1
|
Zeng X, Yu P, Li D, Li Y, Wang X, Yang X, Ren D. Structural characterization and alleviative effects of novel polysaccharides from Artemisia sphaerocephala Krasch seed on obese mice by regulating gut microbiota. Int J Biol Macromol 2025; 310:143407. [PMID: 40274139 DOI: 10.1016/j.ijbiomac.2025.143407] [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: 10/22/2024] [Revised: 04/07/2025] [Accepted: 04/20/2025] [Indexed: 04/26/2025]
Abstract
This study aimed to investigate the efficacy of polysaccharides from Artemisia sphaerocephala Krasch (ASK) seed in alleviating high fat diet (HFD) caused obesity. Here, three polysaccharide fractions (ASKP1, ASKP2 and ASKP3) were purified from ASK seed. Chemical characteristic analysis revealed that ASKP1 is a neutral heteropolysaccharide with the average molecular weight of 9.08 × 105 Da, while ASKP2 and ASKP3 are acidic heteropolysaccharides with the molecular weight of 9.39 × 105 and 8.41 × 105 Da, respectively. Animal experiment found that three ASKP fractions obviously relieved obesity and related metabolic disorders induced by HFD, while ASKP1 was more effective in reducing the blood glucose and serum LDL levels. 16S rDNA sequencing showed that ASKP fractions improved the gut microbiota imbalance of obese mice, and ASKP1 promoted the proliferation of beneficial bacterium Akkermansia more effectively than ASKP2 and ASKP3. Furthermore, ASKP fractions facilitated thermogenesis of brown adipose tissue (BAT) of obese mice, as evidenced by increased expression of thermogenic marker genes UCP1 in BAT, and the thermogenesis effect of ASKP1 was the most obvious. Taken together, our results show that ASKP1 is a novel prebiotic that may be used to treat obesity and its related abnormal metabolism.
Collapse
Affiliation(s)
- Xiaoqian Zeng
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Pinglian Yu
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China; Key Laboratory of YunNan University for Plateau Characteristic Functional Food, School of Chemistry and Chemical Engineering, Zhaotong University, 657000, China.
| | - Donglu Li
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yixiao Li
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xuejie Wang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Daoyuan Ren
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
2
|
Li Z, Song L, Yang Y, Zhao Y, Ma S. Mannose enhances anti-tumor effect of PLX4032 in anaplastic thyroid cancer. Endocr Relat Cancer 2025; 32:e240209. [PMID: 40063000 PMCID: PMC11964477 DOI: 10.1530/erc-24-0209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 03/02/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025]
Abstract
Anaplastic thyroid cancer represents the most aggressive form of thyroid cancer and harbors BRAF mutations in over 40% of cases. Vemurafenib (PLX4032), a BRAF kinase inhibitor, shows promise in BRAFV600E-positive advanced thyroid cancer but may promote resistance in anaplastic cases. This study investigates whether mannose, known to selectively inhibit thyroid cancer, enhances PLX4032 efficacy. To evaluate whether mannose could enhance the response of anaplastic thyroid cancer cells to vemurafenib, we employed several in vitro assays, including MTT, colony formation, flow cytometry, migration and invasion assays. In addition, we performed in vivo assays using mouse models with subcutaneous xenografts. Our findings demonstrated that vemurafenib and mannose synergistically inhibit anaplastic thyroid cancer cell proliferation. The combined treatment significantly impeded anaplastic thyroid cancer cell migration and invasion while promoting apoptosis. In vivo studies corroborated these observations. The underlying mechanism by which mannose potentiates the antitumor effects of vemurafenib was explored using the Seahorse XFe96 Analyzer to measure glycolysis parameters and Western blotting to assess the expression of associated proteins. Mechanistically, vemurafenib reduced the expression of ZIP10, which in turn decreased the enzyme activity of phosphomannose isomerase. This suppression of ZIP10 enhanced mannose-mediated inhibition of glycolysis and thus its antitumor effect, as confirmed by rescue experiments with ZIP10 overexpression. The resulting decrease in glycolysis led to lower ATP levels, which are essential for the phosphorylation of ERK and AKT. Therefore, the combination of vemurafenib and mannose inhibited the levels of pERK and pAKT, thereby improving the effectiveness of PLX4032 in treating anaplastic thyroid cancer.
Collapse
Affiliation(s)
- Zhuolin Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, China
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Disease, College of Stomatology, Xi’an Jiaotong University, Xi’an, China
- Department of Cariology & Endodontics, College of Stomatology, Xi’an Jiaotong University, Xi’an, China
| | - Liumei Song
- Department of Endocrinology, Shaanxi Provincial People’s Hospital, Xi’an, China
- Department of Dermatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yuanxing Yang
- Department of Ultrasound, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yang Zhao
- Department of Endocrinology, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Sharui Ma
- Department of Endocrinology, Shaanxi Provincial People’s Hospital, Xi’an, China
| |
Collapse
|
3
|
Lei H, Zhang Y, Guan T, Liu M, Li Z, Liu J, Zhao J, Liu T. Modification of black soybean (Glycine max(L.)merr.) residue insoluble dietary fiber with ultrasonic, microwave, high temperature and high-pressure, and extrusion. Food Chem 2025; 473:143020. [PMID: 39864176 DOI: 10.1016/j.foodchem.2025.143020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 01/12/2025] [Accepted: 01/20/2025] [Indexed: 01/28/2025]
Abstract
Recent studies have emphasized the modification of Insoluble Dietary Fiber (IDF) to enhance its physicochemical properties and functional performance. This study systematically examined the effects of ultrasonic treatment, microwave irradiation, high-temperature and high-pressure processing, and screw extrusion on the physicochemical characteristics, in vitro antioxidant activity, and adsorption capacities of High-Purity Insoluble Dietary Fiber (HPIDF) derived from black bean residues. Although these physical modifications did not alter the functional group composition or crystalline structure of HPIDF, they significantly enhanced its porosity, water-holding capacity (WHC), oil-holding capacity (OHC), and adsorption capacities for glucose, cholesterol, bile salts, and metal ions. Notably, HPIDF treated under high-temperature and high-pressure conditions exhibited the highest adsorption capacities: 9.86 mmol/g for glucose, 8.69 mg/g (pH 2) and 9.69 mg/g (pH 7) for cholesterol, 0.183 g/g (pH 2) and 0.127 g/g (pH 7) for sodium cholate, and 0.699 mg/g (pH 2) and 0.774 mg/g (pH 7) for Cr2+.
Collapse
Affiliation(s)
- Hongyu Lei
- College of Food Science and Engineering, Changchun University, Changchun 130022, China
| | - Yu Zhang
- College of Food Science and Engineering, Changchun University, Changchun 130022, China
| | - Tianci Guan
- College of Food Science and Engineering, Changchun University, Changchun 130022, China
| | - Mengge Liu
- College of Food Science and Engineering, Changchun University, Changchun 130022, China
| | - Zhiming Li
- College of Food Science and Engineering, Changchun University, Changchun 130022, China
| | - Jiaxin Liu
- Jilin Province Product Quality Supervision and Inspection Institute, Changchun 130103, China
| | - Jun Zhao
- College of Food Science and Engineering, Changchun University, Changchun 130022, China
| | - Tong Liu
- College of Food Science and Engineering, Changchun University, Changchun 130022, China.
| |
Collapse
|
4
|
Agostini D, Bartolacci A, Rotondo R, De Pandis MF, Battistelli M, Micucci M, Potenza L, Polidori E, Ferrini F, Sisti D, Pegreffi F, Pazienza V, Virgili E, Stocchi V, Donati Zeppa S. Homocysteine, Nutrition, and Gut Microbiota: A Comprehensive Review of Current Evidence and Insights. Nutrients 2025; 17:1325. [PMID: 40284190 PMCID: PMC12030302 DOI: 10.3390/nu17081325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Homocysteine, a sulfur-containing amino acid, is an intermediate product during the metabolism of methionine, a vital amino acid. An elevated concentration of homocysteine in the plasma, named hyperhomocysteinemia, has been significantly related to the onset of several diseases, including diabetes, multiple sclerosis, osteoporosis, cancer, and neurodegenerative disorders such as dementia, Alzheimer's and Parkinson's diseases. An interaction between metabolic pathways of homocysteine and gut microbiota has been reported, and specific microbial signatures have been found in individuals experiencing hyperhomocysteinemia. Furthermore, some evidence suggests that gut microbial modulation may exert an influence on homocysteine levels and related disease progression. Conventional approaches for managing hyperhomocysteinemia typically involve dietary interventions alongside the administration of supplements such as B vitamins and betaine. The present review aims to synthesize recent advancements in understanding interventions targeted at mitigating hyperhomocysteinemia, with a particular emphasis on the role of gut microbiota in these strategies. The emerging therapeutic potential of gut microbiota has been reported for several diseases. Indeed, a better understanding of the complex interaction between microbial species and homocysteine metabolism may help in finding novel therapeutic strategies to counteract hyperhomocysteinemia.
Collapse
Affiliation(s)
- Deborah Agostini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (D.A.); (M.B.); (M.M.); (L.P.); (E.P.); (F.F.); (D.S.); (S.D.Z.)
| | - Alessia Bartolacci
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (D.A.); (M.B.); (M.M.); (L.P.); (E.P.); (F.F.); (D.S.); (S.D.Z.)
| | - Rossella Rotondo
- Department of Human Science and Promotion of Quality of Life, San Raffaele Rome Open University, 00166 Rome, Italy; (M.F.D.P.); (V.S.)
- San Raffaele Cassino, 03043 Cassino, Italy
| | - Maria Francesca De Pandis
- Department of Human Science and Promotion of Quality of Life, San Raffaele Rome Open University, 00166 Rome, Italy; (M.F.D.P.); (V.S.)
- San Raffaele Cassino, 03043 Cassino, Italy
| | - Michela Battistelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (D.A.); (M.B.); (M.M.); (L.P.); (E.P.); (F.F.); (D.S.); (S.D.Z.)
| | - Matteo Micucci
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (D.A.); (M.B.); (M.M.); (L.P.); (E.P.); (F.F.); (D.S.); (S.D.Z.)
| | - Lucia Potenza
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (D.A.); (M.B.); (M.M.); (L.P.); (E.P.); (F.F.); (D.S.); (S.D.Z.)
| | - Emanuela Polidori
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (D.A.); (M.B.); (M.M.); (L.P.); (E.P.); (F.F.); (D.S.); (S.D.Z.)
| | - Fabio Ferrini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (D.A.); (M.B.); (M.M.); (L.P.); (E.P.); (F.F.); (D.S.); (S.D.Z.)
| | - Davide Sisti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (D.A.); (M.B.); (M.M.); (L.P.); (E.P.); (F.F.); (D.S.); (S.D.Z.)
| | - Francesco Pegreffi
- Department of Medicine and Surgery, Kore University of Enna, 94100 Enna, Italy;
| | - Valerio Pazienza
- Division of Gastroenterology, “Casa Sollievo della Sofferenza” Hospital, 71013 San Giovanni Rotondo, Italy;
| | - Edy Virgili
- School of Biosciences and Veterinary Medicine, University of Camerino, 62031 Camerino, Italy;
| | - Vilberto Stocchi
- Department of Human Science and Promotion of Quality of Life, San Raffaele Rome Open University, 00166 Rome, Italy; (M.F.D.P.); (V.S.)
| | - Sabrina Donati Zeppa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (D.A.); (M.B.); (M.M.); (L.P.); (E.P.); (F.F.); (D.S.); (S.D.Z.)
- Department of Human Science and Promotion of Quality of Life, San Raffaele Rome Open University, 00166 Rome, Italy; (M.F.D.P.); (V.S.)
| |
Collapse
|
5
|
Yang J, Zhu Y, Wei X, Ni D, Zhang W, Mu W. The use of isomerases and epimerases for the production of the functional sugars mannose, allulose and tagatose from Fructose. World J Microbiol Biotechnol 2025; 41:129. [PMID: 40202705 DOI: 10.1007/s11274-025-04344-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/28/2025] [Indexed: 04/10/2025]
Abstract
Fructose, a common monosaccharide in nature extensively utilized in the food industry, poses a risk of elevated chronic disease incidence with excessive consumption. The global movement for a healthier living has sparked a quest for sugar reduction in foodstuff. The growing concern regarding the adverse impact of excessive sugar consumption on public health has led to significant interest in investigating healthier sugar alternatives. Research efforts have refocused on converting fructose into high-value, reduced-calorie functional sugars. Fructose can undergo direct conversion into three such sugars-mannose, allulose, and tagatose-via a streamlined bioconversion process. Allulose and tagatose, epimers of fructose, are derivable directly from fructose through C-3 and C-4 epimerization processes, whereas mannose, the aldose isomer of fructose, can be synthesized via isomerization pathways. This article aims to present recent advancements in the physiological functions, production methods, and applications of functional sugars derived from fructose. Particularly, it focuses on the bioproduction of mannose, allulose, and tagatose from fructose, encompassing discussions on the recent progress in the related isomerases and epimerases, such as mannose isomerase/lyxose isomerase, ketose 3-epimerase, and tagatose 4-epimerase. This review will provide a fresh perspective on the high-value biological utilization of fructose resources.
Collapse
Affiliation(s)
- Junya Yang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Yingying Zhu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Xu Wei
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Dawei Ni
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Wenli Zhang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Jiangnan University, Wuxi, Jiangsu, 214122, China.
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
6
|
Hong JG, Trotman J, Carbajal Y, Dey P, Glass M, Sclar V, Alter IL, Zhang P, Wang L, Chen L, Petitjean M, Bhattacharya D, Wang S, Friedman SL, DeRossi C, Chu J. Mannose reduces fructose metabolism and reverses MASH in human liver slices and murine models in vivo. Hepatol Commun 2025; 9:e0671. [PMID: 40116750 PMCID: PMC11927666 DOI: 10.1097/hc9.0000000000000671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/20/2025] [Indexed: 03/23/2025] Open
Abstract
BACKGROUND Fibrosis drives liver-related mortality in metabolic dysfunction-associated steatohepatitis (MASH), yet we have limited medical therapies to target MASH-fibrosis progression. Here we report that mannose, a simple sugar, attenuates MASH steatosis and fibrosis in 2 robust murine models and human liver slices. METHODS The well-validated fat-and-tumor MASH murine model for liver steatosis and fibrosis was employed. Mannose was supplied in the drinking water at the start ("Prevention" group) or at week 6 of the 12-week MASH regimen ("Therapy" group). The in vivo antifibrotic effects of mannose supplementation were tested in a second model of carbon tetrachloride (CCl4)-induced liver fibrosis. A quantitative and automated digital pathology approach was used to comprehensively assess steatosis and fibrosis phenotypes. Mannose was also tested in vitro in human and primary mouse hepatocytes conditioned with free fatty acids alone or with fructose, and human precision-cut liver slices from patients with end-stage MASH cirrhosis. RESULTS Oral mannose supplementation improved liver fibrosis in vivo in both fat-and-tumor MASH and CCl4 mouse models, as well as in human precision-cut liver slice MASH samples. Mannose also reduced liver steatosis in fat-and-tumor MASH mice, and in human and mouse hepatocytes in vitro. Ketohexokinase, the main enzyme in fructolysis, was decreased with mannose in whole mouse liver, cultured hepatocytes, and human precision-cut liver slices. Removal of fructose or overexpression of ketohexokinase each abrogated the antisteatotic effects of mannose. CONCLUSIONS This study identifies mannose as a novel therapeutic candidate for MASH that mitigates steatosis by dampening hepatocyte ketohexokinase expression and exerts independent antifibrotic effects in 2 mouse models and human liver tissue slices.
