1
|
Ren X, Shi B, Chang Z, Zhang J, Wang S, Liu R, Sang M, Dong H, Wu Q. Relationship between pathogenic E.coli O78-induced intestinal epithelial barrier damage and Zonulin expression levels in yaks. Front Cell Infect Microbiol 2024; 14:1456356. [PMID: 39376662 PMCID: PMC11456573 DOI: 10.3389/fcimb.2024.1456356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/03/2024] [Indexed: 10/09/2024] Open
Abstract
To explore whether the intestinal damage of yak colibacillosis resulted from the regulation of Zonulin expression by its pathogenic bacteria, the overexpression and interference plasmids of Zonulin were designed and cultured in Tranwell after cell transfection. Then qRT-PCR and Western blot were used to detect the results of cell transfection, 200 mL 1×105 CFU/mL E.coli O78 was added for 4 hours, transmembrane resistance was measured by transmembrane resistance meter, FD4 fluorescence concentration in the lower chamber was detected by enzyme labeling instrument, bacterial translocation was measured by CFU counting method, and epithelial mucin (MUC1, MUC2) and tight junction protein (FABP2, Occludin, ZO-1) were detected by qRT-PCR. Results The Zonulin gene overexpression and knockout cell lines were successfully constructed, the TEER value of the barrier of Zonulin overexpression cell lines began to decrease at 1 h after the addition of E.coli O78 and reached the lowest value at 4 h, and the TEER value of Zonulin interference cell lines decreased within 1-4 h after the addition of E.coli O78. At 4 h, the FD4 passing capacity of Zonulin overexpression cell lines was significantly higher than that of interfering cell lines, reaching twice as much as siRNA-1. The amount of bacterial translocation in overexpressed cell lines increased rapidly within 1-4 h, and the concentration of E.coli in the lower chamber was significantly higher than that in the siRNA-1 group at 4 h, but there was no significant change in the siRNA-1 group in the 1-4 h. There was no significant change in the mRNA level of MUC1 in Zonulin overexpression and interference cell lines after the addition of E.coli O78. In the overexpression group, the mRNA levels of MUC2, Occludin, and ZO-1 were significantly decreased, and the mRNA level of FABP2 was increased considerably. These results suggest stimulate epithelial cells to secrete Zonulin protein. Many Zonulin proteins regulate the opening of tight junction structures, reduce the transmembrane resistance of the cell barrier, and improve the permeability of the cell barrier and the amount of bacterial translocation.
Collapse
Affiliation(s)
- Xiaoli Ren
- Key Laboratory of Clinical Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| | - Bin Shi
- Key Laboratory of Clinical Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, China
- Institute of Animal Science, Xizang Academy of Agricultural and Animal Husbandry Sciences, Xizang Lhasa, China
| | - Zhenyu Chang
- Key Laboratory of Clinical Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| | - Jingbo Zhang
- Key Laboratory of Clinical Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| | - Shuo Wang
- Key Laboratory of Clinical Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| | - Ruidong Liu
- Key Laboratory of Clinical Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| | - Mudan Sang
- Key Laboratory of Clinical Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| | - Hailong Dong
- Key Laboratory of Clinical Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| | - Qingxia Wu
- Key Laboratory of Clinical Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| |
Collapse
|
2
|
Wang X, Lang F, Liu D. High-Salt Diet and Intestinal Microbiota: Influence on Cardiovascular Disease and Inflammatory Bowel Disease. BIOLOGY 2024; 13:674. [PMID: 39336101 PMCID: PMC11429420 DOI: 10.3390/biology13090674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/17/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024]
Abstract
Salt, or sodium chloride, is an essential component of the human diet. Recent studies have demonstrated that dietary patterns characterized by a high intake of salt can influence the abundance and diversity of the gut microbiota, and may play a pivotal role in the etiology and exacerbation of certain diseases, including inflammatory bowel disease and cardiovascular disease. The objective of this review is to synthesize the effects of elevated salt consumption on the gut microbiota, including its influence on gut microbial metabolites and the gut immune system. Additionally, this review will investigate the potential implications of these effects for the development of cardiovascular disease and inflammatory bowel disease. The findings of this study offer novel insights and avenues for the management of two common conditions with significant clinical implications.
