1
|
Gentili M, Sabbatini S, Nunzi E, Lusenti E, Cari L, Mencacci A, Ballet N, Migliorati G, Riccardi C, Ronchetti S, Monari C. Glucocorticoid-Induced Leucine Zipper Protein and Yeast-Extracted Compound Alleviate Colitis and Reduce Fungal Dysbiosis. Biomolecules 2024; 14:1321. [PMID: 39456254 PMCID: PMC11506796 DOI: 10.3390/biom14101321] [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: 09/17/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Inflammatory bowel diseases (IBD) have a complex, poorly understood pathogenesis and lack long-lasting effective treatments. Recent research suggests that intestinal fungal dysbiosis may play a role in IBD development. This study investigates the effects of the glucocorticoid-induced leucine zipper protein (GILZp)", known for its protective role in gut mucosa, and a yeast extract (Py) with prebiotic properties, either alone or combined, in DSS-induced colitis. Both treatments alleviated symptoms via overlapping or distinct mechanisms. In particular, they reduced the transcription levels of pro-inflammatory cytokines IL-1β and TNF-α, as well as the expression of the tight junction protein Claudin-2. Additionally, GILZp increased MUC2 transcription, while Py reduced IL-12p40 and IL-6 levels. Notably, both treatments were effective in restoring the intestinal burden of clinically important Candida and related species. Intestinal mycobiome analysis revealed that they were able to reduce colitis-associated fungal dysbiosis, and this effect was mainly the result of a decreased abundance of the Meyerozima genus, which was dominant in colitic mice. Overall, our results suggest that combined treatment regimens with GILZp and Py could represent a new strategy for the treatment of IBD by targeting multiple mechanisms, including the fungal dysbiosis.
Collapse
Affiliation(s)
- Marco Gentili
- Department of Medicine and Surgery, Pharmacology Division, University of Perugia, 06132 Perugia, Italy (E.L.); (L.C.); (G.M.); (C.R.)
| | - Samuele Sabbatini
- Department of Medicine and Surgery, Medical Microbiology Division, University of Perugia, 06132 Perugia, Italy; (S.S.); (A.M.)
| | - Emilia Nunzi
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy;
| | - Eleonora Lusenti
- Department of Medicine and Surgery, Pharmacology Division, University of Perugia, 06132 Perugia, Italy (E.L.); (L.C.); (G.M.); (C.R.)
| | - Luigi Cari
- Department of Medicine and Surgery, Pharmacology Division, University of Perugia, 06132 Perugia, Italy (E.L.); (L.C.); (G.M.); (C.R.)
| | - Antonella Mencacci
- Department of Medicine and Surgery, Medical Microbiology Division, University of Perugia, 06132 Perugia, Italy; (S.S.); (A.M.)
| | - Nathalie Ballet
- Lesaffre Institute of Science & Technology, Lesaffre International, 59700 Marcq-en-Baroeul, France;
| | - Graziella Migliorati
- Department of Medicine and Surgery, Pharmacology Division, University of Perugia, 06132 Perugia, Italy (E.L.); (L.C.); (G.M.); (C.R.)
| | - Carlo Riccardi
- Department of Medicine and Surgery, Pharmacology Division, University of Perugia, 06132 Perugia, Italy (E.L.); (L.C.); (G.M.); (C.R.)
| | - Simona Ronchetti
- Department of Medicine and Surgery, Pharmacology Division, University of Perugia, 06132 Perugia, Italy (E.L.); (L.C.); (G.M.); (C.R.)
| | - Claudia Monari
- Department of Medicine and Surgery, Medical Microbiology Division, University of Perugia, 06132 Perugia, Italy; (S.S.); (A.M.)
| |
Collapse
|
2
|
Kalashnikova IG, Nekrasova AI, Korobeynikova AV, Bobrova MM, Ashniev GA, Bakoev SY, Zagainova AV, Lukashina MV, Tolkacheva LR, Petryaikina ES, Nekrasov AS, Mitrofanov SI, Shpakova TA, Frolova LV, Bulanova NV, Snigir EA, Mukhin VE, Yudin VS, Makarov VV, Keskinov AA, Yudin SM. The Association between Gut Microbiota and Serum Biomarkers in Children with Atopic Dermatitis. Biomedicines 2024; 12:2351. [PMID: 39457662 PMCID: PMC11505256 DOI: 10.3390/biomedicines12102351] [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: 08/23/2024] [Revised: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Background. Currently, it is known that the gut microbiota plays an important role in the functioning of the immune system, and a rebalancing of the bacterial community can arouse complex immune reactions and lead to immune-mediated responses in an organism, in particular, the development of atopic dermatitis (AD). Cytokines and chemokines are regulators of the innate and adaptive immune response and represent the most important biomarkers of the immune system. It is known that changes in cytokine profiles are a hallmark of many diseases, including atopy. However, it remains unclear how the bacterial imbalance disrupts the function of the immune response in AD. Objectives. We attempted to determine the role of gut bacteria in modulating cytokine pathways and their role in atopic inflammation. Methods. We sequenced the 16S rRNA gene from 50 stool samples of children aged 3-12 years who had confirmed atopic dermatitis, and 50 samples from healthy children to serve as a control group. To evaluate the immune status, we conducted a multiplex immunofluorescence assay and measured the levels of 41 cytokines and chemokines in the serum of all participants. Results. To find out whether changes in the composition of the gut microbiota were significantly associated with changes in the level of inflammatory cytokines, a correlation was calculated between each pair of bacterial family and cytokine. In the AD group, 191 correlations were significant (Spearman's correlation coefficient, p ≤ 0.05), 85 of which were positive and 106 which were negative. Conclusions. It has been demonstrated that intestinal dysbiosis is associated with alterations in cytokine profiles, specifically an increase in proinflammatory cytokine concentrations. This may indicate a systemic impact of these conditions, leading to an imbalance in the immune system's response to the Th2 type. As a result, atopic conditions may develop. Additionally, a correlation between known AD biomarkers (IL-5, IL-8, IL-13, CCL22, IFN-γ, TNF-α) and alterations in the abundance of bacterial families (Pasteurellaceae, Barnesiellaceae, Eubacteriaceae) was observed.
Collapse
Affiliation(s)
- Irina G. Kalashnikova
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.I.N.); (A.V.K.); (M.M.B.); (G.A.A.); (S.Y.B.); (A.V.Z.); (M.V.L.); (L.R.T.); (E.S.P.); (A.S.N.); (S.I.M.); (T.A.S.); (L.V.F.); (N.V.B.); (E.A.S.); (V.E.M.); (V.S.Y.); (V.V.M.); (A.A.K.); (S.M.Y.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Li SY, Tong MM, Li L, Hui F, Meng FZ, Zhao YL, Guo YM, Guo XY, Shi BL, Yan SM. Rectal microbiomes and serum metabolomics reveal the improved effect of Artemisia ordosica crude polysaccharides on the lactation performance, antioxidant status, and immune responses of lactating donkeys. J Dairy Sci 2024; 107:6696-6716. [PMID: 38608958 DOI: 10.3168/jds.2023-24570] [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: 12/21/2023] [Accepted: 03/02/2024] [Indexed: 04/14/2024]
Abstract
This study is aimed at investigating the effects of dietary supplementation with Artemisia ordosica crude polysaccharides (AOCP) on lactation performance, antioxidant status, and immune status of lactating donkeys and analyzing rectal microbiomes and serum metabolomes. Fourteen lactating Dezhou donkeys with similar age (6.16 ± 0.67 yr of BW ± SD), weight (250.06 ± 25.18 kg), DIM (39.11 ± 7.42 d), and average parity of 3 were randomly allocated into 2 treatments: a control group (CON, basal diet) and an AOCP group (AOCP, basal diet with 1.0 g/kg DM AOCP). Ten weeks were allotted for the experiment, 2 wk for adaptation, and 8 wk for collecting data and samples. The results showed that supplementation of donkey diets with AOCP increased lactation performance, including DMI, milking yield, estimated milk yield, solids-corrected milk, ECM, milk fat yield, milk protein yield, milk lactose yield, milk TS yield, and milk SNF yield. The digestibility of DM, CP, ADF, and NDF was increased in the AOCP group compared with the CON group. The AOCP group increased the concentrations of IgA, IgG, and IgM, the activities of the superoxide dismutase, catalase, and total antioxidant capacity in the serum. Artemisia ordosica crude polysaccharides decreased the concentrations of tumor necrosis factor-α, nitric oxide, reactive oxygen species, and malondialdehyde in the serum. Compared with the CON group, AOCP increased propionate, butyrate, isovalerate, and total VFA concentrations in rectal feces (P < 0.05). The addition of AOCP to increased diversity (Shannon index) and altered structure of the rectal microflora. As a result of AOCP supplementation, there has been a significant improvement in the colonization of beneficial bacteria, including Lactobacillus, Unclassified_f_Prevotellacea, Ruminococcus, and Fibrobacter genera. In contrast, a decrease in the colonization of the Clostridium_sensu_stricto_1 bacterial genus and other pathogenic bacteria was observed. Meanwhile, metabolomics analysis found that AOCP supplementation upregulated metabolites l-tyrosine content while downregulating 9(S)-HODE, choline, sucrose, lysophosphatidylcholine (LysoPC) (18:0), LysoPC (18:1(9Z)), and LysoPC (20:2(11Z,14Z)) concentrations. These altered metabolites were involved in the PPAR signaling pathway, prolactin signaling pathway, glycerophospholipid metabolism, carbohydrate digestion and absorption, and tyrosine metabolism pathways, which were mainly related to antioxidant capacity, immune responses, and protein metabolism in the lactating donkeys. As a consequence of feeding AOCP diets, beneficial bacteria were abundant, and antioxidant and protein metabolism-related pathways were enriched, which may enhance lactation performance in donkeys. Therefore, supplementing AOCP diets is a desirable dietary strategy to improve donkey health and lactation performance.
