse
Affiliation(s)
- Ming-Ling Chang
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan
- Division of Hepatology, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Zinger Yang
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA;
| | - Sien-Sing Yang
- Liver Center, Cathay General Hospital Medical Center, Taipei 10630, Taiwan;
| |
Collapse
|
8
|
Liang S, Deng X, Lei L, Zheng Y, Ai J, Chen L, Xiong H, Mei Z, Cheng YC, Ren Y. The Comparative Study of the Therapeutic Effects and Mechanism of Baicalin, Baicalein, and Their Combination on Ulcerative Colitis Rat. Front Pharmacol 2019; 10:1466. [PMID: 31920656 PMCID: PMC6923254 DOI: 10.3389/fphar.2019.01466] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022] Open
Abstract
Introduction: Ulcerative colitis (UC) is an inflammatory bowel disease with a high incidence rate and a difficult treatment regimen. Recently, significant advances in the treatment of intestinal diseases, particularly UC, have been made with the use of the drugs baicalin and baicalein, separately or in combination. However, the therapeutic efficacy and mechanism of action of baicalin, baicalein, and their combination therapy, in the treatment of UC has not been fully elucidated. Materials and Methods: we constructed a UC rat model that encompassed a variety of complex factors, including a high-sugar and high-fat diet, a high temperature and humidity environment (HTHE), excess drinking, and infection of Escherichia coli. Model rats were then treated with baicalin, baicalein, or a combination of the two. Results: The results showed significant differences in the therapeutic effects of baicalin, baicalein, and the combination therapy, in the treatment of UC, as well as differences in the inhibition of inflammation via the nuclear factor-κB and MAPK pathways. The rat model of UC was established as described above. Then, the rats were treated for 7 days with baicalin (100 mg kg-1), baicalein (100 mg kg-1), or both (100 mg kg-1, baicalin: baicalein = 4:1/1:1). Clinical symptoms and signs, body temperature, organ indices, histopathology, blood biochemistry, and metabolites were examined to compare treatment effects and indicators of UC. Baicalin, YSR (Young Scutellaria baicalensis ratio of baicalin and baicalein), baicalein, and WSR (Withered Scutellaria baicalensis ratio of baicalin and baicalein) had significantly different effects in terms of clinical symptoms and signs, body temperature, organ indices, serum inflammatory cytokine levels, blood biochemistry, and histopathology changes in the main organs; YSR exhibited the best treatment effects. LC-MS/MS was used to detect the conversion of baicalin, baicalein, or both, into the six types of metabolites: baicalin, wogonoside, oroxin A, baicalein, wogonin, and oroxylin A. The levels of the six metabolites under the different treatment conditions were significantly different in the large intestine, small intestine, and lungs, but not in the blood. The levels of the six metabolites were significantly different in the large intestine, small intestine, and lung, but not in the serum. Conclusion: All these results indicate that baicalin and baicalein should be used more accurately in specific diseases, especially baicalin or high content of baicalin in Scutellaria baicalensis (Tiaoqin) should be preferred in treatment of UC.
Collapse
Affiliation(s)
- Shuai Liang
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan, China
| | - Xin Deng
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan, China
| | - Lei Lei
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan, China
| | - Yao Zheng
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan, China
| | - Jiao Ai
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan, China
| | - Linlin Chen
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Hui Xiong
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan, China
| | - Zhinan Mei
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan, China
| | - Yung-Chi Cheng
- School of Medicine, Yale University, New Haven, CT, United States
| | - Yongshen Ren
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan, China.,School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
9
|
Pleotropic Roles of Autotaxin in the Nervous System Present Opportunities for the Development of Novel Therapeutics for Neurological Diseases. Mol Neurobiol 2019; 57:372-392. [PMID: 31364025 DOI: 10.1007/s12035-019-01719-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/23/2019] [Indexed: 12/23/2022]
Abstract
Autotaxin (ATX) is a soluble extracellular enzyme that is abundant in mammalian plasma and cerebrospinal fluid (CSF). It has two known enzymatic activities, acting as both a phosphodiesterase and a phospholipase. The majority of its biological effects have been associated with its ability to liberate lysophosphatidic acid (LPA) from its substrate, lysophosphatidylcholine (LPC). LPA has diverse pleiotropic effects in the central nervous system (CNS) and other tissues via the activation of a family of six cognate G protein-coupled receptors. These LPA receptors (LPARs) are expressed in some combination in all known cell types in the CNS where they mediate such fundamental cellular processes as proliferation, differentiation, migration, chronic inflammation, and cytoskeletal organization. As a result, dysregulation of LPA content may contribute to many CNS and PNS disorders such as chronic inflammatory or neuropathic pain, glioblastoma multiforme (GBM), hemorrhagic hydrocephalus, schizophrenia, multiple sclerosis, Alzheimer's disease, metabolic syndrome-induced brain damage, traumatic brain injury, hepatic encephalopathy-induced cerebral edema, macular edema, major depressive disorder, stress-induced psychiatric disorder, alcohol-induced brain damage, HIV-induced brain injury, pruritus, and peripheral nerve injury. ATX activity is now known to be the primary biological source of this bioactive signaling lipid, and as such, represents a potentially high-value drug target. There is currently one ATX inhibitor entering phase III clinical trials, with several additional preclinical compounds under investigation. This review discusses the physiological and pathological significance of the ATX-LPA-LPA receptor signaling axis and summarizes the evidence for targeting this pathway for the treatment of CNS diseases.