Collapse
Affiliation(s)
- John G. Hong
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Joshaya Trotman
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Yvette Carbajal
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Poulomi Dey
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Mariel Glass
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Victoria Sclar
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Isaac L. Alter
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Peng Zhang
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Liheng Wang
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Li Chen
- PharmaNest Inc., Princeton, New Jersey, USA
| | | | - Dipankar Bhattacharya
- Department of Medicine, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shuang Wang
- Department of Medicine, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Scott L. Friedman
- Department of Medicine, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Charles DeRossi
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Jaime Chu
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| |
Collapse
|
7
|
Bhuvaneswari D, Riitvek B, Lakshmi BS. Multi Targeted Activity of Cocculus hirsutus through Modulation of DPP-IV and PTP-1B Leading to Enhancement of Glucose Uptake and Attenuation of Lipid Accumulation. Appl Biochem Biotechnol 2025; 197:2493-2507. [PMID: 39760988 DOI: 10.1007/s12010-024-05142-8] [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] [Accepted: 12/24/2024] [Indexed: 01/07/2025]
Abstract
Multi-targeted therapies are gaining attention in the management of multifactorial diseases due to their poly pharmacology, enhanced potency and reduced toxicity. Metabolic disorders like Type 2 diabetes mellitus (T2DM) and obesity necessitate multi-targeted therapy to improve insulin sensitivity, regulate glucose homeostasis and support weight loss. Medicinal plants rich in bioactive compounds exhibit multi-targetted action with minimal side effects. In the current study, Cocculus hirsutus methanol extract (CME) and its hydromethanolic fraction (HMF) were investigated for their multi-target potential. Significant inhibition of Dipeptidyl peptidase IV (DPP-IV), a key enzyme in glucose metabolism was observed due to CME (54%) and HMF (70%) at 10 µg/ml and 1 µg/ml respectively. Protein Tyrosine Phosphatase 1B (PTP-1B), involved in the regulation of insulin signalling, was also inhibited by CME (67%) and HMF (73%) at 10 µg/ml concentration. An increase in glucose uptake was observed due to CME (62% and 65%) and HMF (63% and 68%) in 3T3-L1 adipocytes and L6 myotubes at 100 ng/ml. Further, investigation of HMF showed a decrease in lipid accumulation by 63% at 1 µg/ml in 3T3-L1 cells. Interestingly, HMF improved insulin sensitivity by upregulating GLUT4 expression (p < 0.05) via the PI3K/AKT pathway in both 3T3-L1 adipocytes and L6 myotubes. An inhibition in lipid accumulation was also observed by suppression of Peroxisome proliferator-activated receptor γ (PPARγ) (p < 0.05), a key regulator of adipogenesis in 3T3-L1 adipocytes. Gas chromatography-mass spectrometry analysis of the HMF showed the major component to be 3-methylmannoside (26.52%).
Collapse
Affiliation(s)
- D Bhuvaneswari
- Tissue Culture and Drug Discovery Laboratory, Department of Biotechnology, Anna University, Chennai, 600 025, India
| | - B Riitvek
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, USA
| | - B S Lakshmi
- Tissue Culture and Drug Discovery Laboratory, Department of Biotechnology, Anna University, Chennai, 600 025, India.
| |
Collapse
|
8
|
Xu J, Zhao Y, Tyler Mertens R, Ding Y, Xiao P. Sweet regulation - The emerging immunoregulatory roles of hexoses. J Adv Res 2025; 69:361-379. [PMID: 38631430 PMCID: PMC11954837 DOI: 10.1016/j.jare.2024.04.014] [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: 10/04/2023] [Revised: 03/20/2024] [Accepted: 04/13/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND It is widely acknowledged that dietary habits have profound impacts on human health and diseases. As the most important sweeteners and energy sources in human diets, hexoses take part in a broad range of physiopathological processes. In recent years, emerging evidence has uncovered the crucial roles of hexoses, such as glucose, fructose, mannose, and galactose, in controlling the differentiation or function of immune cells. AIM OF REVIEW Herein, we reviewed the latest research progresses in the hexose-mediated modulation of immune responses, provided in-depth analyses of the underlying mechanisms, and discussed the unresolved issues in this field. KEY SCIENTIFIC CONCEPTS OF REVIEW Owing to their immunoregulatory effects, hexoses affect the onset and progression of various types of immune disorders, including inflammatory diseases, autoimmune diseases, and tumor immune evasion. Thus, targeting hexose metabolism is becoming a promising strategy for reversing immune abnormalities in diseases.
Collapse
Affiliation(s)
- Junjie Xu
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuening Zhao
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | - Yimin Ding
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Xiao
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China; The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
9
|
Sun Y, Yao J, Gao R, Hao J, Liu Y, Liu S. Interactions of non-starch polysaccharides with the gut microbiota and the effect of non-starch polysaccharides with different structures on the metabolism of the gut microbiota: A review. Int J Biol Macromol 2025; 296:139664. [PMID: 39798752 DOI: 10.1016/j.ijbiomac.2025.139664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/19/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Humans consume large amounts of non-starch polysaccharides(NPs) daily. Some NPs, not absorbed by the body, proceed to the intestines. An increasing number of studies reveal a close relationship between NPs and gut microbiota(GM) that impact the human body. This review not only describes in detail the structures of several common NPs and their effects on GM, but also elucidates the degradation mechanisms of NPs in the intestine. The purpose of this review is to elucidate how NPs interact with GM in the intestine, which can provide valuable information for further studies of NPs.
Collapse
Affiliation(s)
- Yujiao Sun
- Natural Food Macromolecule Research Center, School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710021, PR China; Shaanxi Research Institute of Agricultural Products Processing Technology, Xi'an 710021, PR China.
| | - Jiaxuan Yao
- Natural Food Macromolecule Research Center, School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710021, PR China
| | - Running Gao
- Natural Food Macromolecule Research Center, School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710021, PR China
| | - Junyu Hao
- Natural Food Macromolecule Research Center, School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710021, PR China
| | - Yang Liu
- Shaanxi Academy of Traditional Chinese Medicine, Xi'an 710003, China
| | - Shuai Liu
- Shaanxi Academy of Traditional Chinese Medicine, Xi'an 710003, China.
| |
Collapse
|
10
|
Wei Y, Xu W, Zhang W, Petrova P, Petrov K, Ni D, Mu W. Characterization of Runella zeae D-mannose 2-epimerase and its expression in Bacillus subtilis for D-mannose production from D-glucose. Enzyme Microb Technol 2024; 181:110506. [PMID: 39265454 DOI: 10.1016/j.enzmictec.2024.110506] [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: 06/13/2024] [Revised: 08/12/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024]
Abstract
D-Mannose 2-epimerase (MEase) catalyzes the bioconversion between D-glucose and D-mannose. It is an important potential biocatalyst for large-scale production of D-mannose, a functional monosaccharide used in pharmaceutical and food industries. In this study, a new microbial MEase was characterized from Runella zeae DSM 19591. The enzyme was purified by one-step nickel-affinity chromatography and determined to be a dimeric protein with two identical subunits of approximately 86.1 kDa by gel filtration. The enzyme showed the highest activity at pH 8.0 and 40 °C, with a specific activity of 2.99 U/mg on D-glucose and 3.71 U/mg on D-mannose. The melting temperature (Tm) was 49.4 °C and the half-life was 115.14 and 3.23 h at 35 and 40 °C, respectively. The purified enzyme (1 U/mL) produced 115.7 g/L of D-mannose from 500 g/L of D-glucose for 48 h, with a conversion ratio of 23.14 %. It was successfully expressed in Bacillus subtilis WB600 via pP43NMK as the vector. The highest fermentation activity was 10.58 U/mL after fed-batch cultivation for 28 h, and the whole cells of recombinant B. subtilis produced 114.0 g/L of D-mannose from 500 g/L of D-glucose, with a conversion ratio of 22.8 %.
Collapse
Affiliation(s)
- Yuhan Wei
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wei Xu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wenli Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Penka Petrova
- Institute of Microbiology, Bulgarian Academy of Sciences, Sofia 1113, Bulgaria
| | - Kaloyan Petrov
- Institute of Chemical Engineering, Bulgarian Academy of Sciences, Sofia 1113, Bulgaria
| | - Dawei Ni
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
11
|
Xu X, Senior AM, Le Couteur DG, Cogger VC, Raubenheimer D, James DE, Parker B, Simpson SJ, Muller S, Yang JYH. eNODAL: an experimentally guided nutriomics data clustering method to unravel complex drug-diet interactions. Brief Bioinform 2024; 26:bbaf036. [PMID: 39982203 PMCID: PMC11843446 DOI: 10.1093/bib/bbaf036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/04/2024] [Accepted: 02/19/2025] [Indexed: 02/22/2025] Open
Abstract
Unraveling the complex interplay between nutrients and drugs via their effects on "omics" features could revolutionize our fundamental understanding of nutritional physiology, personalized nutrition, and, ultimately, human health span. Experimental studies in nutrition are starting to use large-scale "omics" experiments to pick apart the effects of such interacting factors. However, the high dimensionality of the omics features, coupled with complex fully factorial experimental designs, poses a challenge to the analysis. Current strategies for analyzing such types of data are based on between-feature correlations. However, these techniques risk overlooking important signals that arise from the experimental design and produce clusters that are hard to interpret. We present a novel approach for analyzing high-dimensional outcomes in nutriomics experiments, termed experiment-guided NutriOmics DatA cLustering ('eNODAL'). This three-step hybrid framework takes advantage of both Analysis of Variance (ANOVA)-type analyses and unsupervised learning methods to extract maximum information from experimental nutriomics studies. First, eNODAL categorizes the omics features into interpretable groups based on the significance of response to the different experimental variables using an ANOVA-like test. Such groups may include the main effects of a nutritional intervention and drug exposure or their interaction. Second, consensus clustering is performed within each interpretable group to further identify subclusters of features with similar response profiles to these experimental factors. Third, eNODAL annotates these subclusters based on their experimental responses and biological pathways enriched within the subcluster. We validate eNODAL using data from a mouse experiment to test for the interaction effects of macronutrient intake and drugs that target aging mechanisms in mice.
Collapse
Affiliation(s)
- Xiangnan Xu
- Chair of Statistics, Humboldt-Universität zu Berlin, Unter den Linden 6, Berlin 10178, Germany
| | - Alistair M Senior
- Charles Perkins Centre, University of Sydney, Johns Hopkins Drive, NSW 2050, Australia
- Sydney Precision Data Science Centre, University of Sydney, F07 Eastern Avenue, NSW 2050, Australia
- Laboratory of Data Discovery for Health Limited (D24H), 19 Science Park W Avenue, Hong Kong SAR 999077, China
| | - David G Le Couteur
- Charles Perkins Centre, University of Sydney, Johns Hopkins Drive, NSW 2050, Australia
- Centre for Education and Research on Ageing, Concord RG Hospital, Hospital Road, NSW 2138, Australia
- ANZAC Research Institute, Concord RG Hospital, Hospital Road, NSW 2138, Australia
| | - Victoria C Cogger
- Centre for Education and Research on Ageing, Concord RG Hospital, Hospital Road, NSW 2138, Australia
- ANZAC Research Institute, Concord RG Hospital, Hospital Road, NSW 2138, Australia
| | - David Raubenheimer
- Charles Perkins Centre, University of Sydney, Johns Hopkins Drive, NSW 2050, Australia
- School of Life and Environmental Science, University of Sydney, F22 Eastern Avenue, NSW 2050, Australia
| | - David E James
- Charles Perkins Centre, University of Sydney, Johns Hopkins Drive, NSW 2050, Australia
- ANZAC Research Institute, Concord RG Hospital, Hospital Road, NSW 2138, Australia
| | - Benjamin Parker
- Department of Anatomy and Physiology, University of Melbourne, 30 Royal Parade, VIC 3052, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, University of Sydney, Johns Hopkins Drive, NSW 2050, Australia
- School of Life and Environmental Science, University of Sydney, F22 Eastern Avenue, NSW 2050, Australia
| | - Samuel Muller
- Sydney Precision Data Science Centre, University of Sydney, F07 Eastern Avenue, NSW 2050, Australia
- School of Mathematical and Physical Sciences, Macquarie University, 18 Wally's Walk, NSW 2109, Australia
- School of Mathematics and Statistics, University of Sydney, F07 Eastern Avenue, NSW 2050, Australia
| | - Jean Y H Yang
- Charles Perkins Centre, University of Sydney, Johns Hopkins Drive, NSW 2050, Australia
- Sydney Precision Data Science Centre, University of Sydney, F07 Eastern Avenue, NSW 2050, Australia
- Laboratory of Data Discovery for Health Limited (D24H), 19 Science Park W Avenue, Hong Kong SAR 999077, China
- School of Mathematics and Statistics, University of Sydney, F07 Eastern Avenue, NSW 2050, Australia
| |
Collapse
|
12
|
Lu Y, Xiong Y, Zhang S, Wang B, Feng Y, Pu Z, Wei K, Chen J, Chen D, Zhang P. D-mannose reduces oxidative stress, inhibits inflammation, and increases treg cell proportions in mice with ulcerative colitis. Front Pharmacol 2024; 15:1454713. [PMID: 39555100 PMCID: PMC11563948 DOI: 10.3389/fphar.2024.1454713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 10/18/2024] [Indexed: 11/19/2024] Open
Abstract
Background Regulatory T (Treg) cells is required to dampen immune responses against intestinal microbiota, which aid in a healthy body to promise that the resident gut microbiota should not attract the attention of the immune system. Inflammation and inflammatory bowel disease (IBD) can be induced if the immune system fails to ignore the resident gut microbiota and targets them instead. D-mannose, a common monosaccharide in nature, has been shown to ameliorate multiple autoimmune diseases. This study aimed to investigate the therapeutic effect of D-mannose on mice ulcerative colitis (UC) induced by 2,4,6-trinitrobenzene sulfonic acid (TNBS), and elucidate its underlying mechanisms. Methods To simulate human IBD, we constructed a mouse model of UC by injecting TNBS into the colon. Results Our results demonstrated that D-mannose treatment effectively alleviated TNBS-induced UC in mice, as evidenced by the amelioration of UC symptoms. D-mannose treatment significantly reduced inflammation by decreasing the expression of proinflammatory cytokines and inflammation mediators. D-mannose treatment also significantly inhibited oxidative stress, promoted the expression of GSH and SOD, decreased the expression of MDA. Mechanistically, D-mannose upregulated the proportion of both CD4(+) Tregs and CD8(+) Tregs. Conclusion In summary, our study provides the first evidence of the therapeutic effect of D-mannose on mice with UC, which is likely mediated by upregulating Treg proportions.