Collapse
Affiliation(s)
- S Y Li
- College of Animal Science, Inner Mongolia Agricultural University, Key Laboratory of Animal Nutrition and Feed Science at Universities of Inner Mongolia Autonomous Region, Hohhot 010018, China
| | - M M Tong
- College of Animal Science, Inner Mongolia Agricultural University, Key Laboratory of Animal Nutrition and Feed Science at Universities of Inner Mongolia Autonomous Region, Hohhot 010018, China
| | - L Li
- College of Animal Science, Inner Mongolia Agricultural University, Key Laboratory of Animal Nutrition and Feed Science at Universities of Inner Mongolia Autonomous Region, Hohhot 010018, China
| | - F Hui
- College of Animal Science, Inner Mongolia Agricultural University, Key Laboratory of Animal Nutrition and Feed Science at Universities of Inner Mongolia Autonomous Region, Hohhot 010018, China
| | - F Z Meng
- College of Animal Science, Inner Mongolia Agricultural University, Key Laboratory of Animal Nutrition and Feed Science at Universities of Inner Mongolia Autonomous Region, Hohhot 010018, China
| | - Y L Zhao
- College of Animal Science, Inner Mongolia Agricultural University, Key Laboratory of Animal Nutrition and Feed Science at Universities of Inner Mongolia Autonomous Region, Hohhot 010018, China
| | - Y M Guo
- College of Animal Science, Inner Mongolia Agricultural University, Key Laboratory of Animal Nutrition and Feed Science at Universities of Inner Mongolia Autonomous Region, Hohhot 010018, China
| | - X Y Guo
- College of Animal Science, Inner Mongolia Agricultural University, Key Laboratory of Animal Nutrition and Feed Science at Universities of Inner Mongolia Autonomous Region, Hohhot 010018, China
| | - B L Shi
- College of Animal Science, Inner Mongolia Agricultural University, Key Laboratory of Animal Nutrition and Feed Science at Universities of Inner Mongolia Autonomous Region, Hohhot 010018, China
| | - S M Yan
- College of Animal Science, Inner Mongolia Agricultural University, Key Laboratory of Animal Nutrition and Feed Science at Universities of Inner Mongolia Autonomous Region, Hohhot 010018, China.
| |
Collapse
|
4
|
Li A, Liu A, Wang J, Song H, Luo P, Zhan M, Zhou X, Chen L, Zhang L. The prophylaxis functions of Lactobacillus fermentum GLF-217 and Lactobacillus plantarum FLP-215 on ulcerative colitis via modulating gut microbiota of mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:5816-5825. [PMID: 38406876 DOI: 10.1002/jsfa.13410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 12/09/2023] [Accepted: 02/18/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND The strong connection between gut microbes and human health has been confirmed by an increasing number of studies. Although probiotics have been found to relieve ulcerative colitis, the mechanism varies by the species involved. In this study, the physiological, immune and pathological factors of mice were measured and shotgun metagenomic sequencing was conducted to investigate the potential mechanisms in preventing ulcerative colitis. RESULTS The results demonstrated that ingestion of Lactobacillus fermentum GLF-217 and Lactobacillus plantarum FLP-215 significantly alleviated ulcerative colitis induced by dextran sulfate sodium (DSS), as evidenced by the increase in body weight, food intake, water intake and colon length as well as the decrease in disease activity index, histopathological score and inflammatory factor. Both strains not only improved intestinal mucosa by increasing mucin-2 and zonula occludens-1, but also improved the immune system response by elevating interleukin-10 levels and decreasing the levels of interleukin-1β, interleukin-6, tumor necrosis factor-α and interferon-γ. Moreover, L. fermentum GLF-217 and L. plantarum FLP-215 play a role in preventing DSS-induced colitis by regulating the structure of gut microbiota and promoting the formation of short-chain fatty acids. CONCLUSIONS This study may provide a reference for the prevention strategy of ulcerative colitis. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Ao Li
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou, China
| | | | - Jun Wang
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou, China
| | - Hainan Song
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou, China
| | | | | | | | | | - Lin Zhang
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou, China
| |
Collapse
|
5
|
Djouina M, Ollivier A, Waxin C, Kervoaze G, Pichavant M, Caboche S, Achour D, Grare C, Beury D, Hot D, Anthérieu S, Lo-Guidice JM, Dubuquoy L, Launay D, Vignal C, Gosset P, Body-Malapel M. Chronic Exposure to Both Electronic and Conventional Cigarettes Alters Ileum and Colon Turnover, Immune Function, and Barrier Integrity in Mice. J Xenobiot 2024; 14:950-969. [PMID: 39051349 PMCID: PMC11270428 DOI: 10.3390/jox14030053] [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: 05/30/2024] [Revised: 07/11/2024] [Accepted: 07/13/2024] [Indexed: 07/27/2024] Open
Abstract
Although the effects of cigarette smoke (CS) on the development of several intestinal diseases is well documented, the impact of e-cigarette aerosol (e-cig) on digestive health is largely unknown. To compare the effects of e-cig and CS on mouse ileum and colon, animals were chronically exposed for 6 months by nose-only inhalation to e-cig at 18 or 30 W power, or to 3R4F CS. Results showed that e-cig exposure decreased colon cell proliferation. Several other proliferative defects were observed in response to both e-cig and CS exposure, including up- and down-regulation of cyclin D1 protein levels in the ileum and colon, respectively. E-cig and CS exposure reduced myeloperoxidase activity in the ileum. In the colon, both exposures disrupted gene expression of cytokines and T cell transcription factors. For tight junction genes, ZO-1- and occludin-protein expression levels were reduced in the ileum and colon, respectively, by e-cig and CS exposure. The 16S sequencing of microbiota showed specific mild dysbiosis, according to the type of exposure. Overall, e-cig exposure led to altered proliferation, inflammation, and barrier function in both the ileum and colon, and therefore may be a gut hazard on par with conventional CS.
Collapse
Affiliation(s)
- Madjid Djouina
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE—Institute for Translational Research in Inflammation, F-59000 Lille, France; (M.D.); (C.W.); (L.D.); (D.L.); (C.V.)
| | - Anaïs Ollivier
- Univ. Lille, CNRS, INSERM, Institut Pasteur de Lille, CHU Lille, Center for Infection and Immunity of Lille (CIIL), UMR9017-U1019, F-59000 Lille, France; (A.O.); (G.K.); (M.P.); (P.G.)
| | - Christophe Waxin
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE—Institute for Translational Research in Inflammation, F-59000 Lille, France; (M.D.); (C.W.); (L.D.); (D.L.); (C.V.)
| | - Gwenola Kervoaze
- Univ. Lille, CNRS, INSERM, Institut Pasteur de Lille, CHU Lille, Center for Infection and Immunity of Lille (CIIL), UMR9017-U1019, F-59000 Lille, France; (A.O.); (G.K.); (M.P.); (P.G.)
| | - Muriel Pichavant
- Univ. Lille, CNRS, INSERM, Institut Pasteur de Lille, CHU Lille, Center for Infection and Immunity of Lille (CIIL), UMR9017-U1019, F-59000 Lille, France; (A.O.); (G.K.); (M.P.); (P.G.)
| | - Ségolène Caboche
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US41-UAR 2014-PLBS, F-59000 Lille, France; (S.C.); (D.B.); (D.H.)
| | - Djamal Achour
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483-IMPECS—IMPact de l’Environnement Chimique sur la Santé, F-59000 Lille, France; (D.A.); (C.G.); (S.A.); (J.-M.L.-G.)
| | - Céline Grare
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483-IMPECS—IMPact de l’Environnement Chimique sur la Santé, F-59000 Lille, France; (D.A.); (C.G.); (S.A.); (J.-M.L.-G.)
| | - Delphine Beury
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US41-UAR 2014-PLBS, F-59000 Lille, France; (S.C.); (D.B.); (D.H.)
| | - David Hot
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US41-UAR 2014-PLBS, F-59000 Lille, France; (S.C.); (D.B.); (D.H.)
| | - Sébastien Anthérieu
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483-IMPECS—IMPact de l’Environnement Chimique sur la Santé, F-59000 Lille, France; (D.A.); (C.G.); (S.A.); (J.-M.L.-G.)
| | - Jean-Marc Lo-Guidice
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483-IMPECS—IMPact de l’Environnement Chimique sur la Santé, F-59000 Lille, France; (D.A.); (C.G.); (S.A.); (J.-M.L.-G.)
| | - Laurent Dubuquoy
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE—Institute for Translational Research in Inflammation, F-59000 Lille, France; (M.D.); (C.W.); (L.D.); (D.L.); (C.V.)
| | - David Launay
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE—Institute for Translational Research in Inflammation, F-59000 Lille, France; (M.D.); (C.W.); (L.D.); (D.L.); (C.V.)
| | - Cécile Vignal
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE—Institute for Translational Research in Inflammation, F-59000 Lille, France; (M.D.); (C.W.); (L.D.); (D.L.); (C.V.)