Collapse
Affiliation(s)
- Xueyang Wang
- Queen Mary College, Nanchang University, Xuefu Road, Nanchang 330001, China; (X.W.); (F.L.)
| | - Fuyuan Lang
- Queen Mary College, Nanchang University, Xuefu Road, Nanchang 330001, China; (X.W.); (F.L.)
| | - Dan Liu
- Queen Mary College, Nanchang University, Xuefu Road, Nanchang 330001, China; (X.W.); (F.L.)
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| |
Collapse
|
3
|
Al-Quraishy S, Abdel-Gaber R, Alamari G, Meryk A, El-Ashram S, Al-Shaebi EM, Dkhil MA. Fighting eimeriosis by using the anti-eimerial and anti-apoptotic properties of rhatany root extract. Front Immunol 2024; 15:1430960. [PMID: 39055709 PMCID: PMC11269128 DOI: 10.3389/fimmu.2024.1430960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 06/12/2024] [Indexed: 07/27/2024] Open
Abstract
Background Over the last decade, extensive use of coccidiostats to treat and control Eimeria infection has developed drug resistance, prompting the search for new alternative therapies. Rhatany is proven to have various pharmacological properties. Objective The present study aimed to in vitro and in vivo evaluate the effect of Rhatany roots extract (RRE) as an anti-eimerial and anti-apoptotic agent against murine eimeriosis induced by Eimeria papillata. Methods Phytochemical screening by gas chromatography-mass spectrometry analysis (GC-MS) was used to detect active compounds in RRE. In vitro anti-eimerial activity of RRE (200, 100, 50 mg/ml), amprolium, phenol, Dettol™, and formalin were studied after incubation with non-sporulated Eimeria oocysts. For the in vivo study, twenty-five male C57BL/6 mice were randomly allocated into five groups. Animals in the first group were just given distilled H2O, while those in the second group were given 200 mg/kg RRE for 5 days. The Eimeria parasite's oocysts were infected into the third, fourth, and fifth groups. For treatment, RRE (200 mg/kg) and amprolium (120 mg/kg) were orally given to the 4th and 5th groups for five days, respectively. All mice were euthanized, on day 5 post-infection, to collect the jejunal tissues under study. Investigations were undertaken into the oocyst output in feces and goblet cells in mice jejuna. Assays for glutathione peroxidase (GPx), hydrogen peroxide (H2O2), and myeloperoxidase (MPO) were also performed. In jejunal tissue, cysteine aspartic acid protease-3 (Caspase-3) was counted using immunohistochemistry, while BCL2-associated X protein (Bax) and B-cell lymphoma-2 (BCL-2) were assayed using ELISA. In addition, mRNA expression of the goblet cell response gene (MUC2) was detected using real-time PCR. Results Phytochemical screening by GC-MS demonstrated the presence of 22 compounds in the RRE. The in vitro study revealed that RRE significantly inhabited the oocyst sporulation in a dose-dependent manner. By day 5 after infection with the Eimeria parasite, the number of oocysts in mice feces was significantly reduced after RRE treatment (1.308 × 106 ± 1.36 × 105 oocysts/g feces) compared to the infected group (5.387 × 106 ± 4.29 × 105 oocysts/g feces). Moreover, the Eimeria infection reduced the number of goblet cells of mice jejuna and its specific gene, MUC2. The treatment with RRE increased the number of goblet cells/villus from 3.45 ± 0.17 to 6.04 ± 0.23, associated with upregulation for MUC2 from 0.26 to 2.39-fold. Also, the Eimeria experimental infection lowered the activity of the antioxidant enzyme represented by GPx (23.99 ± 3.68 mg/g tissue), while increasing the stress parameters of hydrogen peroxide (0.07 ± 0.01 mM/g) as well as the activity of MPO (66.30 ± 3.74 U/mg). The production of apoptotic markers including Caspase-3 (68.89 ± 2.67 U/g) and Bax (159.05 ± 6.50 pg/ml) was significantly elevated while decreasing the anti-apoptotic marker of BCL2 (0.42 ± 0.07 pg/ml). Our study proved that RRE significantly reduced oxidative stress, and apoptotic markers as well as the inflammatory activity of MPO. Also, antioxidant enzyme and anti-apoptotic activity in the jejunum of E. papillata-infected mice were enhanced after RRE treatment. Conclusion Our study highlights the potential of RRE as a natural solution for coccidiosis management by modulating apoptosis in E. papillata host cells. However, further research is needed to fully understand the underlying mechanisms and enhance our understanding of its therapeutic efficacy.