Collapse
Affiliation(s)
- Yuqing Lu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning, China
| | - Yongjian Xiong
- Central Laboratory, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shuangshuang Zhang
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning, China
| | - Boya Wang
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning, China
| | - Yuntao Feng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhuonan Pu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Kun Wei
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning, China
| | - Jun Chen
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning, China
| | - Dapeng Chen
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning, China
| | - Peng Zhang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
13
|
Liu Y, Guan Q, Liu L, Ma L, Duan X, Che J. Metabolomic differences between exanthematous drug eruption and infectious mononucleosis. Skin Res Technol 2024; 30:e70043. [PMID: 39387831 PMCID: PMC11465872 DOI: 10.1111/srt.70043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/17/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Exanthematous drug eruption and infectious mononucleosis (IM) are both exanthematous diseases. Current research on exanthematous drug eruption and IM mainly targets identifying these disorders, the resulting differences at the metabolism level have not yet been systematically analyzed. MATERIALS AND METHODS A total of 30 cases of exanthematous drug eruption and IM, 10 patients without exanthema and 10 healthy volunteers were enrolled, 3 mL of fasting venous blood was collected, the serum metabolite content was detected by gas chromatography-mass spectrometry metabolomics. RESULTS A total of 165 metabolites were identified, exhibiting significant differences in plasma metabolic trends between exanthematous drug eruption and IM, and pinpointed 28 potential biomarkers. Notable changes were seen in the metabolic activities of the pentose phosphate pathway (PPP), tricarboxylic acid cycle (TCA-cycle), and galactose metabolism, characterized by increased levels of gluconate, gluconolactone, glucose, galactaric acid, and mannose, along with decreased amounts of pyruvic acid, succinic acid, malic acid, and glycerol, indicating an impairment in the exanthematous drug eruption group's capacity to endure oxidative stress and regulate energy metabolism. In contrast to its medication without rash counterpart, the exanthematous drug eruption group's plasma displayed distinct metabolic routes, predominantly in the processing of arginine and proline, along with the TCA. This resulted in a marked reduction in urea levels and a rise in pyruvate, citrate, and ornithine, indicating hypoxic stress as the primary cause of these rashes. In contrast to the healthy control group, the IM group showed 26 potential biomarkers, marked by increased levels of ketoglutaric acid, malic acid, pyruvic acid, and oxoglutaric acid, and reduced amounts of glutamine, galacturonic acid, arachidonic acid, trimethylphosphonic acid ester, gluconolactone, and indole acetic acid. Mainly, the metabolic pathways included the TCA, breaking down alanine, aspartate and glutamate metabolism, and the processing of D-glutamine and D-glutamate metabolism, underscoring the body's crucial role in generating energy and inflammatory agents through the citric acid cycle. CONCLUSIONS The comparison of serum metabolomic features of exanthematous drug eruptions and IM outlines a unique pattern closely related to the differences in the pathogenesis of these two exanthematous diseases.
Collapse
Affiliation(s)
- Yanqiu Liu
- Department of Dermatology and VenereologyThe Affiliated Hospital of Chengde Medical UniversityChengdeChina
| | - Qizhen Guan
- Department of Dermatology and VenereologyThe Affiliated Hospital of Chengde Medical UniversityChengdeChina
| | - Liyuan Liu
- Department of Dermatology and VenereologyThe Affiliated Hospital of Chengde Medical UniversityChengdeChina
| | - Lina Ma
- Department of Dermatology and VenereologyThe Affiliated Hospital of Chengde Medical UniversityChengdeChina
| | - Xinsuo Duan
- Department of Dermatology and VenereologyThe Affiliated Hospital of Chengde Medical UniversityChengdeChina
| | - Jiaozi Che
- Clinical labChengde central HospitalChengdeChina
| |
Collapse
|
14
|
Ni D, Wei Y, Zhang Y, Moussa TAA, Zhang W, Mu W. Biochemical identification of D-mannose 2-epimerase from Cytophagaceae bacterium SJW1-29 for efficient bioconversion of D-glucose to D-mannose. Enzyme Microb Technol 2024; 179:110465. [PMID: 38852283 DOI: 10.1016/j.enzmictec.2024.110465] [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: 04/22/2024] [Revised: 05/25/2024] [Accepted: 05/31/2024] [Indexed: 06/11/2024]
Abstract
Enzymatic production of D-mannose attracts increasing attention because of the health effects and commercial values of D-mannose. Several kinds of epimerases or isomerases have been used for enzymatic production of D-mannose from D-glucose or D-fructose. D-Mannose epimerase (MEase), belonging to N-acyl-D-glucosamine 2-epimerase superfamily enzymes, catalyzes the C-2 epimerization between D-glucose and D-mannose. In this study, a novel MEase was identified from Cytophagaceae bacterium SJW1-29. Sequence and structure alignments indicate that it is highly conserved with the reported R. slithyformis MEase with the known crystal structure. It was a metal-independent enzyme, with an optimal pH of 8.0 and an optimal temperature of 40 °C. The specific activities on D-glucose and D-mannose were 2.90 and 2.96 U/mg, respectively. The Km, kcat, and kcat/Km on D-glucose were measured to be 194.9 mM, 2.72 s-1, and 0.014 mM-1 s-1, respectively. The purified enzyme produced 23.15 g/L of D-mannose from 100 g/L of D-glucose at pH 8.0 and 40 °C for 8 h, with a conversion rate of 23.15 %.
Collapse
Affiliation(s)
- Dawei Ni
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Yuhan Wei
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Yulei Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Tarek A A Moussa
- Department of Botany and Microbiology, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Wenli Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, China.
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
15
|
Gao Y, Yang L, Yao K, Wang Y, Shao W, Yang M, Zhang X, Wei Y, Ren W. Exploration of Genes Related to Intramuscular Fat Deposition in Xinjiang Brown Cattle. Genes (Basel) 2024; 15:1121. [PMID: 39336712 PMCID: PMC11430885 DOI: 10.3390/genes15091121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
The aim of this study was to investigate the differentially expressed genes associated with intramuscular fat deposition in the longissimus dorsi muscle of Xinjiang Brown Bulls. The longissimus dorsi muscles of 10 Xinjiang Brown Bulls were selected under the same feeding conditions. The intramuscular fat content of muscle samples was determined by the Soxhlet extraction method, for which 5 samples with high intramuscular fat content (HIMF group) and 5 samples with low intramuscular fat content (LIMF group) were selected. It was found that the intramuscular fat content of the HIMF group was 46.054% higher than that of the LIMF group. Muscle samples produced by paraffin sectioning were selected for morphological observation. It was found that the fat richness of the HIMF group was better than that of the LIMF group. Transcriptome sequencing technology was used to analyze the gene expression differences of longissimus dorsi muscle. Through in-depth analysis of the longissimus dorsi muscle by transcriptome sequencing technology, we screened a total of 165 differentially expressed genes. The results of Gene Ontology (GO) enrichment analysis showed that the differentially expressed genes in the two groups were mainly clustered in biological pathways related to carbohydrate metabolic processes, redox processes and oxidoreductase activities. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis showed that the differentially expressed genes were significantly clustered in 15 metabolic pathways, which mainly covered fatty acid metabolism (related to lipid metabolism and glucose metabolism), the pentose phosphate pathway, the Peroxisome Proliferator-Activated Receptor (PPAR) signaling pathway and other important metabolic processes. The three genes that were predominantly enriched in the glycolipid metabolic pathway by analysis were SCD5, CPT1C and FBP2, all of which directly or indirectly affect intramuscular fat deposition. In summary, the present study investigated the differences in gene expression between high and low intramuscular fat content in the longissimus dorsi muscle of Xinjiang Brown Bulls by transcriptome sequencing technology and revealed the related signaling pathways. Therefore, we hypothesized that SCD5, CPT1C and FBP2 were the key genes responsible for the significant differences in intramuscular fat content of the longissimus dorsi muscles in a population of Xinjiang Brown Bulls. We expect that these findings will provide fundamental support for subsequent studies exploring key genes affecting fat deposition characteristics in Xinjiang Brown Bulls.
Collapse
Affiliation(s)
- Yu Gao
- Xinjiang Key Laboratory of Meat and Milk Production Herbivore Nutrition, College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China
| | - Liang Yang
- Xinjiang Key Laboratory of Meat and Milk Production Herbivore Nutrition, College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China
| | - Kangyu Yao
- Xinjiang Key Laboratory of Meat and Milk Production Herbivore Nutrition, College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China
| | - Yiran Wang
- Xinjiang Key Laboratory of Meat and Milk Production Herbivore Nutrition, College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China
| | - Wei Shao
- Xinjiang Key Laboratory of Meat and Milk Production Herbivore Nutrition, College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China
| | - Min Yang
- Xinjiang Key Laboratory of Meat and Milk Production Herbivore Nutrition, College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China
| | - Xinyu Zhang
- Xinjiang Key Laboratory of Meat and Milk Production Herbivore Nutrition, College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China
| | - Yong Wei
- Xinjiang Key Laboratory of Meat and Milk Production Herbivore Nutrition, College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China
| | - Wanping Ren
- Xinjiang Key Laboratory of Meat and Milk Production Herbivore Nutrition, College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China
| |
Collapse
|
16
|
Semnani-Azad Z, Rahman ML, Arguin M, Doyon M, Perron P, Bouchard L, Hivert MF. Plasma metabolomic profile of adiposity and body composition in childhood: The Genetics of Glucose regulation in Gestation and Growth cohort. Pediatr Obes 2024:e13149. [PMID: 38958048 DOI: 10.1111/ijpo.13149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/21/2024] [Accepted: 06/07/2024] [Indexed: 07/04/2024]
Abstract
OBJECTIVE This study identified metabolite modules associated with adiposity and body fat distribution in childhood using gold-standard measurements. METHODS We used cross-sectional data from 329 children at mid-childhood (age 5.3 ± 0.3 years; BMI 15.7 ± 1.5 kg/m2) from the Genetics of Glucose regulation in Gestation and Growth (Gen3G), a prospective pre-birth cohort. We quantified 1038 plasma metabolites and measured body composition using the gold-standard dual-energy x-ray absorptiometry (DXA), in addition to skinfold, waist circumference, and BMI. We applied weighted-correlation network analysis to identify a network of highly correlated metabolite modules. Spearman's partial correlations were applied to determine the associations of adiposity with metabolite modules and individual metabolites with false discovery rate (FDR) correction. RESULTS We identified a 'green' module of 120 metabolites, primarily comprised of lipids (mostly sphingomyelins and phosphatidylcholine), that showed positive correlations (all FDR p < 0.05) with DXA estimates of total and truncal fat (ρadjusted = 0.11-0.19), skinfold measures (ρadjusted = 0.09-0.26), and BMI and waist circumference (ρadjusted = 0.15 and 0.18, respectively). These correlations were similar when stratified by sex. Within this module, sphingomyelin (d18:2/14:0, d18:1/14:1)*, a sphingomyelin sub-specie that is an important component of cell membranes, showed the strongest associations. CONCLUSIONS A module of metabolites was associated with adiposity measures in childhood.
Collapse
Affiliation(s)
- Zhila Semnani-Azad
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Mohammad L Rahman
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland, USA
| | - Melina Arguin
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, Quebec, Canada
| | - Myriam Doyon
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, Quebec, Canada
| | - Patrice Perron
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, Quebec, Canada
- Faculty of Medicine and Life Sciences, Department of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Luigi Bouchard
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, Quebec, Canada
- Faculty of Medicine and Life Sciences, Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- Department of Medical Biology, CIUSSS du Saguenay-Lac-Saint- Jean, Saguenay, Quebec, Canada
| | - Marie-France Hivert
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, Quebec, Canada
- Diabetes Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
| |
Collapse
|
17
|
Zhang S, Gao YF, Zhang K, Deng GR, He GX, Gao PP, Yu YK, Yuan Y, Xing SJ, Zhao N, Zhang H, Di-Wu YC, Liu YH, Sui BD, Li Z, Ma J, Zheng CX. Integrating network pharmacology and experimental validation reveals therapeutic effects of D-mannose on NAFLD through mTOR suppression. Biochem Biophys Res Commun 2024; 715:149999. [PMID: 38678787 DOI: 10.1016/j.bbrc.2024.149999] [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: 04/11/2024] [Revised: 04/16/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD), a chronic liver condition and metabolic disorder, has emerged as a significant health issue worldwide. D-mannose, a natural monosaccharide widely existing in plants and animals, has demonstrated metabolic regulatory properties. However, the effect and mechanism by which D-mannose may counteract NAFLD have not been studied. In this study, network pharmacology followed by molecular docking analysis was utilized to identify potential targets of mannose against NAFLD, and the leptin receptor-deficient, genetically obese db/db mice was employed as an animal model of NAFLD to validate the regulation of D-mannose on core targets. As a result, 67 targets of mannose are predicted associated with NAFLD, which are surprisingly centered on the mechanistic target of rapamycin (mTOR). Further analyses suggest that mTOR signaling is functionally enriched in potential targets of mannose treating NAFLD, and that mannose putatively binds to mTOR as a core mechanism. Expectedly, repeated oral gavage of supraphysiological D-mannose ameliorates liver steatosis of db/db mice, which is based on suppression of hepatic mTOR signaling. Moreover, daily D-mannose administration reduced hepatic expression of lipogenic regulatory genes in counteracting NAFLD. Together, these findings reveal D-mannose as an effective and potential NAFLD therapeutic through mTOR suppression, which holds translational promise.