| | - Philippe Gosset
- Univ. Lille, CNRS, INSERM, Institut Pasteur de Lille, CHU Lille, Center for Infection and Immunity of Lille (CIIL), UMR9017-U1019, F-59000 Lille, France; (A.O.); (G.K.); (M.P.); (P.G.)
| | - Mathilde Body-Malapel
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE—Institute for Translational Research in Inflammation, F-59000 Lille, France; (M.D.); (C.W.); (L.D.); (D.L.); (C.V.)
| |
Collapse
|
6
|
Mantri A, Klümpen L, Seel W, Krawitz P, Stehle P, Weber B, Koban L, Plassmann H, Simon MC. Beneficial Effects of Synbiotics on the Gut Microbiome in Individuals with Low Fiber Intake: Secondary Analysis of a Double-Blind, Randomized Controlled Trial. Nutrients 2024; 16:2082. [PMID: 38999830 PMCID: PMC11243043 DOI: 10.3390/nu16132082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/14/2024] Open
Abstract
Insufficient dietary fiber intake can negatively affect the intestinal microbiome and, over time, may result in gut dysbiosis, thus potentially harming overall health. This randomized controlled trial aimed to improve the gut microbiome of individuals with low dietary fiber intake (<25 g/day) during a 7-week synbiotic intervention. The metabolically healthy male participants (n = 117, 32 ± 10 y, BMI 25.66 ± 3.1 kg/m2) were divided into two groups: one receiving a synbiotic supplement (Biotic Junior, MensSana AG, Forchtenberg, Germany) and the other a placebo, without altering their dietary habits or physical activity. These groups were further stratified by their dietary fiber intake into a low fiber group (LFG) and a high fiber group (HFG). Stool samples for microbiome analysis were collected before and after intervention. Statistical analysis was performed using linear mixed effects and partial least squares models. At baseline, the microbiomes of the LFG and HFG were partially separated. After seven weeks of intervention, the abundance of SCFA-producing microbes significantly increased in the LFG, which is known to improve gut health; however, this effect was less pronounced in the HFG. Beneficial effects on the gut microbiome in participants with low fiber intake may be achieved using synbiotics, demonstrating the importance of personalized synbiotics.
Collapse
Affiliation(s)
- Aakash Mantri
- Institute of Nutrition and Food Science, Nutrition and Microbiota, University of Bonn, 53115 Bonn, Germany
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, 53127 Bonn, Germany
| | - Linda Klümpen
- Institute of Nutrition and Food Science, Nutrition and Microbiota, University of Bonn, 53115 Bonn, Germany
| | - Waldemar Seel
- Institute of Nutrition and Food Science, Nutrition and Microbiota, University of Bonn, 53115 Bonn, Germany
| | - Peter Krawitz
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, 53127 Bonn, Germany
| | - Peter Stehle
- Institute of Nutrition and Food Science, Nutritional Physiology, University of Bonn, 53115 Bonn, Germany
| | - Bernd Weber
- Institute of Experimental Epileptology and Cognition Research, University of Bonn, 53115 Bonn, Germany
- Center for Economics and Neuroscience, University of Bonn, 53113 Bonn, Germany
| | - Leonie Koban
- Lyon Neuroscience Research Center (CRNL), Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Université Claude Bernard Lyon 1, 69500 Lyon, France
- Institut Européen d‘Administration des Affaires (INSEAD), 77300 Paris, France
- Control-Interoception-Attention Team, Paris Brain Institute (ICM), 75013 Paris, France
| | - Hilke Plassmann
- Lyon Neuroscience Research Center (CRNL), Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Université Claude Bernard Lyon 1, 69500 Lyon, France
- Institut Européen d‘Administration des Affaires (INSEAD), 77300 Paris, France
| | - Marie-Christine Simon
- Institute of Nutrition and Food Science, Nutrition and Microbiota, University of Bonn, 53115 Bonn, Germany
| |
Collapse
|
7
|
Peng S, Lu X, Lin F, Mao N, Yu L, Zhu T, He J, Yang Y, Liu Z, Wang D. Rosa laevigata Polysaccharides Ameliorate Dextran Sulfate Sodium-Induced Ulcerative Colitis of Beagles through Regulating Gut Microbiota. Chem Biodivers 2024; 21:e202302102. [PMID: 38567653 DOI: 10.1002/cbdv.202302102] [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: 12/25/2023] [Revised: 03/15/2024] [Accepted: 03/28/2024] [Indexed: 04/04/2024]
Abstract
Rosa laevigata Michx. polysaccharides (RLP) have been demonstrated to possess antioxidant and anti-inflammatory properties. However, the mechanisms and efficacy of these polysaccharide components in preventing ulcerative colitis (UC) remain to be elucidated. The efficacy and mechanisms of RLP were investigated in a study that utilized healthy adult beagles to establish a UC model, considering the similarities in gut microbiota between humans and dogs. In the study, the beagle model induced by sodium dextran sulfate exhibited typical symptoms of ulcerative colitis, such as weight loss and diarrhea. All these symptoms and changes were significantly ameliorated through oral supplementation of RLP. Additionally, microbial community analysis based on the 16S rDNA gene revealed that RLP alleviated UC by increasing the abundance of beneficial bacteria and reducing the abundance of harmful bacteria. In conclusion, our study has provided that RLP effectively alleviated colitis by preserving the intestinal barrier and regulating the gut microbiota composition.
Collapse
Affiliation(s)
- Song Peng
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Xuanqi Lu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Fangzhu Lin
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Ningning Mao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Lin Yu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Tianyu Zhu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Jing He
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Yang Yang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Zhenguang Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| |
Collapse
|
8
|
Li Y, Liu J, Shi X, Li S, Zhang H, Zhang L, Huang X, Liu S, Wang W, Tian L, Zhang T, Du Z. Casein-quaternary chitosan complexes induced the soft assembly of egg white peptide and curcumin for ulcerative colitis alleviation. Int J Biol Macromol 2024; 269:132107. [PMID: 38710246 DOI: 10.1016/j.ijbiomac.2024.132107] [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: 10/15/2023] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
Soft assembly of peptide and curcumin (Cur) molecules enables functional integration by finding dynamic equilibrium states through non-covalent interactions. Herein, we developed two soft assembly systems, curcumin-egg white peptides (Cur-EWP) aggregations (AGs) and Cur-EWP-casein-quaternary chitosan (Cur-EWP-CA-QC) nanoparticles (NPs) to comparatively investigate their therapeutic effects on ulcerative colitis in mice and elucidate their underlying mechanism. Results revealed that Cur-EWP AGs, despite gastrointestinal tract instability, exhibited a propensity for swift accumulation within the colorectal region, enriching mucus-associated and short-chain fatty acid (SCAF)-producing bacteria, restoring the intestinal barrier damage. Whereas, Cur-EWP-CA-QC NPs, benefiting from their remarkable stability and exceptional mucosal adsorption properties, not only enhanced permeability of Cur and EWP in the small intestine to activate the immune response and boost tight junction protein expression but also, in their unabsorbed state, regulated the intestinal flora, exerting potent anti-inflammatory activity. Soft assembly of peptides and hydrophobic nutraceuticals could synergize biological activities to modulate chronic diseases.
Collapse
Affiliation(s)
- Yajuan Li
- Jilin Provincial Key Laboratory of Nutrition and Functional Food, College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China
| | - Jingbo Liu
- Jilin Provincial Key Laboratory of Nutrition and Functional Food, College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China
| | - Xiaoxia Shi
- Jilin Provincial Key Laboratory of Nutrition and Functional Food, College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China
| | - Shanglin Li
- Jilin Provincial Key Laboratory of Nutrition and Functional Food, College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China
| | - Hui Zhang
- Jilin Provincial Key Laboratory of Nutrition and Functional Food, College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China
| | - Leiyi Zhang
- Jilin Provincial Key Laboratory of Nutrition and Functional Food, College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China
| | - Xinyi Huang
- Jilin Provincial Key Laboratory of Nutrition and Functional Food, College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China
| | - Shuaiyan Liu
- Jilin Provincial Key Laboratory of Nutrition and Functional Food, College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China
| | - Weiyi Wang
- Jilin Provincial Key Laboratory of Nutrition and Functional Food, College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China
| | - Longjiang Tian
- Jilin Provincial Key Laboratory of Nutrition and Functional Food, College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China
| | - Ting Zhang
- Jilin Provincial Key Laboratory of Nutrition and Functional Food, College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China
| | - Zhiyang Du
- Jilin Provincial Key Laboratory of Nutrition and Functional Food, College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China.
| |
Collapse
|
9
|
Wang L, Li T, Cao WX, Zhao JY, Xu XH, Chai JP, Zhang JX, Liu J, Wang FC. To explore the mechanism of acupoint application in the treatment of primary dysmenorrhea by 16S rDNA sequencing and metabolomics. Front Endocrinol (Lausanne) 2024; 15:1397402. [PMID: 38872962 PMCID: PMC11169635 DOI: 10.3389/fendo.2024.1397402] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/16/2024] [Indexed: 06/15/2024] Open
Abstract
Graphene-based warm uterus acupoint paste (GWUAP) is an emerging non-drug alternative therapy for the treatment of primary dysmenorrhea (PD), but the underlying mechanism is still unclear. SD female rats were randomly divided into control group, model group and treatment group to explore the mechanism of GWUAP in the treatment of PD. Combined with 16S rDNA and fecal metabolomics, the diversity of microbiota and metabolites in each group was comprehensively evaluated. In this study, GWUAP reduced the torsion score of PD model rats, improved the pathological morphology of uterine tissue, reduced the pathological damage score of uterine tissue, and reversed the expression levels of inflammatory factors, pain factors and sex hormones. The 16 S rDNA sequencing of fecal samples showed that the abundance of Lactobacillus in the intestinal flora of the model group decreased and the abundance of Romboutsia increased, while the abundance of Lactobacillus in the intestinal flora of the treatment group increased and the abundance of Romboutsia decreased, which improved the imbalance of flora diversity in PD rats. In addition, 32 metabolites related to therapeutic effects were identified by metabolomics of fecal samples. Moreover, there is a close correlation between fecal microbiota and metabolites. Therefore, the mechanism of GWUAP in the treatment of PD remains to be further studied.