Collapse
Affiliation(s)
- Saleh Al-Quraishy
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Rewaida Abdel-Gaber
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ghada Alamari
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Andreas Meryk
- Department of Pediatrics, Medical University of Innsbruck, Innsbruck, Austria
| | - Saeed El-Ashram
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
- Faculty of Science, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| | - Esam M. Al-Shaebi
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed A. Dkhil
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
- Applied Science Research Center, Applied Science Private University, Amman, Jordan
| |
Collapse
|
4
|
Gaur P, Rajendran Y, Srivastava B, Markandey M, Fishbain-Yoskovitz V, Mohapatra G, Suhail A, Chaudhary S, Tyagi S, Yadav SC, Pandey AK, Merbl Y, Bajaj A, Ahuja V, Srikanth C. Rab7-dependent regulation of goblet cell protein CLCA1 modulates gastrointestinal homeostasis. eLife 2024; 12:RP89776. [PMID: 38593125 PMCID: PMC11003743 DOI: 10.7554/elife.89776] [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] [Indexed: 04/11/2024] Open
Abstract
Inflammation in ulcerative colitis is typically restricted to the mucosal layer of distal gut. Disrupted mucus barrier, coupled with microbial dysbiosis, has been reported to occur prior to the onset of inflammation. Here, we show the involvement of vesicular trafficking protein Rab7 in regulating the colonic mucus system. We identified a lowered Rab7 expression in goblet cells of colon during human and murine colitis. In vivo Rab7 knocked down mice (Rab7KD) displayed a compromised mucus layer, increased microbial permeability, and depleted gut microbiota with enhanced susceptibility to dextran sodium-sulfate induced colitis. These abnormalities emerged owing to altered mucus composition, as revealed by mucus proteomics, with increased expression of mucin protease chloride channel accessory 1 (CLCA1). Mechanistically, Rab7 maintained optimal CLCA1 levels by controlling its lysosomal degradation, a process that was dysregulated during colitis. Overall, our work establishes a role for Rab7-dependent control of CLCA1 secretion required for maintaining mucosal homeostasis.
Collapse
Affiliation(s)
- Preksha Gaur
- Laboratory of Gut Inflammation and Infection Biology, Regional Centre for BiotechnologyFaridabadIndia
| | - Yesheswini Rajendran
- Laboratory of Gut Inflammation and Infection Biology, Regional Centre for BiotechnologyFaridabadIndia
| | | | - Manasvini Markandey
- Department of Gastroenterology, All India Institute of Medical SciencesDelhiIndia
| | | | | | - Aamir Suhail
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women’s Hospital, Massachusetts General Hospital and Harvard Medical SchoolBostonUnited States
| | - Shikha Chaudhary
- Department of Anatomy, All India Institute of Medical SciencesNew DelhiIndia
| | - Shaifali Tyagi
- Vaccine and Infectious Disease Research Center, Translational Health Science and Technology InstituteFaridabadIndia
| | | | - Amit Kumar Pandey
- Vaccine and Infectious Disease Research Center, Translational Health Science and Technology InstituteFaridabadIndia
| | - Yifat Merbl
- Department of Immunology, Weizmann Institute of ScienceRehovotIsrael
| | - Avinash Bajaj
- Laboratory of Gut Inflammation and Infection Biology, Regional Centre for BiotechnologyFaridabadIndia
| | - Vineet Ahuja
- Department of Gastroenterology, All India Institute of Medical SciencesDelhiIndia
| | - Chittur Srikanth
- Laboratory of Gut Inflammation and Infection Biology, Regional Centre for BiotechnologyFaridabadIndia
| |
Collapse
|
5
|
Segui-Perez C, Stapels DAC, Ma Z, Su J, Passchier E, Westendorp B, Wubbolts RW, Wu W, van Putten JPM, Strijbis K. MUC13 negatively regulates tight junction proteins and intestinal epithelial barrier integrity via protein kinase C. J Cell Sci 2024; 137:jcs261468. [PMID: 38345099 PMCID: PMC10984281 DOI: 10.1242/jcs.261468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/29/2024] [Indexed: 03/14/2024] Open
Abstract
Glycosylated mucin proteins contribute to the essential barrier function of the intestinal epithelium. The transmembrane mucin MUC13 is an abundant intestinal glycoprotein with important functions for mucosal maintenance that are not yet completely understood. We demonstrate that in human intestinal epithelial monolayers, MUC13 localized to both the apical surface and the tight junction (TJ) region on the lateral membrane. MUC13 deletion resulted in increased transepithelial resistance (TEER) and reduced translocation of small solutes. TEER buildup in ΔMUC13 cells could be prevented by addition of MLCK, ROCK or protein kinase C (PKC) inhibitors. The levels of TJ proteins including claudins and occludin were highly increased in membrane fractions of MUC13 knockout cells. Removal of the MUC13 cytoplasmic tail (CT) also altered TJ composition but did not affect TEER. The increased buildup of TJ complexes in ΔMUC13 and MUC13-ΔCT cells was dependent on PKC. The responsible PKC member might be PKCδ (or PRKCD) based on elevated protein levels in the absence of full-length MUC13. Our results demonstrate for the first time that a mucin protein can negatively regulate TJ function and stimulate intestinal barrier permeability.