Collapse
Affiliation(s)
- Sha Zhang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, China
| | - Ying-Feng Gao
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Kai Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Guo-Rong Deng
- Department of Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Guang-Xiang He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, China
| | - Ping-Ping Gao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yi-Kang Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuan Yuan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Shu-Juan Xing
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Na Zhao
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Hong Zhang
- Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, China
| | - Yong-Chang Di-Wu
- College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, China
| | - Yi-Han Liu
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, Beijing, 100039, China
| | - Bing-Dong Sui
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Zhe Li
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, China.
| | - Jing Ma
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Chen-Xi Zheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
18
|
Chen N, Xu X, Guo Y, Zhao M, Li Y, Zhou T, Zhang X, Gao J, Zhu F, Guo C, Shi Y, Wang Q, Wu W, Zhang L, Li Y. Brain Short-Chain Fatty Acids Induce ACSS2 to Ameliorate Depressive-Like Behavior via PPARγ-TPH2 Axis. RESEARCH (WASHINGTON, D.C.) 2024; 7:0400. [PMID: 38939042 PMCID: PMC11210491 DOI: 10.34133/research.0400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/10/2024] [Indexed: 06/29/2024]
Abstract
Short-chain fatty acids (SCFAs) have been increasingly evidenced to be important bioactive metabolites of the gut microbiota and transducers in controlling diverse psychiatric or neurological disorders via the microbiota-gut-brain axis. However, the precise mechanism by which brain SCFAs extert multiple beneficial effects is not completely understood. Our previous research has demonstrated that the acetyl-coenzyme A synthetase short-chain family member 2 (ACSS2) is a novel target of the rapid and long-lasting antidepressant responses. Here, we show that micromolar SCFAs significantly augment both total cellular and nuclear ACSS2 to trigger tryptophan hydroxylase 2 (TPH2) promoter histone acetylation and its transcription in SH-SY5Y cells. In chronic-restraint-stress-induced depression mice, neuronal ACSS2 knockdown by stereotaxic injection of adeno-associated virus in the hippocampus abolished SCFA-mediated improvements in depressive-like behaviors of mice, supporting that ACSS2 is required for SCFA-mediated antidepressant responses. Mechanistically, the peroxisome-proliferator-activated receptor gamma (PPARγ) is identified as a novel partner of ACSS2 to activate TPH2 transcription. Importantly, PPARγ is also responsible for SCFA-mediated antidepressant-like effects via ACSS2-TPH2 axis. To further support brain SCFAs as a therapeutic target for antidepressant effects, d-mannose, which is a naturally present hexose, can significantly reverse the dysbiosis of gut microbiota in the chronic-restraint-stress-exposure mice and augment brain SCFAs to protect against the depressive-like behaviors via ACSS2-PPARγ-TPH2 axis. In summary, brain SCFAs can activate ACSS2-PPARγ-TPH2 axis to play the antidepressive-like effects, and d-mannose is suggested to be an inducer of brain SCFAs in resisting depression.
Collapse
Affiliation(s)
- Nuo Chen
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Xinyi Xu
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Yaxin Guo
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Ming Zhao
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Yubin Li
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Tian Zhou
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Xinyue Zhang
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Jie Gao
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Faliang Zhu
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Chun Guo
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Yongyu Shi
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Qun Wang
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Wenxian Wu
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology,
Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lining Zhang
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Yan Li
- Department of Pathogen Biology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| |
Collapse
|
19
|
Verdoodt F, Bhatti SFM, Kragic K, Van Ham L, Vanhaecke L, Hesta M, Hemeryck LY. Towards a better understanding of idiopathic epilepsy through metabolic fingerprinting of cerebrospinal fluid in dogs. Sci Rep 2024; 14:14750. [PMID: 38926488 PMCID: PMC11208596 DOI: 10.1038/s41598-024-64777-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Cerebrospinal fluid metabolomics is a promising research technology in the elucidation of nervous system disorders. Therefore, in this work, a cerebrospinal fluid (CSF) metabolomics method using liquid chromatography coupled to mass spectrometry was optimized and validated to cover a wide range of metabolites. An acceptable coefficient of variance regarding instrumental, within-lab and intra-assay precision was found for 95, 70 and 96 of 102 targeted metabolites, together with 1256, 676 and 976 untargeted compounds, respectively. Moreover, approximately 75% of targeted metabolites and 50% of untargeted compounds displayed good linearity across different dilution ranges. Consequently, metabolic alterations in CSF of dogs with idiopathic epilepsy (IE) were studied by comparing CSF of dogs diagnosed with IE (Tier II) to dogs with non-brain related disease. Targeted metabolome analysis revealed higher levels of cortisol, creatinine, glucose, hippuric acid, mannose, pantothenol, and 2-phenylethylamine (P values < 0.05) in CSF of dogs with IE, whereas CSF of dogs with IE showed lower levels of spermidine (P value = 0.02). Untargeted CSF metabolic fingerprints discriminated dogs with IE from dogs with non-brain related disease using Orthogonal Partial Least Squares Discriminant Analysis (R2(Y) = 0.997, Q2(Y) = 0.828), from which norepinephrine was putatively identified as an important discriminative metabolite.
Collapse
Affiliation(s)
- Fien Verdoodt
- Equine and Companion Animal Nutrition, Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
- Laboratory of Integrative Metabolomics, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Sofie F M Bhatti
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Karla Kragic
- Equine and Companion Animal Nutrition, Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Luc Van Ham
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Lynn Vanhaecke
- Laboratory of Integrative Metabolomics, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Myriam Hesta
- Equine and Companion Animal Nutrition, Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Lieselot Y Hemeryck
- Laboratory of Integrative Metabolomics, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium.
| |
Collapse
|
20
|
Yang YN, Han B, Zhang MQ, Chai NN, Yu FL, Qi WH, Tian MY, Sun DZ, Huang Y, Song QX, Li Y, Zhu MC, Zhang Y, Li X. Therapeutic effects and mechanisms of isoxanthohumol on DSS-induced colitis: regulating T cell development, restoring gut microbiota, and improving metabolic disorders. Inflammopharmacology 2024; 32:1983-1998. [PMID: 38642223 DOI: 10.1007/s10787-024-01472-5] [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: 12/29/2023] [Accepted: 03/27/2024] [Indexed: 04/22/2024]
Abstract
Ulcerative colitis (UC) is a severe hazard to human health. Since pathogenesis of UC is still unclear, current therapy for UC treatment is far from optimal. Isoxanthohumol (IXN), a prenylflavonoid from hops and beer, possesses anti-microbial, anti-oxidant, anti-inflammatory, and anti-angiogenic properties. However, the potential effects of IXN on the alleviation of colitis and the action of the mechanism is rarely studied. Here, we found that administration of IXN (60 mg/kg/day, gavage) significantly attenuated dextran sodium sulfate (DSS)-induced colitis, evidenced by reduced DAI scores and histological improvements, as well as suppressed the pro-inflammatory Th17/Th1 cells but promoted the anti-inflammatory Treg cells. Mechanically, oral IXN regulated T cell development, including inhibiting CD4+ T cell proliferation, promoting apoptosis, and regulating Treg/Th17 balance. Furthermore, IXN relieved colitis by restoring gut microbiota disorder and increasing gut microbiota diversity, which was manifested by maintaining the ratio of Firmicutes/Bacteroidetes balance, promoting abundance of Bacteroidetes and Ruminococcus, and suppressing abundance of proteobacteria. At the same time, the untargeted metabolic analysis of serum samples showed that IXN promoted the upregulation of D-( +)-mannose and L-threonine and regulated pyruvate metabolic pathway. Collectively, our findings revealed that IXN could be applied as a functional food component and served as a therapeutic agent for the treatment of UC.
Collapse
Affiliation(s)
- Ya-Na Yang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Bing Han
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Mao-Qing Zhang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Na-Nan Chai
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Feng-Lin Yu
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Wen-Hui Qi
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Meng-Yuan Tian
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Dong-Zhi Sun
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Ying Huang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Qing-Xin Song
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Yan Li
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Mao-Cui Zhu
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Yuan Zhang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China.
| | - Xing Li
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China.
| |
Collapse
|
21
|
Wang C, Guo H, Bai J, Yu L, Tian F, Zhao J, Zhang H, Chen W, Zhai Q. The roles of different Bacteroides uniformis strains in alleviating DSS-induced ulcerative colitis and related functional genes. Food Funct 2024; 15:3327-3339. [PMID: 38465411 DOI: 10.1039/d3fo04863b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Bacteroides is a common intestinal bacterium closely associated with host colitis. However, relevant studies have been focused on the genus level, which could not identify the major Bacteroides species associated with intestinal disease. Thus, we have evaluated the Bacteroides species structure in healthy people and mouse intestinal tracts and explored the change in major Bacteroides species during colitis development. The results demonstrated that B. uniformis with a high abundance in the intestinal tract of healthy people and mice may be a core species that contributes to colitis remission. The results of animal experiments reported that B. uniformis FNMHLBE1K1 (1K1) could alleviate the severity of colitis and enhance the expression of the tight junction protein occludin by regulating gut microbiota. Notably, the protective roles of 1K1 may be attributed to some specific genes. This study revealed that B. uniformis is a key microbe influencing the occurrence and development of colitis and it provides a scientific basis for screening the next generation of probiotics.
Collapse
Affiliation(s)
- Chen Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- College of Food Science, Southwest University, Chongqing, 400715, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Hang Guo
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Junying Bai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- Citrus Research Institute, Southwest University, Chongqing, 400712, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, Jiangsu, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
22
|
Chen N, Zhao M, Wu N, Guo Y, Cao B, Zhan B, Li Y, Zhou T, Zhu F, Guo C, Shi Y, Wang Q, Li Y, Zhang L. ACSS2 controls PPARγ activity homeostasis to potentiate adipose-tissue plasticity. Cell Death Differ 2024; 31:479-496. [PMID: 38332049 PMCID: PMC11043345 DOI: 10.1038/s41418-024-01262-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/10/2024] Open
Abstract
The appropriate transcriptional activity of PPARγ is indispensable for controlling inflammation, tumor and obesity. Therefore, the identification of key switch that couples PPARγ activation with degradation to sustain its activity homeostasis is extremely important. Unexpectedly, we here show that acetyl-CoA synthetase short-chain family member 2 (ACSS2) critically controls PPARγ activity homeostasis via SIRT1 to enhance adipose plasticity via promoting white adipose tissues beiging and brown adipose tissues thermogenesis. Mechanistically, ACSS2 binds directly acetylated PPARγ in the presence of ligand and recruits SIRT1 and PRDM16 to activate UCP1 expression. In turn, SIRT1 triggers ACSS2 translocation from deacetylated PPARγ to P300 and thereafter induces PPARγ polyubiquitination and degradation. Interestingly, D-mannose rapidly activates ACSS2-PPARγ-UCP1 axis to resist high fat diet induced obesity in mice. We thus reveal a novel ACSS2 function in coupling PPARγ activation with degradation via SIRT1 and suggest D-mannose as a novel adipose plasticity regulator via ACSS2 to prevent obesity.
Collapse
Affiliation(s)
- Nuo Chen
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ming Zhao
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Nan Wu
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yaxin Guo
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Baihui Cao
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bing Zhan
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yubin Li
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tian Zhou
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Faliang Zhu
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chun Guo
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yongyu Shi
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qun Wang
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yan Li
- Department of Pathogen Biology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Lining Zhang
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
23
|
Li Y, Shi C, Deng J, Qiu X, Zhang S, Wang H, Qin X, He Y, Cao B, Su H. Effects of Grape Pomace on Growth Performance, Nitrogen Metabolism, Antioxidants, and Microbial Diversity in Angus Bulls. Antioxidants (Basel) 2024; 13:412. [PMID: 38671860 PMCID: PMC11047470 DOI: 10.3390/antiox13040412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/21/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Polyphenol-rich grape pomace (GP) represents a valuable processing by-product with considerable potential as sustainable livestock feed. This study aimed to investigate the effects of different levels of GP on the growth performance and nitrogen utilization efficiency, antioxidant activity, and rumen and rectum microbiota of Angus bulls. Thirty Angus bulls were allocated three dietary treatments according to a completely randomized design: 0% (G0), 10% (G10), and 20% (G20) corn silage dry matter replaced with dried GP dry matter. The results showed that the average daily gain (ADG) of the G0 group and G10 group was higher than that of the G20 group (p < 0.05); urinary nitrogen levels decreased linearly with the addition of GP (linear, p < 0.05). In terms of antioxidants, the levels of catalase (CAT) in the G10 group were higher than in the G0 and G20 groups (p < 0.05), and the total antioxidative capacity (T-AOC) was significantly higher than that in the G20 group (p < 0.05). In addition, in the analysis of a microbial network diagram, the G10 group had better microbial community complexity and stability. Overall, these findings offer valuable insights into the potential benefits of incorporating GP into the diet of ruminants.
Collapse
Affiliation(s)
- Yingqi Li
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.L.); (C.S.); (J.D.); (S.Z.); (H.W.); (X.Q.); (Y.H.); (B.C.)
| | - Changxiao Shi
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.L.); (C.S.); (J.D.); (S.Z.); (H.W.); (X.Q.); (Y.H.); (B.C.)
| | - Jiajie Deng
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.L.); (C.S.); (J.D.); (S.Z.); (H.W.); (X.Q.); (Y.H.); (B.C.)
| | - Xinjun Qiu
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China;
| | - Siyu Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.L.); (C.S.); (J.D.); (S.Z.); (H.W.); (X.Q.); (Y.H.); (B.C.)
| | - Huili Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.L.); (C.S.); (J.D.); (S.Z.); (H.W.); (X.Q.); (Y.H.); (B.C.)
| | - Xiaoli Qin
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.L.); (C.S.); (J.D.); (S.Z.); (H.W.); (X.Q.); (Y.H.); (B.C.)
| | - Yang He
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.L.); (C.S.); (J.D.); (S.Z.); (H.W.); (X.Q.); (Y.H.); (B.C.)
| | - Binghai Cao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.L.); (C.S.); (J.D.); (S.Z.); (H.W.); (X.Q.); (Y.H.); (B.C.)
| | - Huawei Su
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.L.); (C.S.); (J.D.); (S.Z.); (H.W.); (X.Q.); (Y.H.); (B.C.)
| |
Collapse
|
24
|
Yang Y, Ma Q, Wang Q, Zhao L, Liu H, Chen Y. Mannose enhances intestinal immune barrier function and dextran sulfate sodium salt-induced colitis in mice by regulating intestinal microbiota. Front Immunol 2024; 15:1365457. [PMID: 38529272 PMCID: PMC10961387 DOI: 10.3389/fimmu.2024.1365457] [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: 01/04/2024] [Accepted: 02/27/2024] [Indexed: 03/27/2024] Open
Abstract
Background Inflammatory bowel disease (IBD) greatly affects human quality of life. Mannose has been reported to be used to treat IBD, but the mechanism is currently unknown. Methods C57/BL mice were used as research subjects, and the mouse acute colitis model was induced using dextran sulfate sodium salt (DSS). After oral administration of mannose, the body weights and disease activity index (DAI) scores of the mice were observed. The colon lengths, histopathological sections, fecal content microbial sequencing, colon epithelial inflammatory genes, and tight junction protein Occludin-1 expression levels were measured. We further used the feces of mice that had been orally administered mannose to perform fecal bacterial transplantation on the mice with DSS-induced colitis and detected the colitis-related indicators. Results Oral administration of mannose increased body weights and colon lengths and reduced DAI scores in mice with DSS-induced colitis. In addition, it reduced the expression of colon inflammatory genes and the levels of serum inflammatory factors (TNF-α, IL-6, and IL-1β), further enhancing the expression level of the colonic Occludin-1 protein and alleviating the toxic response of DSS to the intestinal epithelium of the mice. In addition, gut microbial sequencing revealed that mannose increased the abundance and diversity of intestinal flora. Additionally, after using the feces of the mannose-treated mice to perform fecal bacterial transplantation on the mice with DSS-induced colitis, they showed the same phenotype as the mannose-treated mice, and both of them alleviated the intestinal toxic reaction induced by the DSS. It also reduced the expression of intestinal inflammatory genes (TNF-α, IL-6, and IL-1β) and enhanced the expression level of the colonic Occludin-1 protein. Conclusion Mannose can treat DSS-induced colitis in mice, possibly by regulating intestinal microorganisms to enhance the intestinal immune barrier function and reduce the intestinal inflammatory response.