Collapse
Affiliation(s)
- Lin Wang
- Department of Acupuncture and Tuina, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Tie Li
- Department of Acupuncture and Tuina, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Wen-Xuan Cao
- Department of Pharmacy, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jin-Ying Zhao
- Department of Acupuncture and Tuina, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xiao-Hong Xu
- Department of Acupuncture and Tuina, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jia-Peng Chai
- Department of Acupuncture and Tuina, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jia-Xun Zhang
- Department of Acupuncture and Tuina, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jia Liu
- Department of Pharmacy, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Fu-Chun Wang
- Department of Acupuncture and Tuina, Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
10
|
Wei X, Ma N, Yang W, Tian J, Liu H, Fang H. Polyphenol Extracts from Ziziphus jujuba Mill. "Junzao" Attenuates Ulcerative Colitis by Inhibiting the NLRP3 and MAPKs Signaling Pathways and Regulating Gut Microbiota Homeostasis in Mice. Mol Nutr Food Res 2024; 68:e2300643. [PMID: 38600887 DOI: 10.1002/mnfr.202300643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/06/2024] [Indexed: 04/12/2024]
Abstract
SCOPE Polyphenols are the major active substances in red jujube fruit, and their anti-inflammatory and antioxidant activities suggest their potential utility in the prevention of ulcerative colitis (UC). METHODS AND RESULTS In this study, the effect of polyphenol extracts from red jujube (Ziziphus jujuba Mill. "Junzao") (PERJ) on the dextron sulfate sodium (DSS)-induced UC mice is investigated. The result shows that PERJ effectively improves clinical symptoms, including food and water intake, the disease activity insex (DAI) and spleen index, and routine blood levels, and alleviates the shortening of the colon, in mice with DSS-induced UC. Meanwhile, PERJ remarkably decreases the expression of proinflammatory factors. Moreover, PERJ repairs intestinal barrier damage by increasing the expression level of mucin 2 and mucin 3, and the result is also confirmed in the histological assessment. Besides, the expression levels of Nod-like receptor family pyrin domain-containing 3 (NLRP3) and mitogen-activated protein kinase cascade (MAPKs) signaling pathway-related proteins are inhibited by the PERJ administration. Finally, 16S rRNA sequencing analyses reveal that PERJ reverses intestinal microbiota dysbiosis by enhancing the abundance of Firmicutes and decreasing that of Proteobacteria and Bacteroidetes. CONCLUSION PERJ probably inhibits the development of UC by suppressing the NLRP3 and MAPKs signaling pathways and regulating gut microbiota homeostasis, and can be considered as a potential resource for preventing UC.
Collapse
Affiliation(s)
- Xiaobo Wei
- School of Food Science and Engineering, Ningxia University, Ningxia Key Laboratory for Food Microbial-Applications Technology and Safety, Control, Yinchuan, 750021, China
| | - Ni Ma
- School of Food Science and Engineering, Ningxia University, Ningxia Key Laboratory for Food Microbial-Applications Technology and Safety, Control, Yinchuan, 750021, China
| | - Wen Yang
- School of Food Science and Engineering, Ningxia University, Ningxia Key Laboratory for Food Microbial-Applications Technology and Safety, Control, Yinchuan, 750021, China
| | - Jinhu Tian
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agri-Food Processing, Integrated Research Base of Southern Fruit and Vegetable Preservation Technology, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Fuli Institute of Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Huiyan Liu
- School of Food Science and Engineering, Ningxia University, Ningxia Key Laboratory for Food Microbial-Applications Technology and Safety, Control, Yinchuan, 750021, China
| | - Haitian Fang
- School of Food Science and Engineering, Ningxia University, Ningxia Key Laboratory for Food Microbial-Applications Technology and Safety, Control, Yinchuan, 750021, China
| |
Collapse
|
11
|
Lee HY, Park YM, Shin DY, Hwang HM, Jeong H, Jeong SJ, Yang HJ, Ryu MS, Seo JW, Jeong DY, Kim BS, Kim JG. Gochujang, a traditional Korean fermented food, protects through suppressed inflammatory pathways and histological structure disruption in dextran sodium sulfate (DSS)-induced colitis mice. Heliyon 2024; 10:e27383. [PMID: 38515681 PMCID: PMC10955233 DOI: 10.1016/j.heliyon.2024.e27383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/23/2024] Open
Abstract
The mechanisms underlying chronic inflammatory diseases remain unclear. Therefore, researchers have explored the mechanisms underlying colitis using diverse materials. Recently, there has been an increasing interest in fermented products and bioconversion materials, their potential efficacy is being actively studied. Gochujang, a traditional Korean fermented product, is crafted by blending fermented Meju powder, gochu (Korean chili) powder, glutinous rice, and salt. In our study, we explored the effectiveness of Gochujang (500 mg/kg; Cheongju and Hongcheon, Korea) in dextran sulfate sodium (DSS)-induced colitis mice model. Gochujang was orally administered for 2 weeks, followed by the induction of colitis using 3% DSS in the previous week. During our investigation, Gochujang variants (TCG22-25, Cheongju and TCG22-48, Hongcheon) did not exhibit significant inhibition of weight reduction (p = 0.061) but notably (p = 0.001) suppressed the reduction in large intestine length in DSS-induced colitis mice. In the serum from colitis mice, TCG22-48 demonstrated reduced levels of the inflammatory cytokines interleukin (IL)-6 (p = 0.001) and tumor necrosis factor (TNF)-α (p = 0.001). Additionally, it inhibited the phosphorylation of Erk (p = 0.028), p38, and NF-κB (p = 0.001) the inflammatory mechanism. In our study, TCG22-25 demonstrated a reduction in the IL-6 level (p = 0.001) in serum and inhibited the phosphorylation of p38 and NF-κB (p = 0.001). Histological analysis revealed a significant (p = 0.001) reduction in the pathological score of the large intestine from TCG22-25 and TCG22-48. In conclusion, the intake of Gochujang demonstrates potent anti-inflammatory effects, mitigating colitis by preventing the large intestine length reduction of animals with colitis, lowering serum levels of TNF-α and IL-6 cytokines, and inhibiting histological disruption and inflammatory mechanism phosphorylation.
Collapse
Affiliation(s)
- Hak Yong Lee
- Invivo Co., Ltd., 121, Deahak-ro, Seongbuk-gu, Nonsan, 32992, South Korea
| | - Young Mi Park
- Invivo Co., Ltd., 121, Deahak-ro, Seongbuk-gu, Nonsan, 32992, South Korea
- Department of Pathology, College of Korean Medicine, Wonkwang University, Iksan, South Korea
| | - Dong Yeop Shin
- Invivo Co., Ltd., 121, Deahak-ro, Seongbuk-gu, Nonsan, 32992, South Korea
- Department of Integrated Life Science and Technology, Kongju National University, 32439, South Korea
| | - Hai Min Hwang
- Invivo Co., Ltd., 121, Deahak-ro, Seongbuk-gu, Nonsan, 32992, South Korea
| | - Hanna Jeong
- Invivo Co., Ltd., 121, Deahak-ro, Seongbuk-gu, Nonsan, 32992, South Korea
| | - Su-Ji Jeong
- Microbial Institute for Fermentation Industry (MIFI), Sunchang, 56048, South Korea
| | - Hee-Jong Yang
- Microbial Institute for Fermentation Industry (MIFI), Sunchang, 56048, South Korea
| | - Myeong Seon Ryu
- Microbial Institute for Fermentation Industry (MIFI), Sunchang, 56048, South Korea
| | - Ji Won Seo
- Microbial Institute for Fermentation Industry (MIFI), Sunchang, 56048, South Korea
| | - Do-Youn Jeong
- Microbial Institute for Fermentation Industry (MIFI), Sunchang, 56048, South Korea
| | - Byeong Soo Kim
- Department of Integrated Life Science and Technology, Kongju National University, 32439, South Korea
| | - Jae Gon Kim
- Invivo Co., Ltd., 121, Deahak-ro, Seongbuk-gu, Nonsan, 32992, South Korea
| |
Collapse
|
12
|
Fu Z, Yang X, Jiang Y, Mao X, Liu H, Yang Y, Chen J, Chen Z, Li H, Zhang XS, Mao X, Li N, Wang D, Jiang J. Microbiota profiling reveals alteration of gut microbial neurotransmitters in a mouse model of autism-associated 16p11.2 microduplication. Front Microbiol 2024; 15:1331130. [PMID: 38596370 PMCID: PMC11002229 DOI: 10.3389/fmicb.2024.1331130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/27/2024] [Indexed: 04/11/2024] Open
Abstract
The gut-brain axis is evident in modulating neuropsychiatric diseases including autism spectrum disorder (ASD). Chromosomal 16p11.2 microduplication 16p11.2dp/+ is among the most prevalent genetic copy number variations (CNV) linked with ASD. However, the implications of gut microbiota status underlying the development of ASD-like impairments induced by 16p11.2dp/+ remains unclear. To address this, we initially investigated a mouse model of 16p11.2dp/+, which exhibits social novelty deficit and repetitive behavior characteristic of ASD. Subsequently, we conducted a comparative analysis of the gut microbial community and metabolomic profiles between 16p11.2dp/+ and their wild-type counterparts using 16S rRNA sequencing and liquid chromatography-mass spectrometry (LC/MS). Our microbiota analysis revealed structural dysbiosis in 16p11.2dp/+ mice, characterized by reduced biodiversity and alterations in species abundance, as indicated by α/β-diversity analysis. Specifically, we observed reduced relative abundances of Faecalibaculum and Romboutsia, accompanied by an increase in Turicibacter and Prevotellaceae UCG_001 in 16p11.2dp/+ group. Metabolomic analysis identified 19 significantly altered metabolites and unveiled enriched amino acid metabolism pathways. Notably, a disruption in the predominantly histamine-centered neurotransmitter network was observed in 16p11.2dp/+ mice. Collectively, our findings delineate potential alterations and correlations among the gut microbiota and microbial neurotransmitters in 16p11.2dp/+ mice, providing new insights into the pathogenesis of and treatment for 16p11.2 CNV-associated ASD.