Collapse
Affiliation(s)
- Celia Segui-Perez
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Daphne A. C. Stapels
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Ziliang Ma
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
- Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research (A*STAR), 138648 Singapore, Singapore
- Department of Pharmacy, National University of Singapore, 117543 Singapore, Singapore
| | - Jinyi Su
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Elsemieke Passchier
- UMAB, Department of Laboratory Pharmacy and Biomedical Genetics, Center for Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Bart Westendorp
- Department of Biomolecular Health Sciences, Division of Cell Biology, Metabolism and Cancer, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Richard W. Wubbolts
- Department of Biomolecular Health Sciences, Division of Cell Biology, Metabolism and Cancer, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Wei Wu
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
- Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research (A*STAR), 138648 Singapore, Singapore
- Department of Pharmacy, National University of Singapore, 117543 Singapore, Singapore
| | - Jos P. M. van Putten
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Karin Strijbis
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| |
Collapse
|
6
|
Falco EC, Ribaldone DG, Canavese G. Hyper Mucinous Proliferations in the Mucosa of Patients with Inflammatory Bowel Disease: Histological Lesions with a Real Potential for Neoplastic Evolution? Diagnostics (Basel) 2024; 14:499. [PMID: 38472971 DOI: 10.3390/diagnostics14050499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/15/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND AND AIMS Mucin disfunction is a critical event in the pathogenesis of inflammatory bowel disease (IBD). Although hyper mucinous conditions have a still debated implication in the clinical evolution of this disorder, hyper mucinous villous proliferations were found to have a preneoplastic biologic potential. We studied morphologic and immunophenotypic characteristics of these lesions in ileocolonic resections for IBD to add evidence about the evolutive potential of these lesions in samples with well oriented wall structures. METHODS Morphologic characteristics of bowel samples from 20 patients resected for IBD and with raised lesions at gross examination were studied and sections from cases with hyper mucinous lesions were stained with the following antibodies: Ki 67, p21, and p27, which were employed to evaluate the characteristics of the proliferative and differentiative activity of the epithelial structures; mismatch repair proteins and p53 have been studied as proteins implicated in carcinogenesis in IBD-affected mucosa; mucins subtypes in hyper mucinous structures were evaluated with MUC-2 and MUC-6. The results in 11 cases of saplings were that they harbored hyper mucinous proliferations. The occurrence of hyper mucinous structures was not related to dysplastic lesions, pseudo pyloric metaplasia, subtype of disease, or activity. In only one of our cases, mild cytologic atypia in the proliferative compartment was detected. Proliferation markers (Ki 67, p53) were expressed in the proliferative compartments of mucosal crypts and antiproliferative proteins p21 and p27 were expressed in differentiated epithelium. MMR proteins expression was limited to the proliferative compartment of the hyper mucinous projections. Mucin subtypes distribution was regular in the epithelium of hyper mucinous proliferations. CONCLUSIONS The present monocentric retrospective study was conducted on surgical samplings with well oriented crypts. Collected data show that hyper mucinous features are frequent occurrences in raised lesions in IBD patients. In hyper mucinous proliferations of the selected cases, the status of the proliferative cycle, the expression of the proteins most frequently involved in carcinogenetic pathways of mucosa affected by IBD, and the mucins subtypes expression have no evident anomalies. Findings are not consistent with the increased risk of neoplastic evolution observed in other studies; rather, they suggest a hyperplastic nature. However, the capacity of hyper mucinous raised lesions for neoplastic evolution should be ruled out with more extensive prospective studies to identify functional defects that could explain the hypothesized neoplastic potential.