Collapse
Affiliation(s)
- Yi Yang
- Department of Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Qiming Ma
- Department of Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Qingyu Wang
- Department of Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Lifeng Zhao
- Department of Pharmacy, Affiliated Cancer Hospital of Inner Mongolia Medical University, Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, China
| | - Hengshan Liu
- Department of Emergency and trauma, Yichang Central People’s Hospital, Yichang, Hubei, China
| | - Yanjun Chen
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
25
|
Eberhart T, Stanley FU, Ricci L, Chirico T, Ferrarese R, Sisti S, Scagliola A, Baj A, Badurek S, Sommer A, Culp-Hill R, Dzieciatkowska M, Shokry E, Sumpton D, D'Alessandro A, Clementi N, Mancini N, Cardaci S. ACOD1 deficiency offers protection in a mouse model of diet-induced obesity by maintaining a healthy gut microbiota. Cell Death Dis 2024; 15:105. [PMID: 38302438 PMCID: PMC10834593 DOI: 10.1038/s41419-024-06483-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/03/2024]
Abstract
Aconitate decarboxylase 1 (ACOD1) is the enzyme synthesizing itaconate, an immuno-regulatory metabolite tuning host-pathogen interactions. Such functions are achieved by affecting metabolic pathways regulating inflammation and microbe survival. However, at the whole-body level, metabolic roles of itaconate remain largely unresolved. By using multiomics-integrated approaches, here we show that ACOD1 responds to high-fat diet consumption in mice by promoting gut microbiota alterations supporting metabolic disease. Genetic disruption of itaconate biosynthesis protects mice against obesity, alterations in glucose homeostasis and liver metabolic dysfunctions by decreasing meta-inflammatory responses to dietary lipid overload. Mechanistically, fecal metagenomics and microbiota transplantation experiments demonstrate such effects are dependent on an amelioration of the intestinal ecosystem composition, skewed by high-fat diet feeding towards obesogenic phenotype. In particular, unbiased fecal microbiota profiling and axenic culture experiments point towards a primary role for itaconate in inhibiting growth of Bacteroidaceae and Bacteroides, family and genus of Bacteroidetes phylum, the major gut microbial taxon associated with metabolic health. Specularly to the effects imposed by Acod1 deficiency on fecal microbiota, oral itaconate consumption enhances diet-induced gut dysbiosis and associated obesogenic responses in mice. Unveiling an unrecognized role of itaconate, either endogenously produced or exogenously administered, in supporting microbiota alterations underlying diet-induced obesity in mice, our study points ACOD1 as a target against inflammatory consequences of overnutrition.
Collapse
Affiliation(s)
- Tanja Eberhart
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Federico Uchenna Stanley
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Luisa Ricci
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Tiziana Chirico
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Roberto Ferrarese
- Laboratory of Medical Microbiology and Virology, Vita-Salute San Raffaele University, Milan, 20100, Italy
- IRCCS San Raffaele Hospital, Milan, 20100, Italy
- Synlab Italia, Castenedolo, BS, Italy
| | - Sofia Sisti
- Laboratory of Medical Microbiology and Virology, Vita-Salute San Raffaele University, Milan, 20100, Italy
- IRCCS San Raffaele Hospital, Milan, 20100, Italy
| | - Alessandra Scagliola
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
- Istituto Nazionale di Genetica Molecolare, INGM, "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Andreina Baj
- Department of Medicine and Technological Innovation, University of Insubria, Varese, Italy
| | - Sylvia Badurek
- Preclinical Phenotyping Facility, Vienna BioCenter Core Facilities (VBCF), member of the Vienna BioCenter (VBC), Vienna, Austria
| | - Andreas Sommer
- Next Generation Sequencing Facility, Vienna BioCenter Core Facilities (VBCF), member of the Vienna BioCenter (VBC), Vienna, Austria
| | - Rachel Culp-Hill
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | | | | | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Nicola Clementi
- Laboratory of Medical Microbiology and Virology, Vita-Salute San Raffaele University, Milan, 20100, Italy
- IRCCS San Raffaele Hospital, Milan, 20100, Italy
| | - Nicasio Mancini
- Laboratory of Medical Microbiology and Virology, Vita-Salute San Raffaele University, Milan, 20100, Italy
- IRCCS San Raffaele Hospital, Milan, 20100, Italy
- Laboratory of Medical Microbiology and Virology, Department of Medicine and Technological Innovation, University of Insubria, Varese, Italy
- Laboratory of Medical Microbiology and Virology, Fondazione Macchi University Hospital, Varese, Italy
| | - Simone Cardaci
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy.
| |
Collapse
|
26
|
Li Q, Yi X, Li L, Sun Y, Nie Z, Du J, Cao L, Gao J, Xu G. Effects of effective microorganisms on the physiological status, intestinal microbiome, and serum metabolites of Eriocheir sinensis. Int Microbiol 2024; 27:167-178. [PMID: 37261580 DOI: 10.1007/s10123-023-00375-9] [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: 09/22/2022] [Revised: 05/06/2023] [Accepted: 05/09/2023] [Indexed: 06/02/2023]
Abstract
The compound known as effective microorganisms (EMs) is widely used in aquaculture to improve water quality, but how they affect the health of Chinese mitten crab (Eriocheir sinensis) is unclear, especially in terms of intestinal microbiota and serum metabolites. In this study, we fed juvenile crabs with an EM-containing diet to explore the effects of EM on the physiological status, intestinal microbiome, and metabolites of E. sinensis. The activities of alanine aminotransferase and alkaline phosphatase were significantly enhanced by EM, indicating that EM supplementation effectively enhanced the antioxidant capacity of E. sinensis. Proteobacteria, Tenericutes, Firmicutes, Bacteroidetes, and Actinobacteria were the main intestinal microbes in both the control and EM groups. Linear discriminant effect size analysis showed that Fusobacteriaceae, Desulfovibrio, and Morganella were biomarkers in the control group, and Exiguobacterium and Rhodobacteraceae were biomarkers in the EM group. Metabolomics analysis revealed that EM supplementation increased cellular energy sources and decreased protein consumption, and oxidative stress. Together, these results indicate that EM can optimize the intestinal microbiome and serum metabolites, thereby benefiting the health of E. sinensis.
Collapse
Affiliation(s)
- Quanjie Li
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Xiangyu Yi
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, Jiangsu, China
| | - Le Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, Jiangsu, China
| | - Yi Sun
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Zhijuan Nie
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Jinliang Du
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Liping Cao
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Jiancao Gao
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Gangchun Xu
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China.
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, Jiangsu, China.
| |
Collapse
|
27
|
Liu W, Xie J, Jiang H, Zhou J, Lu X, Zuo D, Dong L, Chen Q. Mannose attenuates intestinal epithelial tight junction damage in experimental colitis mice by activating the AXIN-AMPK pathway. Int Immunopharmacol 2024; 127:111319. [PMID: 38064812 DOI: 10.1016/j.intimp.2023.111319] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/20/2023] [Accepted: 11/27/2023] [Indexed: 01/18/2024]
Abstract
Mannose is a unique natural sugar that can be found in a variety of fruits and vegetables. During the past decades, mannose has been reported to be effective in promoting immune tolerance and suppressing inflammatory diseases. Metabolic dysfunction and altered inflammation have clear implications for the development and progression of inflammatory diseases. Herein, we intended to reveal the molecular mechanism of mannose in protecting against intestinal epithelial damage in experimental colitis. We showed that mannose treatment significantly attenuated dextran sodium sulfate (DSS)-induced intestinal barrier damage. The AMPK pathway was responsible for the mannose-mediated protective effect in DSS-induced intestinal epithelial damage. Mechanistically, mannose promoted the axis inhibition protein (AXIN)-based AMPK activation, thereby preventing mitochondrial dysfunction and tight junction disruption in response to the DSS challenge. Cumulatively, the results indicate the use of mannose as a novel approach to treat IBD and other diseases involving tight junction dysfunction. The therapeutic effect of mannose is related to its regulatory function in AMPK pathway activation.
Collapse
Affiliation(s)
- Wenxin Liu
- Clinical Research Institute of Zhanjiang, Central People's Hospital of Zhanjiang, Guangdong Medical University, Zhanjiang, Guangdong 524045, China
| | - Jingwen Xie
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Honglian Jiang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, Guangzhou, Guangdong 510030, China
| | - Jia Zhou
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xiao Lu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Daming Zuo
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, China; Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Lijun Dong
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Qingyun Chen
- Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou 510080, Guangdong 519041, China.
| |
Collapse
|
28
|
Zhang H, Zhao X, Gao Y, Shi Y, Wei L, Li J, Liu C, Ma X. D-Mannose promotes recovery from experimental colitis by inducing AMPK phosphorylation to stimulate epithelial repair. Food Funct 2024; 15:625-646. [PMID: 38099724 DOI: 10.1039/d3fo03146b] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Delayed mucosal healing and impaired intestinal epithelial barrier function have been implicated in the pathogenesis of ulcerative colitis (UC). Accordingly, restoration of epithelial barrier function as a means to reshape mucosal homeostasis represents an important strategy for use in the treatment of UC. In this study, we examined the role and mechanisms of D-mannose in the recovery of colitis as assessed in both animal and cell models. We found that D-mannose ameliorated inflammation, promoted mucosal healing in the colon and therefore was able to induce the recovery of UC. Furthermore, D-mannose increased the expression of tight junction (TJ) proteins and reduced the intestinal permeability during the recovery of colitis. Moreover, D-mannose inhibited M1 macrophage polarization and promoted M2 macrophage polarization via inducing AMPK phosphorylation while reducing mTOR phosphorylation in both models. In addition, increased TJ protein expression and decreased paracellular permeability were observed in NCM460 cells when incubated with the supernatants of D-mannose-treated RAW264.7 cells, suggesting that M1/M2 polarization induced by D-mannose modulates the expression of TJ proteins. Further study showed that D-mannose significantly upregulated the expression of TJ proteins in DSS-treated NCM460 cells by inducing AMPK phosphorylation, indicating a direct protective effect on epithelial cells. Finally, the protective effects of D-mannose were significantly abrogated by the presence of compound C, an AMPK inhibitor. Taken together, our data indicate that D-mannose can alleviate inflammation and foster epithelial restitution in UC recovery by inducing the TJ protein expression, which are achieved by inducing AMPK phosphorylation in the epithelium and/or macrophages.
Collapse
Affiliation(s)
- Haojie Zhang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.
| | - Xue Zhao
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.
| | - Yifei Gao
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.
| | - Yao Shi
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.
| | - Lina Wei
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.
| | - Jingxin Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.
| | - Chuanyong Liu
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.
| | - Xuelian Ma
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
29
|
Hong JG, Carbajal Y, Trotman J, Glass M, Sclar V, Alter IL, Zhang P, Wang L, Chen L, Petitjean M, Friedman SL, DeRossi C, Chu J. Mannose Supplementation Curbs Liver Steatosis and Fibrosis in Murine MASH by Inhibiting Fructose Metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.17.576067. [PMID: 38293175 PMCID: PMC10827199 DOI: 10.1101/2024.01.17.576067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) can progress to cirrhosis and liver cancer. There are no approved medical therapies to prevent or reverse disease progression. Fructose and its metabolism in the liver play integral roles in MASH pathogenesis and progression. Here we focus on mannose, a simple sugar, which dampens hepatic stellate cell activation and mitigates alcoholic liver disease in vitro and in vivo . In the well-validated FAT-MASH murine model, oral mannose supplementation improved both liver steatosis and fibrosis at low and high doses, whether administered either at the onset of the model ("Prevention") or at week 6 of the 12-week MASH regimen ("Reversal"). The in vivo anti-fibrotic effects of mannose supplementation were validated in a second model of carbon tetrachloride-induced liver fibrosis. In vitro human and mouse primary hepatocytes revealed that the anti-steatotic effects of mannose are dependent on the presence of fructose, which attenuates expression of ketohexokinase (KHK), the main enzyme in fructolysis. KHK is decreased with mannose supplementation in vivo and in vitro, and overexpression of KHK abrogated the anti-steatotic effects of mannose. Our study identifies mannose as a simple, novel therapeutic candidate for MASH that mitigates metabolic dysregulation and exerts anti-fibrotic effects.
Collapse
|
30
|
Shen HH, Zhang YY, Wang XY, Wang CJ, Wang Y, Ye JF, Li MQ. Potential Causal Association between Plasma Metabolites, Immunophenotypes, and Female Reproductive Disorders: A Two-Sample Mendelian Randomization Analysis. Biomolecules 2024; 14:116. [PMID: 38254716 PMCID: PMC10813709 DOI: 10.3390/biom14010116] [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: 12/06/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND While extensive research highlighted the involvement of metabolism and immune cells in female reproductive diseases, causality remains unestablished. METHODS Instrumental variables for 486 circulating metabolites (N = 7824) and 731 immunophenotypes (N = 3757) were derived from a genome-wide association study (GWAS) meta-analysis. FinnGen contributed data on 14 female reproductive disorders. A bidirectional two-sample Mendelian randomization study was performed to determine the relationships between exposures and outcomes. The robustness of results, potential heterogeneity, and horizontal pleiotropy were examined through sensitivity analysis. RESULTS High levels of mannose were found to be causally associated with increased risks of gestational diabetes (GDM) (OR [95% CI], 6.02 [2.85-12.73], p = 2.55 × 10-6). A genetically predicted elevation in the relative count of circulating CD28-CD25++CD8+ T cells was causally related to increased female infertility risk (OR [95% CI], 1.26 [1.14-1.40], p = 1.07 × 10-5), whereas a high absolute count of NKT cells reduced the risk of ectopic pregnancy (OR [95% CI], 0.87 [0.82-0.93], p = 5.94 × 10-6). These results remained consistent in sensitivity analyses. CONCLUSIONS Our study supports mannose as a promising GDM biomarker and intervention target by integrating metabolomics and genomics.