Collapse
Affiliation(s)
- Zhang Fu
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xiuyan Yang
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Youheng Jiang
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xinliang Mao
- Guangdong Perfect Life Health Science and Technology Research Institute Co., Ltd., Zhongshan, Guangdong, China
| | - Hualin Liu
- Guangdong Perfect Life Health Science and Technology Research Institute Co., Ltd., Zhongshan, Guangdong, China
| | - Yanming Yang
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Jia Chen
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Zhumei Chen
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-Sen University (SYSU), Shenzhen, Guangdong, China
| | - Huiliang Li
- Division of Medicine, Wolfson Institute for Biomedical Research, Faculty of Medical Sciences, University College London, London, United Kingdom
- China-UK Institute for Frontier Science, Shenzhen, Guangdong, China
| | - Xue-Song Zhang
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States
| | - Xinjun Mao
- Department of Anesthesiology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Ningning Li
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- China-UK Institute for Frontier Science, Shenzhen, Guangdong, China
| | - Dilong Wang
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jian Jiang
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| |
Collapse
|
13
|
Kumar M, Muthurayar T, Karthika S, Gayathri S, Varalakshmi P, Ashokkumar B. Anti-Diabetic Potentials of Lactobacillus Strains by Modulating Gut Microbiota Structure and β-Cells Regeneration in the Pancreatic Islets of Alloxan-Induced Diabetic Rats. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10221-7. [PMID: 38329697 DOI: 10.1007/s12602-024-10221-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2024] [Indexed: 02/09/2024]
Abstract
Diabetes mellitus, a most common endocrine disorder of glucose metabolism, has become a global epidemic and poses a serious public health threat with an increased socio-economic burden. Escalating incidence of diabetes is correlated with changes in lifestyle and food habits that cause gut microbiome dysbiosis and β-cells damage, which can be addressed with dietary interventions containing probiotics. Hence, the search for probiotics of human origin with anti-diabetic, anti-AGE, and anti-ACE potentials has gained renewed interest for the effective management of diabetes and its associated complications. The present study used an alloxan (AXN)-induced diabetic rat model to investigate the effects of potential probiotic Lacticaseibacillus casei MKU1, Lactiplantibacillus pentosus MKU3, and Lactiplantibacillus plantarum MKU7 administration individually on physiochemical parameters related to diabetic pathogenesis. Experimental animals were randomly allotted into six groups viz. NCG (control), DCG (AXN), DGM (metformin), DGP1 (MKU1), DGP2 (MKU3), and DGP3 (MKU7), and biochemical data like serum glucose, insulin, AngII, ACE, HbA1c, and TNF-α levels were measured until 90 days. Our results suggest that oral administration with MKU1, MKU3, or MKU7 significantly improved serum insulin levels, glycemic control, glucose tolerance, and body weight. Additionally, β-cell mass was increased by preserving islet integrity in Lactobacillus-treated diabetic rats, whereas TNF-α (~40%), AngII (~30%), and ACE levels (~50%) were strongly inhibited and enhanced sIgA production (5.8 folds) abundantly. Furthermore, Lactobacillus administration positively influenced the gut microbiome with a significant increase in the abundance of Lactobacillus species and the beneficial Bacteroides uniformis and Bacteroides fragilis, while decreased the pathogenic Proteus vulgaris and Parabacteroides distasonis. Among the probiotic treatment groups, L. pentosus MKU3 performed greatly in almost all parameters, indicating its potential use for alleviating diabetes-associated complications.
Collapse
Affiliation(s)
- Manoj Kumar
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India
| | - Tharmar Muthurayar
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India
| | - Sukumaran Karthika
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India
| | - Santhalingam Gayathri
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India
| | - Perumal Varalakshmi
- Department of Molecular Microbiology, School of Biotechnology, Madurai Kamaraj University, Madurai, India
| | - Balasubramaniem Ashokkumar
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India.
| |
Collapse
|
14
|
Wang M, Wang Z, Lessing DJ, Guo M, Chu W. Fusobacterium nucleatum and its metabolite hydrogen sulfide alter gut microbiota composition and autophagy process and promote colorectal cancer progression. Microbiol Spectr 2023; 11:e0229223. [PMID: 37889013 PMCID: PMC10714730 DOI: 10.1128/spectrum.02292-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/19/2023] [Indexed: 10/28/2023] Open
Abstract
IMPORTANCE Colorectal cancer (CRC) is the second most common cancer in the world; the main treatment for CRC is immunosuppressive therapy, but this therapy is only effective for a small percentage of CRC patients, so there is an urgent need for a treatment with fewer side effects and higher efficacy. This study demonstrated that Fusobacterium nucleatum with increased abundance in CRC can regulate the autophagy process and disrupt normal intestinal microbiota by producing hydrogen sulfide, factors that may be involved in the development and progression of CRC. This study may provide a reference for future CRC treatment options that are efficient and have fewer side effects.
Collapse
Affiliation(s)
- Minyu Wang
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zheng Wang
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Duncan James Lessing
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Min Guo
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Weihua Chu
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
15
|
Banc R, Rusu ME, Filip L, Popa DS. Phytochemical Profiling and Biological Activities of Quercus sp. Galls (Oak Galls): A Systematic Review of Studies Published in the Last 5 Years. PLANTS (BASEL, SWITZERLAND) 2023; 12:3873. [PMID: 38005770 PMCID: PMC10674842 DOI: 10.3390/plants12223873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/12/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023]
Abstract
Quercus species have been widely used in traditional medicine, and recently, researchers' attention has focused on galls of the genus Quercus as a source of health-promoting phytochemicals. This review presents a summary of the most recent findings on the phytochemistry and bioactivity of oak galls, following the screening of scientific papers published in two relevant databases, PubMed and Embase, between January 2018 and June 2023. The oak galls are rich in active compounds, mostly gallotannins and phenolic acids. Due to these secondary metabolites, the reviewed studies have demonstrated a wide range of biological activities, including antioxidant and anti-inflammatory actions, antimicrobial properties, tissue-protective effects, and antitumor, anti-aging, and hypoglycemic potential. Thus, oak galls are a promising natural matrix, to be considered in obtaining pharmaceutical and cosmetic preparations used in anti-aging strategies and, together with medications, in the management of age-related diseases. In further evaluations, the valuable functional properties of oak galls, reported mostly in preclinical studies, should be confirmed with clinical studies that would also take into account the potential health risks of their use.
Collapse
Affiliation(s)
- Roxana Banc
- Department of Bromatology, Hygiene, Nutrition, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania; (R.B.); (L.F.)
| | - Marius Emil Rusu
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 12 Ion Creangǎ Street, 400010 Cluj-Napoca, Romania
| | - Lorena Filip
- Department of Bromatology, Hygiene, Nutrition, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania; (R.B.); (L.F.)
| | - Daniela-Saveta Popa
- Department of Toxicology, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania;
| |
Collapse
|
16
|
Elzayat H, Mesto G, Al-Marzooq F. Unraveling the Impact of Gut and Oral Microbiome on Gut Health in Inflammatory Bowel Diseases. Nutrients 2023; 15:3377. [PMID: 37571313 PMCID: PMC10421146 DOI: 10.3390/nu15153377] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a complex disorder characterized by chronic inflammation of the gastrointestinal tract (GIT). IBD mainly includes two distinct diseases, namely Crohn's disease and ulcerative colitis. To date, the precise etiology of these conditions is not fully elucidated. Recent research has shed light on the significant role of the oral and gut microbiome in the development and progression of IBD and its collective influence on gut health. This review aims to investigate the connection between the oral and gut microbiome in the context of IBD, exploring the intricate interplay between these microbial communities and their impact on overall gut health. Recent advances in microbiome research have revealed a compelling link between the oral and gut microbiome, highlighting their pivotal role in maintaining overall health. The oral cavity and GIT are two interconnected ecosystems that harbor complex microbial communities implicated in IBD pathogenesis in several ways. Reduction in diversity and abundance of beneficial bacterial species with the colonization of opportunistic pathogens can induce gut inflammation. Some of these pathogens can arise from oral origin, especially in patients with oral diseases such as periodontitis. It is essential to discern the mechanisms of microbial transmission, the impact of oral health on the gut microbiome, and the potential role of dysbiosis in disease development. By elucidating this relationship, we can enhance our understanding of IBD pathogenesis and identify potential therapeutic avenues for managing the disease. Furthermore, innovative strategies for modulating the oral and gut microbiome can promote health and prevent disease occurrence and progression.