Collapse
Affiliation(s)
| | | | - Gabriella Canavese
- Department of Pathology, Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| |
Collapse
|
7
|
Leoncini G, Cari L, Ronchetti S, Donato F, Caruso L, Calafà C, Villanacci V. Mucin Expression Profiles in Ulcerative Colitis: New Insights on the Histological Mucosal Healing. Int J Mol Sci 2024; 25:1858. [PMID: 38339134 PMCID: PMC10855303 DOI: 10.3390/ijms25031858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
A structural weakness of the mucus barrier (MB) is thought to be a cause of ulcerative colitis (UC). This study aims to investigate the mucin (MUC) composition of MB in normal mucosa and UC. Ileocolonic biopsies were taken at disease onset and after treatment in 40 patients, including 20 with relapsing and 20 with remitting UC. Ileocolonic biopsies from 10 non-IBD patients were included as controls. Gut-specific MUC1, MUC2, MUC4, MUC5B, MUC12, MUC13, MUC15, and MUC17 were evaluated immunohistochemically. The promoters of mucin genes were also examined. Normal mucosa showed MUC2, MUC5B, and MUC13 in terminal ileum and colon, MUC17 in ileum, and MUC1, MUC4, MUC12, and MUC15 in colon. Membranous, cytoplasmic and vacuolar expressions were highlighted. Overall, the mucin expression was abnormal in UC. Derangements in MUC1, MUC4, and MUC5B were detected both at onset and after treatment. MUC2 and MUC13 were unaffected. Sequence analysis revealed glucocorticoid-responsive elements in the MUC1 promoter, retinoic-acid-responsive elements in the MUC4 promoter, and butyrate-responsive elements in the MUC5B promoter. In conclusion, MUCs exhibited distinct expression patterns in the gut. Their expression was disrupted in UC, regardless of the treatment protocols. Abnormal MUC1, MUC4, and MUC5B expression marked the barrier dysfunction in UC.
Collapse
Affiliation(s)
- Giuseppe Leoncini
- First Pathology Division, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Luigi Cari
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Simona Ronchetti
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Francesco Donato
- Unit of Hygiene, Epidemiology and Public Health, University of Brescia, 25123 Brescia, Italy
| | - Laura Caruso
- Pathology Unit, Department of Pathology and Laboratory Medicine, Desenzano del Garda Hospital, ASST del Garda, 25015 Brescia, Italy
| | - Cristina Calafà
- Pathology Unit, Department of Pathology and Laboratory Medicine, Desenzano del Garda Hospital, ASST del Garda, 25015 Brescia, Italy
| | | |
Collapse
|
8
|
Kim YM, Kim HY, Jang JT, Hong S. Preventive Effect of Ecklonia cava Extract on DSS-Induced Colitis by Elevating Intestinal Barrier Function and Improving Pathogenic Inflammation. Molecules 2023; 28:8099. [PMID: 38138587 PMCID: PMC10745772 DOI: 10.3390/molecules28248099] [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: 11/21/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Inflammatory bowel disease (IBD), including ulcerative colitis and Crohn's disease, is a complex gastrointestinal disorder with a multifactorial etiology, including environmental triggers, autoimmune mechanisms, and genetic predisposition. Despite advancements in therapeutic strategies for IBD, its associated mortality rate continues to rise, which is often attributed to unforeseen side effects of conventional treatments. In this context, we explored the potential of Ecklonia cava extract (ECE), derived from an edible marine alga known for its anti-inflammatory and antioxidant properties, in mitigating IBD. This study investigated the effectiveness of ECE as a preventive agent in a murine model of dextran sulfate sodium (DSS)-induced colitis. Our findings revealed that pretreatment with ECE significantly ameliorated colitis severity, as evidenced by increased colon length, reduced spleen weight, and histological improvements demonstrated by immunohistochemical analysis. Furthermore, ECE significantly attenuated the upregulation of inflammatory cytokines and mediators and the infiltration of immune cells known to be prominent features of colitis in mice. Notably, ECE alleviated dysbiosis of intestinal microflora and aided in the recovery of damaged intestinal mucosa. Mechanistically, ECE exhibited protective effects against pathogenic colitis by inhibiting the NLRP3/NF-κB pathways known to be pivotal regulators in the inflammatory signaling cascade. These compelling results suggest that ECE holds promise as a potential candidate for IBD prevention. It might be developed into a functional food for promoting gastrointestinal health. This research sheds light on the preventive potential of natural compounds like ECE in the management of IBD, offering a safer and more effective approach to combating this challenging disease.