Collapse
Affiliation(s)
- Hui-Hui Shen
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, China
| | - Yang-Yang Zhang
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, China
- Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xuan-Yu Wang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanpo Xinchengxi District, Jinghai District, Tianjin 301617, China
| | - Cheng-Jie Wang
- Department of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200011, China
| | - Ying Wang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Ji’nan 250012, China
| | - Jiang-Feng Ye
- Institute for Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138632, Singapore
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, China
- Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, China
| |
Collapse
|
31
|
Chen S, Wang K, Wang Q. Mannose: A Promising Player in Clinical and Biomedical Applications. Curr Drug Deliv 2024; 21:1435-1444. [PMID: 38310442 DOI: 10.2174/0115672018275954231220101637] [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: 09/20/2023] [Revised: 12/02/2023] [Accepted: 12/08/2023] [Indexed: 02/05/2024]
Abstract
Mannose, an isomer of glucose, exhibits a distinct molecular structure with the same formula but a different atom arrangement, contributing to its specific biological functions. Widely distributed in body fluids and tissues, particularly in the nervous system, skin, testes, and retinas, mannose plays a crucial role as a direct precursor for glycoprotein synthesis. Glycoproteins, essential for immune regulation and glycosylation processes, underscore the significance of mannose in these physiological activities. The clinical and biomedical applications of mannose are diverse, encompassing its anti-inflammatory properties, potential to inhibit bacterial infections, role in metabolism regulation, and suggested involvement in alleviating diabetes and obesity. Additionally, mannose shows promise in antitumor effects, immune modulation, and the construction of drug carriers, indicating a broad spectrum of therapeutic potential. The article aims to present a comprehensive review of mannose, focusing on its molecular structure, metabolic pathways, and clinical and biomedical applications, and also to emphasize its status as a promising therapeutic agent.
Collapse
Affiliation(s)
- Sijing Chen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
- The Department of Gynecologic Oncology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kana Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
- The Department of Gynecologic Oncology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiao Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
- The Department of Gynecologic Oncology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
32
|
Zhang L, Chen N, Zhan L, Bi T, Zhou W, Zhang L, Zhu L. Erchen Decoction alleviates obesity-related hepatic steatosis via modulating gut microbiota-drived butyric acid contents and promoting fatty acid β-oxidation. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116811. [PMID: 37336336 DOI: 10.1016/j.jep.2023.116811] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/11/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Erchen decoction (ECD) is a traditional Chinese medicine formula comprising six distinct herbs and has been documented to possess a protective effect against obesity. The study conducted previously demonstrated that ECD has the potential to effectively modulate the composition of gut microbiota and levels of short-chain fatty acids (SCFAs) in obese rat. However, the regulatory mechanism of ECD on gut microbiota and SCFAs and further improvement of obesity have not been thoroughly explained. AIM OF THE STUDY The objective of this study was to examine the therapeutic effect and molecular mechanism of ECD in a rat model of high-fat diet (HFD) feeding. MATERIALS AND METHODS Rats with HFD-induced obesity were treated with ECD. Upon completion of the study, serum and liver samples were procured to conduct biochemical, pathological, and Western blotting analyses. The investigation of alterations in the gut microbiota subsequent to ECD treatment was conducted through the utilization of 16S rRNA sequencing. The metabolic alterations in the cecal contents were examined through the utilization of mass spectrometry-ultraperformance liquid chromatography. RESULTS ECD treatment improved lipid metabolic disorders and reduced hepatic steatosis in HFD-induced obese rats. Obese rat treated with ECD showed a higher abundance of SCFA-producing bacteria, including Lactobacillus, Bifidobacterium, and Butyricicoccus, and lower abundance of disease-related bacteria, such as Bacteroides, Parabacteroides, and Sediminibacterium. Additionally, ECD caused an increase in total SCFAs levels; in particular, butyric acid was dramatically increased in the HFD group. Rats treated with ECD also exhibited significantly increased butyric acid concentrations in the serum and liver. The subsequent reduction in histone deacetylase 1 expression and increase in acetyl-histone 3-lysine 9 (H3K9ac) levels contributed to the promotion of fatty acid β-oxidation (FAO) in liver by ECD. CONCLUSION This study demonstrates that ECD regulates the gut microbiota and promotes butyric acid production to ameliorate obesity-related hepatic steatosis. The mechanism might be related to the promotion of FAO via a butyric acid-mediated increase in H3K9ac levels in the liver.
Collapse
Affiliation(s)
- Ling Zhang
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Ning Chen
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Libin Zhan
- Center for Innovative Engineering Technology in Traditional Chinese Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, China; Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, China; Key Laboratory of Liaoning Province for TCM Spleen-Viscera-State Modern Research, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, China.
| | - Tingting Bi
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Wen Zhou
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Lijing Zhang
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Lianlian Zhu
- Center for Innovative Engineering Technology in Traditional Chinese Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, China; Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, China; Key Laboratory of Liaoning Province for TCM Spleen-Viscera-State Modern Research, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, China.
| |
Collapse
|
33
|
Jin H, Liu X, Liu HX. Biological function, regulatory mechanism, and clinical application of mannose in cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188970. [PMID: 37657682 DOI: 10.1016/j.bbcan.2023.188970] [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: 04/19/2023] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 09/03/2023]
Abstract
Studies examining the regulatory roles and clinical applications of monosaccharides other than glucose in cancer have been neglected. Mannose, a common type of monosaccharide found in human body fluids and tissues, primarily functions in protein glycosylation rather than carbohydrate metabolism. Recent research has demonstrated direct anticancer effects of mannose in vitro and in vivo. Simply supplementing cell culture medium or drinking water with mannose achieved these effects. Moreover, mannose enhances the effectiveness of current cancer treatments including chemotherapy, radiotherapy, targeted therapy, and immune therapy. Besides the advancements in basic research on the anticancer effects of mannose, recent studies have reported its application as a biomarker for cancer or in the delivery of anticancer drugs using mannose-modified drug delivery systems. This review discusses the progress made in understanding the regulatory roles of mannose in cancer progression, the mechanisms underlying its anticancer effects, and its current application in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Haoyi Jin
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, Liaoning, China
| | - Xi Liu
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, Liaoning, China
| | - Hong-Xu Liu
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, Liaoning, China; Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, Liaoning, China.
| |
Collapse
|
34
|
Lin H, Li X, Zhao J, Wang L, Liu Y, Gao C. D-mannose reduces adipogenesis by inhibiting the PI3K/AKT signaling pathway. Histol Histopathol 2023; 38:1283-1294. [PMID: 37246829 DOI: 10.14670/hh-18-631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
PURPOSE To explore the effects and potential mechanisms of D-mannose on adipogenic differentiation of two kinds of representative mesenchymal stem cells (MSCs). METHODS We cultured two kinds of representative MSCs, human adipose tissue-derived stromal cells (hADSCs) as well as human bone marrow mesenchymal stem cells (hBMSCs), with adipogenic-induced medium containing D-mannose or D-fructose as the control. Oil red O staining, quantitative real-time polymerase chain reaction (qRT-PCR), and western blot (WB) were used to detect whether D-mannose had effects on adipogenic differentiation of MSCs. RNA sequencing (RNA-seq) transcriptomic analysis was further used to explore the potential mechanisms of D-mannose on adipogenic differentiation of MSCs. After that, qRT-PCR and WB were used to verify the results of RNA-seq. Last, we removed bilateral ovaries of female rats to establish an estrogen deficiency obesity model, and gave D-mannose intragastric administration. One month later, the femurs of rats were sliced for oil red O staining, and the inhibitory effect of D-mannose on lipid formation in vivo was studied. RESULTS Oil red O staining, qRT-PCR and WB in vitro demonstrated that D-mannose inhibited the adipogenic differentiation of both hADSCs and hBMSCs. Oil red O staining of femur sections proved that D-mannose was able to reduce in vivo adipogenesis. The results of RNA-seq transcriptomic analysis revealed that the adipogenesis-inhibition effects of D-mannose were performed by antagonizing the PI3K/AKT signaling pathway. Besides, qRT-PCR and WB further verified the results of RNA-seq. CONCLUSION Our study indicated that D-mannose was able to reduce adipogenic differentiation of both hADSCs and hBMSCs by antagonizing the PI3K/AKT signaling pathway. D-mannose is expected to be a safe and effective treatment strategy for obesity.
Collapse
Affiliation(s)
- Haozhi Lin
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shan Dong Province, PR China
| | - Xin Li
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shan Dong Province, PR China
| | - Jiping Zhao
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shan Dong Province, PR China
| | - Lei Wang
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shan Dong Province, PR China
| | - Yizhen Liu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shan Dong Province, PR China
| | - Cui Gao
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shan Dong Province, PR China.
| |
Collapse
|
35
|
Xie J, Wu S, Liao W, Ning J, Ding K. Src is a target molecule of mannose against pancreatic cancer cells growth in vitro & in vivo. Glycobiology 2023; 33:766-783. [PMID: 37658770 DOI: 10.1093/glycob/cwad070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 08/04/2023] [Accepted: 08/05/2023] [Indexed: 09/05/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant cancer with limited treatment options. Mannose, a common monosaccharide taken up by cells through the same transporters as glucose, has been shown to induce growth retardation and enhance cell death in response to chemotherapy in several cancers, including PDAC. However, the molecular targets and mechanisms underlying mannose's action against PDAC are not well understood. In this study, we used an integrative approach of network pharmacology, bioinformatics analysis, and experimental verification to investigate the pharmacological targets and mechanisms of mannose against PDAC. Our results showed that the protein Src is a key target of mannose in PDAC. Additionally, computational analysis revealed that mannose is a highly soluble compound that meets Lipinski's rule of five and that the expression of its target molecules is correlated with survival rates and prognosis in PDAC patients. Finally, we validated our findings through in vitro and in vivo experiments. In conclusion, our study provides evidence that mannose plays a critical role in inhibiting PDAC growth by targeting Src, suggesting that it may be a promising therapeutic candidate for PDAC.
Collapse
Affiliation(s)
- Jianhao Xie
- Carbohydrate-Based Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd, Pudong New district, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Beijing 100049, China
| | - Shengjie Wu
- Carbohydrate-Based Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd, Pudong New district, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Beijing 100049, China
| | - Wenfeng Liao
- Carbohydrate-Based Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd, Pudong New district, Shanghai 201203, China
| | - Jingru Ning
- Carbohydrate-Based Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd, Pudong New district, Shanghai 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Rd, Qixia District, Nanjing 210023, China
| | - Kan Ding
- Carbohydrate-Based Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd, Pudong New district, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Beijing 100049, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, SSIP Healthcare and Medicine Demonstration Zone, Zhongshan Tsuihang New District, Zhongshan, Guangdong 528400, China
| |
Collapse
|
36
|
LI C, YANG Y, FENG C, LI H, QU Y, WANG Y, WANG D, WANG Q, GUO J, SHI T, SUN X, WANG X, HOU Y, SUN Z, YANG T. Integrated 'omics analysis for the gut microbiota response to moxibustion in a rat model of chronic fatigue syndrome. J TRADIT CHIN MED 2023; 43:1176-1189. [PMID: 37946480 PMCID: PMC10623263 DOI: 10.19852/j.cnki.jtcm.20231018.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/08/2023] [Indexed: 11/12/2023]
Abstract
OBJECTIVE To observe the efficacy of moxibustion in the treatment of chronic fatigue syndrome (CFS) and explore the effects on gut microbiota and metabolic profiles. METHODS Forty-eight male Sprague-Dawley rats were randomly assigned to control group (Con), CFS model group (Mod, established by multiple chronic stress for 35 d), MoxA group (CFS model with moxibustion Shenque (CV8) and Guanyuan (CV4), 10 min/d, 28 d) and MoxB group (CFS model with moxibustion Zusanli (ST36), 10 min/d, 28 d). Open-field test (OFT) and Morris-water-maze test (MWMT) were determined for assessment the CFS model and the therapeutic effects of moxibustion.16S rRNA gene sequencing analysis based gut microbiota integrated untargeted liquid chromatograph-mass spectrometer (LC-MS) based fecal metabolomics were executed, as well as Spearman correlation analysis, was utilized to uncover the functional relevance between the potential metabolites and gut microbiota. RESULTS The results of our behavioral tests showed that moxibustion improved the performance of CFS rats in the OFT and the MWMT. Microbiome profiling analysis revealed that the gut microbiomes of CFS rats were less diverse with altered composition, including increases in pro-inflammatory species (such as Proteobacteria) and decreases in anti-inflammatory species (such as Bacteroides, Lactobacillus, Ruminococcus, and Prevotella). Moxibustion partially normalized these changes in the gut microbiota. Furthermore, CFS was associated with metabolic disorders, which were effectively ameliorated by moxibustion. This was demonstrated by the normalization of 33 microbiota-related metabolites, including mannose (P = 0.001), aspartic acid (P = 0.009), alanine (P = 0.007), serine (P = 0.000), threonine (P = 0.027), methionine (P = 0.023), 5-hydroxytryptamine (P = 0.008), alpha-linolenic acid (P = 0.003), eicosapentaenoic acid (P = 0.006), hypoxanthine (P = 0.000), vitamin B6 (P = 0.000), cholic acid (P = 0.013), and taurocholate (P = 0.002). Correlation analysis showed a significant association between the perturbed fecal microbiota and metabolite levels, with a notable negative relationship between LCA and Bacteroides. CONCLUSIONS In this study, we demonstrated that moxibustion has an antifatigue-like effect. The results from the 16S rRNA gene sequencing and metabolomics analysis suggest that the therapeutic effects of moxibustion on CFS are related to the regulation of gut microorganisms and their metabolites. The increase in Bacteroides and decrease in LCA may be key targets for the moxibustion treatment of CFS.