Collapse
Affiliation(s)
- Hala Elzayat
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 15551, United Arab Emirates
| | - Ghaidaa Mesto
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 15551, United Arab Emirates
| | - Farah Al-Marzooq
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 15551, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain 15551, United Arab Emirates
| |
Collapse
|
17
|
Guo M, Xing D, Wang J, Zhang Y, Li Z, Jiao X. Potent Intestinal Mucosal Barrier Enhancement of Nostoc commune Vaucher Polysaccharide Supplementation Ameliorates Acute Ulcerative Colitis in Mice Mediated by Gut Microbiota. Nutrients 2023; 15:3054. [PMID: 37447380 DOI: 10.3390/nu15133054] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Ulcerative colitis (UC) is evolving into a global burden with a substantially increasing incidence in developing countries. It is characterized by inflammation confined to mucosa and is recognized as an intestinal barrier disease. The intestinal microbiota plays a crucial role in UC pathogenesis. N. commune has long been appreciated as a healthy food and supplement worldwide and polysaccharides account for 60%. Here, we examined the amelioration of N. commune polysaccharides against acute colitis in mice induced by DSS and assessed the mediating role of gut microbiota. An integrated analysis of microbiome, metabolomics, and transcriptomics fully elaborated it markedly enhanced intestinal mucosal barrier function, including: increasing the relative abundance of Akkermansia muciniphila, uncultured_bacterium_g__norank_f__Muribaculaceae, and unclassified_g__norank_f__norank_o__Clostridia_UCG-014; decreasing microbiota-derived phosphatidylcholines and thromboxane 2 levels mapped to arachidonic acid metabolism; improving mucin2 biosynthesis and secretion; enhancing ZO-1 and occludin expression; reducing neutrophil infiltration; regulating the level of colitis-related inflammatory cytokines; involving inflammation and immune function-associated signaling pathways. Further, the mediation effect of gut microbiota was evaluated by administering a cocktail of antibiotics. In conclusion, our results demonstrated that N. commune polysaccharides predominantly reinforced the gut microbiota-mediated intestinal mucosal barrier to confer protection against UC and exhibited dramatic prebiotic-like functions, providing an alternative or complementary treatment for UC.
Collapse
Affiliation(s)
- Min Guo
- Department of Physiology, Key Laboratory of Physiology of Shanxi Province, Key Laboratory of Cellular Physiology of Ministry of Education, Shanxi Medical University, Taiyuan 030001, China
| | - Dehai Xing
- Department of Physiology, Key Laboratory of Physiology of Shanxi Province, Key Laboratory of Cellular Physiology of Ministry of Education, Shanxi Medical University, Taiyuan 030001, China
| | - Jin Wang
- Department of Physiology, Key Laboratory of Physiology of Shanxi Province, Key Laboratory of Cellular Physiology of Ministry of Education, Shanxi Medical University, Taiyuan 030001, China
| | - Ying Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
| | - Zhuoyu Li
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Xiangying Jiao
- Department of Physiology, Key Laboratory of Physiology of Shanxi Province, Key Laboratory of Cellular Physiology of Ministry of Education, Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
18
|
McGuire BD, Dees A, Hao L, Buckendahl P, Ogilvie AR, Sun H, Rezaee T, Barrett LO, Karim L, Dominguez-Bello MG, Bello NT, Shapses SA. A vitamin D deficient diet increases weight gain and compromises bone biomechanical properties without a reduction in BMD in adult female mice. J Steroid Biochem Mol Biol 2023; 231:106314. [PMID: 37088440 DOI: 10.1016/j.jsbmb.2023.106314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/27/2023] [Accepted: 04/20/2023] [Indexed: 04/25/2023]
Abstract
Vitamin D contributes to the development and maintenance of bone. Evidence suggests vitamin D status can also alter energy balance and gut health. In young animals, vitamin D deficiency (VDD) negatively affects bone mineral density (BMD) and bone microarchitecture, and these effects may also occur due to chronic ethanol intake. However, evidence is limited in mature models, and addressing this was a goal of the current study. Seven-month-old female C57BL/6 mice (n = 40) were weight-matched and randomized to one of four ad libitum diets: control, alcohol (Alc), vitamin D deficient (0 IU/d), or Alc+VDD for 8 weeks. A purified (AIN-93) diet was provided with water or alcohol (10 %) ad libitum. Body weight and food intake were recorded weekly, and feces were collected at 0, 4, and 8 weeks. At the age of 9 months, intestinal permeability was assessed by oral gavage of fluorescein isothiocyanate-dextran. Thereafter, bone mineral density (BMD) was measured by dual-energy X-ray absorptiometry. The microarchitecture of the distal femur was assessed by micro-computed tomography and biomechanical properties were evaluated by cyclic reference point indentation. VDD did not affect BMD or most bone microarchitecture parameters, however, the polar moment of inertia (p < 0.05) was higher in the VDD groups compared to vitamin D sufficient groups. VDD mice also had lower whole bone water content (p < 0.05) and a greater average unloading slope (p < 0.01), and energy dissipated (p < 0.01), indicating the femur displayed a brittle phenotype. In addition, VDD caused a greater increase in energy intake (p < 0.05), weight gain (p < 0.05), and a trend for higher intestinal permeability (p = 0.08). The gut microbiota of the VDD group had a reduction in alpha diversity (p < 0.05) and a lower abundance of ASVs from Rikenellaceae, Clostridia_UCG-014, Oscillospiraceae, and Lachnospiraceae (p < 0.01). There was little to no effect of alcohol supplementation on outcomes. Overall, these findings suggest that vitamin D deficiency causes excess weight gain and reduces the biomechanical strength of the femur as indicated by the higher average unloading slope and energy dissipated without an effect on BMD in a mature murine model.
Collapse
Affiliation(s)
- Brandon D McGuire
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, USA
| | - Azra Dees
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, USA
| | - Lihong Hao
- Department of Animal Sciences, Rutgers University, New Brunswick, NJ, USA
| | | | - Anna R Ogilvie
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, USA
| | - Haipeng Sun
- Department of Microbiology and Biochemistry, New Brunswick, NJ, USA
| | - Taraneh Rezaee
- Department of Bioengineering, University of Massachusetts Dartmouth, Dartmouth, MA 02747, USA
| | - Leland O Barrett
- Department of Bioengineering, University of Massachusetts Dartmouth, Dartmouth, MA 02747, USA
| | - Lamya Karim
- Department of Bioengineering, University of Massachusetts Dartmouth, Dartmouth, MA 02747, USA
| | - Maria Gloria Dominguez-Bello
- Department of Microbiology and Biochemistry, New Brunswick, NJ, USA; NJ Institute of Food, Nutrition and Health, Rutgers University, New Brunswick, NJ, USA
| | - Nicholas T Bello
- Department of Animal Sciences, Rutgers University, New Brunswick, NJ, USA; NJ Institute of Food, Nutrition and Health, Rutgers University, New Brunswick, NJ, USA
| | - Sue A Shapses
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, USA; NJ Institute of Food, Nutrition and Health, Rutgers University, New Brunswick, NJ, USA; Department of Medicine, Rutgers-Robert Wood Johnson Univ. Hospital, New Brunswick, NJ, USA.
| |
Collapse
|
19
|
Yi S, Zhang G, Liu M, Yu W, Cheng G, Luo L, Ning F. Citrus Honey Ameliorates Liver Disease and Restores Gut Microbiota in Alcohol-Feeding Mice. Nutrients 2023; 15:nu15051078. [PMID: 36904078 PMCID: PMC10005585 DOI: 10.3390/nu15051078] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Citrus honey (CH) is rich in nutrients that have a wide variety of biological functions, such as antibacterial, anti-inflammatory, and antioxidant activities, and which demonstrate therapeutic properties, such as anti-cancer and wound-healing abilities. However, the effects of CH on alcohol-related liver disease (ALD) and the intestinal microbiota remain unknown. This study aimed to determine the alleviating effects of CH on ALD and its regulatory effects on the gut microbiota in mice. In total, 26 metabolites were identified and quantified in CH, and the results suggested that the primary metabolites were abscisic acid, 3,4-dimethoxycinnamic acid, rutin, and two markers of CH, hesperetin and hesperidin. CH lowered the levels of aspartate aminotransferase, glutamate aminotransferase, and alcohol-induced hepatic edema. CH could promote the proliferation of Bacteroidetes while reducing the abundance of Firmicutes. Additionally, CH also showed some inhibitory effects on the growth of Campylobacterota and Turicibacter. CH enhanced the secretion of short-chain fatty acids (SCFAs), such as acetic acid, propionic acid, butyric acid, and valeric acid. Given its alleviating functions in liver tissue damage and its regulatory effects on the gut microbiota and SCFAs, CH could be a promising candidate for the therapeutic treatment of ALD.