Collapse
|
10
|
Deelen P, van Dam S, Herkert JC, Karjalainen JM, Brugge H, Abbott KM, van Diemen CC, van der Zwaag PA, Gerkes EH, Zonneveld-Huijssoon E, Boer-Bergsma JJ, Folkertsma P, Gillett T, van der Velde KJ, Kanninga R, van den Akker PC, Jan SZ, Hoorntje ET, Te Rijdt WP, Vos YJ, Jongbloed JDH, van Ravenswaaij-Arts CMA, Sinke R, Sikkema-Raddatz B, Kerstjens-Frederikse WS, Swertz MA, Franke L. Improving the diagnostic yield of exome- sequencing by predicting gene-phenotype associations using large-scale gene expression analysis. Nat Commun 2019; 10:2837. [PMID: 31253775 PMCID: PMC6599066 DOI: 10.1038/s41467-019-10649-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 05/23/2019] [Indexed: 02/06/2023] Open
Abstract
The diagnostic yield of exome and genome sequencing remains low (8-70%), due to incomplete knowledge on the genes that cause disease. To improve this, we use RNA-seq data from 31,499 samples to predict which genes cause specific disease phenotypes, and develop GeneNetwork Assisted Diagnostic Optimization (GADO). We show that this unbiased method, which does not rely upon specific knowledge on individual genes, is effective in both identifying previously unknown disease gene associations, and flagging genes that have previously been incorrectly implicated in disease. GADO can be run on www.genenetwork.nl by supplying HPO-terms and a list of genes that contain candidate variants. Finally, applying GADO to a cohort of 61 patients for whom exome-sequencing analysis had not resulted in a genetic diagnosis, yields likely causative genes for ten cases.
Collapse
Affiliation(s)
- Patrick Deelen
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Genomics Coordination Center, 9700 VB, Groningen, The Netherlands
| | - Sipko van Dam
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Johanna C Herkert
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Juha M Karjalainen
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Harm Brugge
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Kristin M Abbott
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Cleo C van Diemen
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Paul A van der Zwaag
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Erica H Gerkes
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Evelien Zonneveld-Huijssoon
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Jelkje J Boer-Bergsma
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Pytrik Folkertsma
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Tessa Gillett
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - K Joeri van der Velde
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Genomics Coordination Center, 9700 VB, Groningen, The Netherlands
| | - Roan Kanninga
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Genomics Coordination Center, 9700 VB, Groningen, The Netherlands
| | - Peter C van den Akker
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Sabrina Z Jan
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Edgar T Hoorntje
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands.,Netherlands Heart Institute, 3511 EP, Utrecht, The Netherlands
| | - Wouter P Te Rijdt
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands.,Netherlands Heart Institute, 3511 EP, Utrecht, The Netherlands
| | - Yvonne J Vos
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Jan D H Jongbloed
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Conny M A van Ravenswaaij-Arts
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Richard Sinke
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Birgit Sikkema-Raddatz
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | | | - Morris A Swertz
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Genomics Coordination Center, 9700 VB, Groningen, The Netherlands
| | - Lude Franke
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands.
| |
Collapse
|
11
|
Ninou I, Magkrioti C, Aidinis V. Autotaxin in Pathophysiology and Pulmonary Fibrosis. Front Med (Lausanne) 2018; 5:180. [PMID: 29951481 PMCID: PMC6008954 DOI: 10.3389/fmed.2018.00180] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/25/2018] [Indexed: 12/17/2022] Open
Abstract
Lysophospholipid signaling is emerging as a druggable regulator of pathophysiological responses, and especially fibrosis, exemplified by the relative ongoing clinical trials in idiopathic pulmonary fibrosis (IPF) patients. In this review, we focus on ectonucleotide pyrophosphatase-phosphodiesterase 2 (ENPP2), or as more widely known Autotaxin (ATX), a secreted lysophospholipase D (lysoPLD) largely responsible for extracellular lysophosphatidic acid (LPA) production. In turn, LPA is a bioactive phospholipid autacoid, forming locally upon increased ATX levels and acting also locally through its receptors, likely guided by ATX's structural conformation and cell surface associations. Increased ATX activity levels have been detected in many inflammatory and fibroproliferative conditions, while genetic and pharmacologic studies have confirmed a pleiotropic participation of ATX/LPA in different processes and disorders. In pulmonary fibrosis, ATX levels rise in the broncheoalveolar fluid (BALF) and stimulate LPA production. LPA engagement of its receptors activate multiple G-protein mediated signal transduction pathways leading to different responses from pulmonary cells including the production of pro-inflammatory signals from stressed epithelial cells, the modulation of endothelial physiology, the activation of TGF signaling and the stimulation of fibroblast accumulation. Genetic or pharmacologic targeting of the ATX/LPA axis attenuated disease development in animal models, thus providing the proof of principle for therapeutic interventions.
Collapse
Affiliation(s)
- Ioanna Ninou
- Division of Immunology, Alexander Fleming Biomedical Sciences Research Center, Athens, Greece
| | - Christiana Magkrioti
- Division of Immunology, Alexander Fleming Biomedical Sciences Research Center, Athens, Greece
| | - Vassilis Aidinis
- Division of Immunology, Alexander Fleming Biomedical Sciences Research Center, Athens, Greece
| |
Collapse
|