Collapse
Affiliation(s)
- Chaoran LI
- 1 Department of Acupuncture, Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Yan YANG
- 2 Department of Chinese Medical Literature, College of Basic Medicine, Heilongjiang University of Chinese medicine, Harbin 150040, China
| | - Chuwen FENG
- 3 Department of Rehabilitation, the First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin 150040, China
| | - Heng LI
- 7 Shanghai Applied Protein Technology Co., Ltd., Shanghai 200233, China
| | - Yuanyuan QU
- 5 Graduate School, the Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin 150040, China
| | - Yulin WANG
- 6 Department of Acupuncture, the Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin 150040, China
| | - Delong WANG
- 6 Department of Acupuncture, the Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin 150040, China
| | - Qingyong WANG
- 5 Graduate School, the Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin 150040, China
| | - Jing GUO
- 5 Graduate School, the Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin 150040, China
| | - Tianyu SHI
- 5 Graduate School, the Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin 150040, China
| | - Xiaowei SUN
- 4 Department of Acupuncture, the First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin 150040, China
| | - Xue WANG
- 8 Department of Acupuncture, Chongqing Changshou District People's Hospital, Chongqing 401220, China
| | - Yunlong HOU
- 9 College of integrated Chinese and Western Medicine, Hebei University of Chinese Medicine, and National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine, Hebei 050000, China
| | - Zhongren SUN
- 6 Department of Acupuncture, the Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin 150040, China
| | - Tiansong YANG
- 10 Department of Rehabilitation, the First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, and Traditional Chinese Medicine Informatics Key Laboratory of Heilongjiang Province, Harbin 150040, China
| |
Collapse
|
37
|
Perry RN, Albarracin D, Aherrahrou R, Civelek M. Network Preservation Analysis Reveals Dysregulated Metabolic Pathways in Human Vascular Smooth Muscle Cell Phenotypic Switching. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:372-381. [PMID: 37387208 PMCID: PMC10434832 DOI: 10.1161/circgen.122.003781] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/06/2023] [Indexed: 07/01/2023]
Abstract
BACKGROUND Vascular smooth muscle cells are key players involved in atherosclerosis, the underlying cause of coronary artery disease. They can play either beneficial or detrimental roles in lesion pathogenesis, depending on the nature of their phenotypic changes. An in-depth characterization of their gene regulatory networks can help better understand how their dysfunction may impact disease progression. METHODS We conducted a gene expression network preservation analysis in aortic smooth muscle cells isolated from 151 multiethnic heart transplant donors cultured under quiescent or proliferative conditions. RESULTS We identified 86 groups of coexpressed genes (modules) across the 2 conditions and focused on the 18 modules that are least preserved between the phenotypic conditions. Three of these modules were significantly enriched for genes belonging to proliferation, migration, cell adhesion, and cell differentiation pathways, characteristic of phenotypically modulated proliferative vascular smooth muscle cells. The majority of the modules, however, were enriched for metabolic pathways consisting of both nitrogen-related and glycolysis-related processes. Therefore, we explored correlations between nitrogen metabolism-related genes and coronary artery disease-associated genes and found significant correlations, suggesting the involvement of the nitrogen metabolism pathway in coronary artery disease pathogenesis. We also created gene regulatory networks enriched for genes in glycolysis and predicted key regulatory genes driving glycolysis dysregulation. CONCLUSIONS Our work suggests that dysregulation of vascular smooth muscle cell metabolism participates in phenotypic transitioning, which may contribute to disease progression, and suggests that AMT (aminomethyltransferase) and MPI (mannose phosphate isomerase) may play an important role in regulating nitrogen and glycolysis-related metabolism in smooth muscle cells.
Collapse
Affiliation(s)
- R. Noah Perry
- Center for Public Health Genomics (R.N.P., R.A., M.C.), University of Virginia, Charlottesville
- Department of Biomedical Engineering (R.N.P., D.A., M.C.), University of Virginia, Charlottesville
| | - Diana Albarracin
- Department of Biomedical Engineering (R.N.P., D.A., M.C.), University of Virginia, Charlottesville
| | - Redouane Aherrahrou
- Center for Public Health Genomics (R.N.P., R.A., M.C.), University of Virginia, Charlottesville
| | - Mete Civelek
- Center for Public Health Genomics (R.N.P., R.A., M.C.), University of Virginia, Charlottesville
- Department of Biomedical Engineering (R.N.P., D.A., M.C.), University of Virginia, Charlottesville
| |
Collapse
|
38
|
Li W, Jia Y, Gong Z, Dong Z, Yu F, Fu Y, Jiang C, Kong W. Ablation of the gut microbiota alleviates high-methionine diet-induced hyperhomocysteinemia and glucose intolerance in mice. NPJ Sci Food 2023; 7:36. [PMID: 37460578 DOI: 10.1038/s41538-023-00212-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 07/12/2023] [Indexed: 07/20/2023] Open
Abstract
A high-methionine (HM) diet leads to hyperhomocysteinemia (HHcy), while gastrointestinal tissue is an important site of net homocysteine (Hcy) production. However, the role of the gut microbiota in host HHcy remains obscure. This study aimed to determine whether gut microbiota ablation could alleviate host HHcy and glucose intolerance and reveal the underlying mechanism. The results showed that the HM diet-induced HHcy and glucose intolerance in mice, while antibiotic administration decreased the plasma level of Hcy and reversed glucose intolerance. HM diet increased intestinal epithelial homocysteine levels, while antibiotic treatment decreased intestinal epithelial homocysteine levels under the HM diet. Gut microbiota depletion had no effect on the gene expression and enzyme activity of CBS and BHMT in the livers of HM diet-fed mice. The HM diet altered the composition of the gut microbiota with marked increases in the abundances of Faecalibaculum and Dubosiella, which were also positively correlated with plasma Hcy concentrations. An in-depth analysis of the bacterial cysteine and methionine metabolism pathways showed that the abundances of two homocysteine biosynthesis-related KEGG orthologies (KOs) were markedly increased in the gut microbiota in HM diet-fed mice. Hcy was detected from Dubosiella newyorkensis-cultured supernatant by liquid chromatography-tandem mass spectrometry (LC‒MS) analysis. In conclusion, these findings suggested that the HM diet-induced HHcy and glucose intolerance in mice, by reshaping the composition of the gut microbiota, which might produce and secrete Hcy.
Collapse
Affiliation(s)
- Wenqiang Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yiting Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Ze Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Zhao Dong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Fang Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Third Hospital, Peking University, Beijing, China.
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China.
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| |
Collapse
|
39
|
Yao H, Wang F, Chong H, Wang J, Bai Y, Du M, Yuan X, Yang X, Wu M, Li Y, Pang H. A Curcumin-Modified Coordination Polymers with ROS Scavenging and Macrophage Phenotype Regulating Properties for Efficient Ulcerative Colitis Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300601. [PMID: 37195012 PMCID: PMC10323614 DOI: 10.1002/advs.202300601] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/01/2023] [Indexed: 05/18/2023]
Abstract
Overexpression of classically activated macrophages (M1) subtypes and assessed reactive oxygen species (ROS) levels are often observed in patients with ulcerative colitis. At present, the treatment system of these two problems has yet to be established. Here, the chemotherapy drug curcumin (CCM) is decorated with Prussian blue analogs in a straightforward and cost-saving manner. Modified CCM can be released in inflammatory tissue (acidic environment), eventually causing M1 macrophages to transform into M2 macrophages and inhibiting pro-inflammatory factors. Co(III) and Fe(II) have abundant valence variations, and the lower REDOX potential in CCM-CoFe PBA enables ROS clearance through multi-nanomase activity. In addition, CCM-CoFe PBA effectively alleviated the symptoms of UC mice induced by DSS and inhibited the progression of the disease. Therefore, the present material may be used as a new therapeutic agent for UC.
Collapse
Affiliation(s)
- Hang Yao
- School of Chemistry and Chemical EngineeringYangzhou UniversityJiangsu225002China
| | - Feifei Wang
- School of Chemistry and Chemical EngineeringYangzhou UniversityJiangsu225002China
| | - Hui Chong
- School of Chemistry and Chemical EngineeringYangzhou UniversityJiangsu225002China
| | - Jingjing Wang
- School of Chemistry and Chemical EngineeringYangzhou UniversityJiangsu225002China
| | - Yang Bai
- School of PharmacyChangzhou UniversityChangzhouJiangsu213164P. R. China
- State Key Laboratory of Coordination ChemistryNanjing UniversityNanjingJiangsu210023P. R. China
| | - Meng Du
- School of Chemistry and Chemical EngineeringYangzhou UniversityJiangsu225002China
| | - Xiaohui Yuan
- School of Chemistry and Chemical EngineeringYangzhou UniversityJiangsu225002China
| | - Xiaofei Yang
- School of Chemistry and Chemical EngineeringYangzhou UniversityJiangsu225002China
| | - Ming Wu
- School of Chemistry and Chemical EngineeringYangzhou UniversityJiangsu225002China
| | - Yuping Li
- Department of NeurosurgeryClinical Medical CollegeYangzhou UniversityJiangsu225002P. R. China
| | - Huan Pang
- School of Chemistry and Chemical EngineeringYangzhou UniversityJiangsu225002China
| |
Collapse
|
40
|
Jiang Q, Wei B, You M, Zhou X. d-mannose blocks the interaction between keratinocytes and Th17 cells to alleviate psoriasis by inhibiting HIF-1α/CCL20 in mice. Int Immunopharmacol 2023; 118:110087. [PMID: 37001381 DOI: 10.1016/j.intimp.2023.110087] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023]
Abstract
Psoriasis is an autoimmune chronic inflammatory skin disease with an unclear pathogenesis that is difficult to cure, causing serious physical and mental burdens for patients. Previous research showed that a mutually reinforcing vicious cycle caused by keratinocytes (KC) and a variety of immune cells plays an important role in psoriatic inflammation. d-Mannose, a widely distributed metabolite in the body, has been found to treat several metabolic diseases, but its impact on psoriasis remains unknown. Our study aims to investigate the effects of d-mannose on psoriasis and its specific mechanism. Here, we found that d-mannose alleviates psoriasis in mice both as oral and topical agents. Specifically, d-mannose down-regulated the expression of hypoxia-inducible factor 1A(HIF-1α) and inhibited the expression of chemokine CCL20 in keratinocytes, thereby inhibiting the local infiltration of Th17 cells and breaking the cycle of keratinocytes-Th17 cells. Overall, our study indicates that d-mannose alleviates cutaneous inflammation in psoriasis by inhibiting the HIF-1α/CCL20/Th17 cells axis, and d-mannose has the potential to be used as an oral and topical agent in the treatment of psoriasis.
Collapse
|
41
|
Jones KR, Hughey MC, Belden LK. Colonization order of bacterial isolates on treefrog embryos impacts microbiome structure in tadpoles. Proc Biol Sci 2023; 290:20230308. [PMID: 36946107 PMCID: PMC10031419 DOI: 10.1098/rspb.2023.0308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 03/06/2023] [Indexed: 03/23/2023] Open
Abstract
Priority effects, or impacts of colonization order, may have lasting influence on ecological community composition. The embryonic microbiome is subject to stochasticity in colonization order of bacteria. Stochasticity may be especially impactful for embryos developing in bacteria-rich environments, such as the embryos of many amphibians. To determine if priority effects experienced as embryos impacted bacterial community composition in newly hatched tadpoles, we selectively inoculated the embryos of laboratory-raised hourglass treefrogs, Dendropsophus ebraccatus, with bacteria initially isolated from the skin of wild D. ebraccatus adults over 2 days. First, embryos were inoculated with two bacteria in alternating sequences. Next, we evaluated the outcomes of priority effects in an in vitro co-culture assay absent of host factors. We then performed a second embryo experiment, inoculating embryos with one of three bacteria on the first day and a community of five target bacteria on the second. Through 16S rRNA gene amplicon sequencing, we observed relative abundance shifts in tadpole bacteria communities due to priority effects. Our results suggest that the initial bacterial source pools of embryos shape bacterial communities at later life stages; however, the magnitude of those changes is dependent on the host environment and the identity of bacterial colonists.
Collapse
Affiliation(s)
- Korin Rex Jones
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061-0131, USA
| | - Myra C. Hughey
- Department of Biology, Vassar College, Poughkeepsie, NY 12604, USA
| | - Lisa K. Belden
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061-0131, USA
| |
Collapse
|
42
|
Chen X, Cao J, Geng A, Zhang X, Wang H, Chu Q, Yan Z, Zhang Y, Liu H, Zhang J. Integration of GC-MS and LC-MS for metabolite characteristics of thigh meat between fast- and slow-growing broilers at marketable age. Food Chem 2023; 403:134362. [PMID: 36183464 DOI: 10.1016/j.foodchem.2022.134362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 09/07/2022] [Accepted: 09/18/2022] [Indexed: 11/26/2022]
|
43
|
Ye J, Yang H, Hu W, Tang K, Liu A, Bi S. Changed cecal microbiota involved in growth depression of broiler chickens induced by immune stress. Poult Sci 2023; 102:102598. [PMID: 36913756 PMCID: PMC10023976 DOI: 10.1016/j.psj.2023.102598] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
A previous study identified genes and metabolites associated with amino acid metabolism, glycerophospholipid metabolism, and inflammatory response in the liver of broilers with immune stress. The present research was designed to investigate the effect of immune stress on the cecal microbiome in broilers. In addition, the correlation between altered microbiota and liver gene expression, the correlation between altered microbiota and serum metabolites were compared using the Spearman correlation coefficients. Eighty broiler chicks were randomly assigned to 2 groups with 4 replicate pens per group and 10 birds per pen. The model broilers were intraperitoneally injected of 250 µg/kg LPS at 12, 14, 33, and 35 d of age to induce immunological stress. Cecal contents were taken after the experiment and kept at -80°C for 16S rDNA gene sequencing. Then the Pearson's correlation between gut microbiome and liver transcriptome, between gut microbiome and serum metabolites were calculated using R software. The results showed that immune stress significantly changed microbiota composition at different taxonomic levels. KEGG pathways analysis suggested that these gut microbiota were mainly involved in biosynthesis of ansamycins, glycan degradation, D-glutamine and D-glutamate metabolism, valine, leucine, and isoleucine biosynthesis and biosynthesis of vancomycin group antibiotics. Moreover, immune stress increased the activities of metabolism of cofactors and vitamins, as well as decreased the ability of energy metabolism and digestive system. Pearson's correlation analysis identified several bacteria were positively correlated with the gene expression while a few of bacteria were negatively correlated with the gene expression. The results identified potential microbiota involvement in growth depression mediated by immune stress and provided strategies such as supplement of probiotic for alleviating immune stress in broiler chickens.