Collapse
Affiliation(s)
- Shengxiang Yi
- School of Life Sciences, Nanchang University, Nanchang 330031, China
| | - Gaowei Zhang
- School of Life Sciences, Nanchang University, Nanchang 330031, China
| | - Mingyan Liu
- School of Life Sciences, Nanchang University, Nanchang 330031, China
| | - Wenjie Yu
- School of Life Sciences, Nanchang University, Nanchang 330031, China
- College of Food and Health, Beijing Technology and Business University (BTBU), Beijing 100048, China
| | - Guohua Cheng
- Agriculture and Rural Affairs Bureau of Nanfeng County, Fuzhou 344500, China
| | - Liping Luo
- School of Life Sciences, Nanchang University, Nanchang 330031, China
- College of Food and Health, Beijing Technology and Business University (BTBU), Beijing 100048, China
- Correspondence: (L.L.); (F.N.); Tel./Fax: +86-0791-83969519 (L.L.)
| | - Fangjian Ning
- School of Life Sciences, Nanchang University, Nanchang 330031, China
- College of Food and Health, Beijing Technology and Business University (BTBU), Beijing 100048, China
- Correspondence: (L.L.); (F.N.); Tel./Fax: +86-0791-83969519 (L.L.)
| |
Collapse
|
20
|
Wang F, Zhang M, Yuan M, Xia Z, Yang F, Zhang S, Lin T, Luo L, Tang J, Zhang Y. A novel sorbicillinoid compound as a potent anti-inflammation agent through inducing NLRP3 protein degradation. Br J Pharmacol 2023. [PMID: 36788033 PMCID: PMC10330222 DOI: 10.1111/bph.16058] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 12/14/2022] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND AND PURPOSE Chronic inflammation is pathogenic and contributes to human diseases, causing a significant threat to public health. The NLR family pyrin domain-containing protein 3 (NLRP3) is the best-characterized factor regulating inflammation. Therefore, targeting NLRP3 has the potential to treat inflammatory diseases and improve human health. EXPERIMENTAL APPROACH Lipopolysaccharide was used to induce inflammation in cell cultures. Lipopolysaccharide/d-galactosamine and dextran sulfate sodium salt were used to induce acute liver inflammation and ulcerative colitis respectively in C57BL/6J mice. Western blotting, immunofluorescence, immunoprecipitation, quantitative PCR and enzyme-linked immunosorbent assay (ELISA) were used to evaluate the activation of the inflammatory response in cell cultures and in mice. KEY RESULTS JNUTS013, a novel sorbicillinoid compound recently synthesized by us, significantly inhibited inflammation both in cell cultures and in mouse models. Mechanistically, JNUTS013 induced proteasome-dependent degradation of NLRP3. Hence, it suppressed the formation of the NLRP3 inflammasome and the production of downstream inflammatory cytokines and chemokines. The inhibitory effect of JNUTS013 on NLRP3 protein expression was confirmed in mice. Importantly, JNUTS013 failed to ameliorate bowel inflammation in Nlrp3-/- knockout mice, supporting NLRP3 as the biological target by which JNUTS013 inhibits inflammation. Further studies revealed critical chemical moieties of JNUTS013 required for inducing NLRP3 degradation. CONCLUSION AND IMPLICATIONS This study identifies a novel compound JNUTS013 that inhibits inflammation through inducing NLRP3 protein degradation in vitro and in vivo, which not only supports the development of JNUTS013 as an anti-inflammation agent but also creates a new way for the treatment of inflammation by chemically inducing NLRP3 degradation.
Collapse
Affiliation(s)
- Fangfang Wang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drug Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou, China.,Department of Pharmacology, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Meng Zhang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drug Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou, China
| | - Meng Yuan
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drug Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou, China
| | - Zixuan Xia
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drug Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou, China
| | - Fengge Yang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drug Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou, China
| | - Sihao Zhang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drug Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou, China
| | - Tengyu Lin
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Jinshan Tang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drug Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou, China
| | - Youwei Zhang
- Department of Pharmacology, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
21
|
Shen L, Shen Y, You L, Zhang Y, Su Z, Peng G, Deng J, Zuo Z, Zhong Z, Ren Z, Yu S, Zong X, Zhu Y, Cao S. Pueraria lobata polysaccharides alleviate neonatal calf diarrhea by modulating gut microbiota and metabolites. Front Vet Sci 2023; 9:1024392. [PMID: 36686167 PMCID: PMC9845628 DOI: 10.3389/fvets.2022.1024392] [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: 08/21/2022] [Accepted: 11/23/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction Neonatal calf diarrhea (NCD) is still one of the most critical diseases in calf rearing. Studies have shown that Pueraria lobata polysaccharides (PLP) have intense antioxidant and immunomodulatory activity and modulate gut microbiota. This randomized clinical trial aimed to determine the effect of PLP on the neonatal calf with diarrhea. Methods In this study, we recorded the fecal score of experimental calves, and calves with fecal scores ≥ 2 were determined as diarrhea and assessed their serum concentrations of inflammatory cytokines and oxidative damage-related indices. Fecal microbiota and metabolomics of diarrheal calves were further investigated. Results Results showed that treatment with PLP decreased the fecal score of diarrheal calves, serum concentrations of IL-1β, TNF-γ, and malondialdehyde, and also elevated the level of superoxide dismutase. In addition, PLP treatment altered the gut microbiota, significantly increased the relative abundances of beneficial bacteria, including the phyla Bacteroidetes and Actinobacteria, the genus Collinsella, Megamonas, and Bifidobacterium; decreased the relative abundances of pathogenetic or diarrhea related bacteria, such as Proteobacteria, Fusobacteria, Clostridium_sensu_stricto_1, and Escherichia_Shigella. Moreover, PLP can increase the fecal concentrations of isobutyric acid, propionic acid, and pantothenate; lower the levels of PC [18:0/18:1(9Z)], arachidonic acid, and docosahexaenoic acid. Discussion Thus, the results suggested that the PLP may perform the therapeutic activity via alleviating intestinal inflammation and regulating gut microbiota, avoiding further dysbiosis to restore the metabolism of gut microbiota, and finally promoting the recovery of diarrhea. The change further mitigated intestinal inflammation and oxidative damage in diarrheal calves. This indicated that PLP might be a promising treatment to attenuate diarrhea in neonatal calves.
Collapse
Affiliation(s)
- Liuhong Shen
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yu Shen
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Liuchao You
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yue Zhang
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zhetong Su
- Guangxi Innovates Medical Technology Co., Ltd., Lipu, Guangxi, China
| | - Guangneng Peng
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Junliang Deng
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zhicai Zuo
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zhijun Zhong
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zhihua Ren
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Shumin Yu
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xiaolan Zong
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yingkun Zhu
- School of Agriculture and Food Science, University College Dublin, Belfield, Ireland,*Correspondence: Yingkun Zhu ✉
| | - Suizhong Cao
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China,Suizhong Cao ✉
| |
Collapse
|
22
|
Huang L, Chen C. Employing pigs to decipher the host genetic effect on gut microbiome: advantages, challenges, and perspectives. Gut Microbes 2023; 15:2205410. [PMID: 37122143 PMCID: PMC10153013 DOI: 10.1080/19490976.2023.2205410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
The gut microbiota is a complex and diverse ecosystem comprised of trillions of microbes and plays an essential role in host's immunity, metabolism, and even behaviors. Environmental and host factors drive the huge variations in the gut microbiome among individuals. Here, we summarize accumulated evidences about host genetic effect on the gut microbial compositions with emphases on the correlation between host genetic kinship and the similarity of microbial compositions, heritability estimates of microbial taxa, and identification of genomic variants associated with the gut microbiome in pigs as well as in humans. A proportion of bacterial taxa have been reported to be heritable, and numerous variants associated with the diversity of the gut microbiota or specific taxa have been identified in both humans and pigs. LCT and ABO gene have been replicated in multiple studies, and its mechanism have been elucidated clearly. We also discuss the main advantages and challenges using pigs as experimental animals in exploring host genetic effect on the gut microbial composition and provided our insights on the perspectives in this area.
Collapse
Affiliation(s)
- Lusheng Huang
- National Key Laboratory of Pig Genetic Improvement, Jiangxi Agricultural University, Nanchang, China
| | - Congying Chen
- National Key Laboratory of Pig Genetic Improvement, Jiangxi Agricultural University, Nanchang, China
| |
Collapse
|
23
|
Li X, Xu S, Zhang Y, Li K, Gao XJ, Guo MY. Berberine Depresses Inflammation and Adjusts Smooth Muscle to Ameliorate Ulcerative Colitis of Cats by Regulating Gut Microbiota. Microbiol Spectr 2022; 10:e0320722. [PMID: 36287004 PMCID: PMC9769923 DOI: 10.1128/spectrum.03207-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 09/27/2022] [Indexed: 01/10/2023] Open
Abstract
Intestinal microbiota dysbiosis is a well established characteristic of ulcerative colitis (UC). Regulating the gut microbiota is an effective UC treatment strategy. Berberine (BBR), an alkaloid extracted from several Chinese herbs, is a common traditional Chinese medicine. To establish the efficacy and mechanism of action of BBR, we constructed a UC model using healthy adult shorthair cats to conduct a systematic study of colonic tissue pathology, inflammatory factor expression, and gut microbiota structure. We investigated the therapeutic capacity of BBR for regulating the gut microbiota and thus work against UC in cats using 16S rRNA genes amplicon sequencing technology. Our results revealed that dextran sulfate sodium (DSS)-induced cat models of UC showed weight loss, diarrhea accompanied by mucous and blood, histological abnormalities, and shortening of the colon, all of which were significantly alleviated by supplementation with BBR. A 16S rRNA gene-based microbiota analysis demonstrated that BBR could significantly benefit gut microbiota. Western blot, quantitative PCR, and enzyme-linked immunosorbent assays (ELISAs) showed that in DSS-induced cat models, the expression of the inflammatory factors was increased, activating the JAK2/STAT3 signaling pathway, and treatment with BBR reversed this effect. The myosin light chain (MLC) phosphorylation in the smooth muscle of the intestines is associated with motility of inflammation-related diarrhea in cats. This study used gut flora analyses to demonstrate the anti-UC effects of BBR and its potential therapeutic mechanisms and offers novel insights into the prevention of inflammatory diseases using natural products. IMPORTANCE Ulcerative colitis (UC) is common in clinics. Intestinal microbiota disorder is correlated with ulcerative colitis. Although there are many studies on ulcerative colitis in rats, there are few studies on colitis in cats. Therefore, this study explored the possibility of the use of BBR as a safe and efficient treatment for colitis in cats. The results demonstrated the therapeutic effects of BBR on UC based on the state of the intestinal flora. The study found BBR supplementation to be effective against dextran sulfate sodium (DSS)-induced colitis, smooth muscle damage, and gut microbiota dysbiosis.
Collapse
Affiliation(s)
- Xueying Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People’s Republic of China
| | - Shuang Xu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People’s Republic of China
| | - Yanhe Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People’s Republic of China
| | - Kan Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People’s Republic of China
| | - Xue-Jiao Gao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People’s Republic of China
| | - Meng-yao Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People’s Republic of China
| |
Collapse
|
24
|
Liu L, Sui W, Yang Y, Liu L, Li Q, Guo A. Establishment of an Enteric Inflammation Model in Broiler Chickens by Oral Administration with Dextran Sulfate Sodium. Animals (Basel) 2022; 12:ani12243552. [PMID: 36552471 PMCID: PMC9774581 DOI: 10.3390/ani12243552] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/10/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
This study aimed to evaluate the effectiveness of oral gavage of dextran sodium sulfate (DSS) to establish an enteric inflammation model in broilers. Forty 1-day-old male, yellow-feathered broilers were randomly divided into 2 groups with 5 replicates of 4 birds each for a 42-day trial. The experiment design used 2 groups: (1) the control group (CT), normal broilers fed a basal diet, and (2) the DSS group, DSS-treated broilers fed a basal diet. The DSS group received 1 mL of 2.5% DSS solution once a day by oral gavage from 21 to 29 days of age. The results showed that compared with those in CT, DSS treatment significantly increased histological scores for enteritis and mucosal damage at 29 and 42 days of age (p < 0.01) and the disease activity index (DAI) from 23 to 29 days of age (p < 0.01). DSS-treated broilers showed poor growth performance at 42 days of age, including decreased body weight and average daily gain and an increased feed conversion ratio (p < 0.01). DSS also caused gross lesions and histopathological damage in the jejunum of broilers, such as obvious hemorrhagic spots, loss of villus architecture, epithelial cell disruption, inflammatory cell infiltration, and decreased villus height. These results suggest that oral gavage of DSS is an effective method for inducing mild and non-necrotic enteric inflammation in broilers.
Collapse
Affiliation(s)
- Lixuan Liu
- Faculty of Life Sciences, Southwest Forestry University, No. 300, Bailong Road, Panlong District, Kunming 650224, China
| | - Wenjing Sui
- Faculty of Life Sciences, Southwest Forestry University, No. 300, Bailong Road, Panlong District, Kunming 650224, China
| | - Yajin Yang
- Faculty of Life Sciences, Southwest Forestry University, No. 300, Bailong Road, Panlong District, Kunming 650224, China
| | - Lily Liu
- Faculty of Life Sciences, Southwest Forestry University, No. 300, Bailong Road, Panlong District, Kunming 650224, China
| | - Qingqing Li
- Faculty of Life Sciences, Southwest Forestry University, No. 300, Bailong Road, Panlong District, Kunming 650224, China
- Kunming Xianghao Technology Co., Ltd., Kunming 650204, China
| | - Aiwei Guo
- Faculty of Life Sciences, Southwest Forestry University, No. 300, Bailong Road, Panlong District, Kunming 650224, China
- Correspondence:
| |
Collapse
|
25
|
Liu S, Yin R, Yang Z, Wei F, Hu J. The effects of rhein on D-GalN/LPS-induced acute liver injury in mice: Results from gut microbiome-metabolomics and host transcriptome analysis. Front Immunol 2022; 13:971409. [PMID: 36389730 PMCID: PMC9648667 DOI: 10.3389/fimmu.2022.971409] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 10/11/2022] [Indexed: 11/24/2022] Open
Abstract
Background Rhubarb is an important traditional Chinese medicine, and rhein is one of its most important active ingredients. Studies have found that rhein can improve ulcerative colitis by regulating gut microbes, but there are few reports on its effects on liver diseases. Therefore, this study aims to investigate these effects and underlying mechanisms. Methods Mice were given rhein (100 mg/kg), with both a normal control group and a model group receiving the same amount of normal saline for one week. Acute liver injury was induced in mice by intraperitoneal injection of D-GalN (800 mg/kg)/LPS (10 ug/kg). Samples (blood, liver, and stool) were then collected and assessed for histological lesions and used for 16S rRNA gene sequencing, high-performance liquid chromatography-mass spectrometry (LC-MS) and RNA-seq analysis. Results The levels of ALT and AST in the Model group were abnormal higher compared to the normal control group, and the levels of ALT and AST were significantly relieved in the rhein group. Hepatic HE staining showed that the degree of liver injury in the rhein group was lighter than that in the model group, and microbiological results showed that norank_o:Clostridia_UCG-014, Lachnoclostridium, and Roseburia were more abundant in the model group compared to the normal control group. Notably, the rhein treatment group showed reshaped disturbance of intestinal microbial community by D-GalN/LPS and these mice also had higher levels of Verrucomicrobia, Akkermansiaceae and Bacteroidetes. Additionally, There were multiple metabolites that were significantly different between the normal control group and the model group, such as L-α-amino acid, ofloxacin-N-oxide, 1-hydroxy-1,3-diphenylpropan-2-one,and L-4-hydroxyglutamate semialdehyde, but that returned to normal levels after rhein treatment. The gene expression level in the model group also changed significantly, various genes such as Cxcl2, S100a9, Tnf, Ereg, and IL-10 were up-regulated, while Mfsd2a and Bhlhe41 were down-regulated, which were recovered after rhein treatment. Conclusion Overall, our results show that rhein alleviated D-GalN/LPS-induced acute liver injury in mice. It may help modulate gut microbiota in mice, thereby changing metabolism in the intestine. Meanwhile, rhein also may help regulate genes expression level to alleviate D-GalN/LPS-induced acute liver injury.
Collapse
Affiliation(s)
- Shuhui Liu
- Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Ruiying Yin
- Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Ziwei Yang
- Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Feili Wei
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- *Correspondence: Feili Wei, ; Jianhua Hu,
| | - Jianhua Hu
- Beijing Youan Hospital, Capital Medical University, Beijing, China
- *Correspondence: Feili Wei, ; Jianhua Hu,
| |
Collapse
|
26
|
Yu T, Zhou Z, Liu S, Li C, Zhang ZW, Zhang Y, Jin W, Liu K, Mao S, Zhu L, Xie L, Wang G, Liang Y. The role of phosphatidylcholine 34:1 in the occurrence, development and treatment of ulcerative colitis. Acta Pharm Sin B 2022; 13:1231-1245. [PMID: 36970218 PMCID: PMC10031229 DOI: 10.1016/j.apsb.2022.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/16/2022] [Accepted: 08/23/2022] [Indexed: 11/29/2022] Open
Abstract
Lipid homeostasis is considered to be related to intestinal metabolic balance, while its role in the pathogenesis and treatment of ulcerative colitis (UC) remains largely unexplored. The present study aimed to identify the target lipids related to the occurrence, development and treatment of UC by comparing the lipidomics of UC patients, mice and colonic organoids with the corresponding healthy controls. Here, multi-dimensional lipidomics based on LC-QTOF/MS, LC-MS/MS and iMScope systems were constructed and used to decipher the alteration of lipidomic profiles. The results indicated that UC patients and mice were often accompanied by dysregulation of lipid homeostasis, in which triglycerides and phosphatidylcholines were significantly reduced. Notably, phosphatidylcholine 34:1 (PC34:1) was characterized by high abundance and closely correlation with UC disease. Our results also revealed that down-regulation of PC synthase PCYT1α and Pemt caused by UC modeling was the main factor leading to the reduction of PC34:1, and exogenous PC34:1 could greatly enhance the fumarate level via inhibiting the transformation of glutamate to N-acetylglutamate, thus exerting an anti-UC effect. Collectively, our study not only supplies common technologies and strategies for exploring lipid metabolism in mammals, but also provides opportunities for the discovery of therapeutic agents and biomarkers of UC.
Collapse
Affiliation(s)
- Tengjie Yu
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Zhihao Zhou
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Shijia Liu
- Affliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, China
| | - Changjian Li
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Zhi-Wei Zhang
- College of Chemical & Pharmaceutical Engineering, Hebei University of Science & Technology, Shijiazhuang 050018, China
| | - Yong Zhang
- College of Chemical & Pharmaceutical Engineering, Hebei University of Science & Technology, Shijiazhuang 050018, China
| | - Wei Jin
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Keanqi Liu
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Shuying Mao
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Lei Zhu
- Affliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, China
| | - Lin Xie
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Guangji Wang
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Corresponding authors. Tel./fax: +86 25 83271060.
| | - Yan Liang
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Corresponding authors. Tel./fax: +86 25 83271060.
| |
Collapse
|