Collapse
Affiliation(s)
- Jixuan Ye
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Rongchang, Chongqing, China
| | - Huaao Yang
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Rongchang, Chongqing, China
| | - Weidong Hu
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Rongchang, Chongqing, China
| | - Keyi Tang
- College of Life Sciences, Sichuan Normal University, Chengdu, Sichuan, China
| | - Anfang Liu
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Southwest University, Rongchang, Chongqing, China
| | - Shicheng Bi
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Rongchang, Chongqing, China.
| |
Collapse
|
44
|
Xiao P, Hu Z, Lang J, Pan T, Mertens RT, Zhang H, Guo K, Shen M, Cheng H, Zhang X, Cao Q, Ke Y. Mannose metabolism normalizes gut homeostasis by blocking the TNF-α-mediated proinflammatory circuit. Cell Mol Immunol 2023; 20:119-130. [PMID: 36471112 PMCID: PMC9887054 DOI: 10.1038/s41423-022-00955-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 11/07/2022] [Indexed: 12/11/2022] Open
Abstract
Mannose is a naturally occurring sugar widely consumed in the daily diet; however, mechanistic insights into how mannose metabolism affects intestinal inflammation remain lacking. Herein, we reported that mannose supplementation ameliorated colitis development and promoted colitis recovery. Macrophage-secreted inflammatory cytokines, particularly TNF-α, induced pathological endoplasmic reticulum stress (ERS) in intestinal epithelial cells (IECs), which was prevented by mannose via normalization of protein N-glycosylation. By preserving epithelial integrity, mannose reduced the inflammatory activation of colonic macrophages. On the other hand, mannose directly suppressed macrophage TNF-α production translationally by reducing the glyceraldehyde 3-phosphate level, thus promoting GAPDH binding to TNF-α mRNA. Additionally, we found dysregulated mannose metabolism in the colonic mucosa of patients with inflammatory bowel disease. Finally, we revealed that activating PMM2 activity with epalrestat, a clinically approved drug for the treatment of diabetic neuropathy, elicited further sensitization to the therapeutic effect of mannose. Therefore, mannose metabolism prevents TNF-α-mediated pathogenic crosstalk between IECs and intestinal macrophages, thereby normalizing aberrant immunometabolism in the gut.
Collapse
Affiliation(s)
- Peng Xiao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Pathology and Pathophysiology, and Department of Gastroenterology at Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Immunological Disease Research Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, China.
| | - Ziwei Hu
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaheng Lang
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianyuan Pan
- Department of General Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | - Huilun Zhang
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Ke Guo
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Immunological Disease Research Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Manlu Shen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Immunological Disease Research Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongqiang Cheng
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xue Zhang
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Qian Cao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Immunological Disease Research Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Yuehai Ke
- Department of Pathology and Pathophysiology, and Department of Gastroenterology at Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, Hangzhou, China.
| |
Collapse
|
45
|
Dhanalakshmi M, Sruthi D, Jinuraj KR, Das K, Dave S, Andal NM, Das J. Mannose: a potential saccharide candidate in disease management. Med Chem Res 2023; 32:391-408. [PMID: 36694836 PMCID: PMC9852811 DOI: 10.1007/s00044-023-03015-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 01/04/2023] [Indexed: 01/21/2023]
Abstract
There are a plethora of antibiotic resistance cases and humans are marching towards another big survival test of evolution along with drastic climate change and infectious diseases. Ever since the first antibiotic [penicillin], and the myriad of vaccines, we were privileged to escape many infectious disease threats. The survival technique of pathogens seems rapidly changing and sometimes mimicking our own systems in such a perfect manner that we are left unarmed against them. Apart from searching for natural alternatives, repurposing existing drugs more effectively is becoming a familiar approach to new therapeutic opportunities. The ingenious use of revolutionary artificial intelligence-enabled drug discovery techniques is coping with the speed of such alterations. D-Mannose is a great hope as a nutraceutical in drug discovery, against CDG, diabetes, obesity, lung disease, and autoimmune diseases and recent findings of anti-tumor activity make it interesting along with its role in drug delivery enhancing techniques. A very unique work done in the present investigation is the collection of data from the ChEMBL database and presenting the targetable proteins on pathogens as well as on humans. It shows Mannose has 50 targets and the majority of them are on human beings. The structure and conformation of certain monosaccharides have a decisive role in receptor pathogen interactions and here we attempt to review the multifaceted roles of Mannose sugar, its targets associated with different diseases, as a natural molecule having many success stories as a drug and future hope for disease management. Graphical abstract
Collapse
Affiliation(s)
- M. Dhanalakshmi
- Research and Development Centre, Bharathiar University, Coimbatore, 641046 Tamil Nadu India
| | - D. Sruthi
- Department of Biochemistry, Indian Institute of Science, Bengaluru, 560012 India
| | - K. R. Jinuraj
- OSPF-NIAS Drug Discovery Lab, NIAS, IISc Campus, Bengaluru, 560012 India
| | - Kajari Das
- Department of Biotechnology, College of Basic Science and Humanities, Odisha University of Agriculture and Technology, Bhubaneswar-3, Odisha India
| | - Sushma Dave
- Department of Applied Sciences, JIET, Jodhpur, Rajasthan India
| | - N. Muthulakshmi Andal
- Department of Chemistry, PSGR Krishnammal College for Women, Coimbatore, 641004 Tamil Nadu India
| | - Jayashankar Das
- Valnizen Healthcare, Vile Parle West, Mumbai, 400056 Maharashtra India
| |
Collapse
|
46
|
Chen J, Zeng P, Gong L, Zhang X, Ling Z, Bi K, Shi F, Wang K, Zhang Q, Jiang J, Zhang Y, Uede T, El-Omar EM, Diao H. Osteopontin Exacerbates High-Fat Diet-Induced Metabolic Disorders in a Microbiome-Dependent Manner. mBio 2022; 13:e0253122. [PMID: 36300928 PMCID: PMC9765578 DOI: 10.1128/mbio.02531-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 09/26/2022] [Indexed: 11/20/2022] Open
Abstract
The gut microbiome is involved in metabolic disorders. Osteopontin (OPN), as a key cytokine, contributes to various inflammation-related diseases. The underlying role of OPN in the microbiome remains poorly understood. Here, we investigated whether OPN could modulate metabolic disorders by affecting gut microbiota. In our present study, we found that the expression of OPN was elevated in individuals with obesity compared to that observed in healthy controls. There was a positive correlation between plasma OPN levels and body mass index (BMI) in humans. Moreover, OPN significantly exacerbated lipid accumulation and metabolic disorders in high-fat diet (HFD)-fed mice. Importantly, OPN significantly aggravated HFD-induced gut dysbiosis with a key signature profile. Fecal microbiota transplantation also supported the role of OPN in HFD-induced metabolic disorders in a microbiota-dependent manner. Moreover, the microbiome shift of OPN-deficient mice would be compensated to resemble those of wild-type mice by feeding with either OPN-containing milk or recombinant OPN protein in vivo. Furthermore, metagenomic analysis showed that OPN induced a higher abundance of Dorea and a lower abundance of Lactobacillus, which were positively and negatively correlated with body weight, respectively. Indeed, the abundance of Dorea was significantly decreased after Lactobacillus administration, suggesting that OPN may regulate the intestinal abundance of Dorea by reducing the colonization of Lactobacillus. We further confirmed that OPN decreased the adhesion of Lactobacillus to intestinal epithelial cells through the Notch signaling pathway. This study suggested that OPN could exacerbate HFD-induced metabolic dysfunctions through the OPN-induced alteration of the gut microbiome. Therefore, OPN could be a potential therapeutic target for metabolic syndrome. IMPORTANCE Gut microbiota are involved in metabolic disorders. However, microbiome-based therapeutic interventions are not always effective, which might be due to interference of the host factors. Here, we identified a strong positive correlation between OPN levels and BMI in humans. Next, we confirmed that OPN could aggravate high-fat diet-induced metabolic disorders in mice. Importantly, we found that fecal microbiota transplantation from OPN-deficient mice significantly alleviated metabolic disorders in WT mice. OPN directly induces the remodeling of the gut microbiota both in vitro and in vivo. These findings indicate that OPN could contribute to metabolic disorders by inducing an alteration of gut microbiota. OPN regulated the relative abundance of Lactobacillus by decreasing the adhesion of Lactobacillus to intestinal epithelial cells through the Notch signaling pathway. These data identify OPN as a potential pharmaceutical target for weight control and for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Jianing Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ping Zeng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lan Gong
- Microbiome Research Centre, St George and Sutherland Clinical School, University of New South Wales, Sydney, Australia
| | - Xujun Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zongxin Ling
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Kefan Bi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Fan Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Kaihang Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Qiong Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jingjing Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yanhui Zhang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| | - Toshimitsu Uede
- Division of Molecular Immunology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Emad M. El-Omar
- Microbiome Research Centre, St George and Sutherland Clinical School, University of New South Wales, Sydney, Australia
| | - Hongyan Diao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
47
|
Luo J, Li Y, Zhai Y, Liu Y, Zeng J, Wang D, Li L, Zhu Z, Chang B, Deng F, Zhang J, Zhou J, Sun L. D-Mannose ameliorates DNCB-induced atopic dermatitis in mice and TNF-α-induced inflammation in human keratinocytes via mTOR/NF-κB pathway. Int Immunopharmacol 2022; 113:109378. [DOI: 10.1016/j.intimp.2022.109378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/01/2022] [Accepted: 10/16/2022] [Indexed: 11/05/2022]
|
48
|
Chen S, Ren Z, Huo Y, Yang W, Peng L, Lv H, Nie L, Wei H, Wan C. Targeting the gut microbiota to investigate the mechanism of Lactiplantibacillus plantarum 1201 in negating colitis aggravated by a high-salt diet. Food Res Int 2022; 162:112010. [DOI: 10.1016/j.foodres.2022.112010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/29/2022]
|
49
|
Wang X, Li L, Bai M, Zhao J, Sun X, Gao Y, Yu H, Chen X, Zhang C. Dietary supplementation with Tolypocladium sinense mycelium prevents dyslipidemia inflammation in high fat diet mice by modulation of gut microbiota in mice. Front Immunol 2022; 13:977528. [PMID: 36420262 PMCID: PMC9677100 DOI: 10.3389/fimmu.2022.977528] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 10/19/2022] [Indexed: 03/09/2024] Open
Abstract
Obesity is a risk factor for many serious health problems, associated with inflammation, hyperlipidemia, and gut dysbiosis. Prevention of obesity is especially important for human health. Tolypocladium sinense is one of the fungi isolated from Chinese caterpillar fungus, which is a traditional Chinese medicine with putative gut microbiota modulation effects. Here, we established a high-fat diet (HFD)-induced hyperlipidemia mice model, which was supplemented with lyophilized T. sinense mycelium (TSP) daily to evaluate its anti-obesity effects. The results indicated that TSP supplementation can effectively alleviate the inflammatory response and oxidative stress levels caused by obesity. TSP significantly prevented obesity and suppressed dyslipidemia by regulating the expression of lipid metabolism genes in the liver. TSP is also effective in preventing the HFD-induced decline in short-chain fatty acid (SCFA) content. Gut microbiota profiling showed that TSP supplementation reversed HFD diet-induced bacterial abundance and also altered the metabolic pathways of functional microorganisms, as revealed by KEGG analysis. It is noteworthy that, correlation analysis reveals the up-regulated gut microbiota (Lactobacillus and Prevotella_9) are closely correlated with lipid metabolism parameters, gene expression of liver lipid metabolism and inflammatory. Additionally, the role of TSP in the regulation of lipid metabolism was reconfirmed by fecal microbiota transplantation. To sum up, our results provide the evidence that TSP may be used as prebiotic agents to prevent obesity by altering the gut microbiota, alleviating the inflammatory response and regulating gene expression of liver lipid metabolism.
Collapse
Affiliation(s)
- Xiaolong Wang
- Department of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Lin Li
- Department of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Mingjian Bai
- Department of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Jiaxin Zhao
- National & Local United Engineering Laboratory for Chinese Herbal Medicine Breeding and Cultivation, School of Life Sciences, Jilin University, Changchun, China
| | - Xiaojie Sun
- Department of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Yu Gao
- Department of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Haitao Yu
- Department of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Xia Chen
- National & Local United Engineering Laboratory for Chinese Herbal Medicine Breeding and Cultivation, School of Life Sciences, Jilin University, Changchun, China
| | - Chunjing Zhang
- Department of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| |
Collapse
|
50
|
Liu Q, An X, Chen Y, Deng Y, Niu H, Ma R, Zhao H, Cao W, Wang X, Wang M. Effects of Auricularia auricula Polysaccharides on Gut Microbiota and Metabolic Phenotype in Mice. Foods 2022; 11:foods11172700. [PMID: 36076885 PMCID: PMC9455240 DOI: 10.3390/foods11172700] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/17/2022] [Accepted: 09/01/2022] [Indexed: 11/26/2022] Open
Abstract
Personalized diets change the internal metabolism of organisms, which, in turn, affects the health of the body; this study was performed to explore the regulatory effects of polysaccharides extracted from Auricularia auricula on the overall metabolism and gut microbiota in normal C57BL/6J mice. The study was conducted using metabolomic and microbiomic methods to provide a scientific basis for further development and use of Auricularia auricula resources in the Qinba Mountains and in nutritional food with Auricularia auricula polysaccharides (AAP) as the main functional component. Based on LC-MS/MS metabolomic results, 51 AAP-regulated metabolites were found, mainly enriched in the arginine biosynthesis pathway, which had the highest correlation, followed by the following metabolisms: arginine and proline; glycine, serine and threonine; and glycerophospholipid, along with the sphingolipid metabolism pathway. Furthermore, supplementation of AAP significantly changed the composition of the mice intestinal flora. The relative abundance levels of Lactobacillus johnsonii, Weissella cibaria, Kosakonia cowanii, Enterococcus faecalis, Bifidobacterium animalis and Bacteroides uniformis were markedly up-regulated, while the relative abundance of Firmicutes bacterium M10-2 was down-regulated. The bioactivities of AAP may be related to the regulatory effects of endogenous metabolism and gut microbiota composition.
Collapse
Affiliation(s)
- Qian Liu
- College of Food Science and Technology, Northwest University, 229 Taibai North Road, Xi’an 710069, China
- Shaanxi Functional Food Engineering Center Co., Ltd., Xi’an 710069, China
- Correspondence: ; Tel./Fax: +86-29-88305208
| | - Xin An
- College of Food Science and Technology, Northwest University, 229 Taibai North Road, Xi’an 710069, China
| | - Yuan Chen
- College of Food Science and Technology, Northwest University, 229 Taibai North Road, Xi’an 710069, China
| | - Yuxuan Deng
- College of Food Science and Technology, Northwest University, 229 Taibai North Road, Xi’an 710069, China
| | - Haili Niu
- College of Food Science and Technology, Northwest University, 229 Taibai North Road, Xi’an 710069, China
| | - Ruisen Ma
- College of Food Science and Technology, Northwest University, 229 Taibai North Road, Xi’an 710069, China
| | - Haoan Zhao
- College of Food Science and Technology, Northwest University, 229 Taibai North Road, Xi’an 710069, China
| | - Wei Cao
- College of Food Science and Technology, Northwest University, 229 Taibai North Road, Xi’an 710069, China
| | - Xiaoru Wang
- Shaanxi Functional Food Engineering Center Co., Ltd., Xi’an 710069, China
| | - Meng Wang
- Shaanxi Functional Food Engineering Center Co., Ltd., Xi’an 710069, China
| |
Collapse
|