Collapse
Affiliation(s)
- Young-Mi Kim
- Lee Gil Ya Cancer and Diabetes Institute, Department of Biochemistry, Gachon University College of Medicine, Incheon 21999, Republic of Korea; (Y.-M.K.); (H.-Y.K.)
| | - Hye-Youn Kim
- Lee Gil Ya Cancer and Diabetes Institute, Department of Biochemistry, Gachon University College of Medicine, Incheon 21999, Republic of Korea; (Y.-M.K.); (H.-Y.K.)
| | - Ji-Tae Jang
- Aqua Green Technology Co., Ltd., Smart Building, Jeju Science Park, Jeju 63309, Republic of Korea;
| | - Suntaek Hong
- Lee Gil Ya Cancer and Diabetes Institute, Department of Biochemistry, Gachon University College of Medicine, Incheon 21999, Republic of Korea; (Y.-M.K.); (H.-Y.K.)
| |
Collapse
|
9
|
Zong Y, Meng J, Mao T, Han Q, Zhang P, Shi L. Repairing the intestinal mucosal barrier of traditional Chinese medicine for ulcerative colitis: a review. Front Pharmacol 2023; 14:1273407. [PMID: 37942490 PMCID: PMC10628444 DOI: 10.3389/fphar.2023.1273407] [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: 08/06/2023] [Accepted: 10/10/2023] [Indexed: 11/10/2023] Open
Abstract
Damage to the intestinal mucosal barrier play an important role in the pathogenesis of ulcerative colitis (UC). Discovering the key regulators and repairing the disturbed barrier are crucial for preventing and treating UC. Traditional Chinese medicine (TCM) has been proved to be effective on treating UC and has exhibited its role in repairing the intestinal mucosal barrier. We summarized the evidence of TCM against UC by protecting and repairing the physical barrier, chemical barrier, immune barrier, and biological barrier. Mechanisms of increasing intestinal epithelial cells, tight junction proteins, and mucins, promoting intestinal stem cell proliferation, restoring the abundance of the intestinal microbiota, and modulating the innate and adaptive immunity in gut, were all involved in. Some upstream proteins and signaling pathways have been elucidated. Based on the existing problems, we suggested future studies paying attention to patients' samples and animal models of UC and TCM syndromes, conducting rescue experiments, exploring more upstream regulators, and adopting new technical methods. We hope this review can provide a theoretical basis and novel ideas for clarifying the mechanisms of TCM against UC via repairing the intestinal mucosal barrier.
Collapse
Affiliation(s)
- Yichen Zong
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Meng
- Department of Gastroenterology and Hepatology, Beijing University of Chinese Medicine Affiliated Dongfang Hospital, Beijing, China
| | - Tangyou Mao
- Department of Gastroenterology and Hepatology, Beijing University of Chinese Medicine Affiliated Dongfang Hospital, Beijing, China
| | - Qiang Han
- Department of Traditional Chinese Medicine, Health Service Center of Beiyuan Community, Beijing, China
| | - Peng Zhang
- Department of Gastroenterology and Hepatology, Beijing University of Chinese Medicine Affiliated Dongfang Hospital, Beijing, China
| | - Lei Shi
- Department of Gastroenterology and Hepatology, Beijing University of Chinese Medicine Affiliated Dongfang Hospital, Beijing, China
| |
Collapse
|
10
|
Machado GC, Ferrer VP. MUC17 mutations and methylation are associated with poor prognosis in adult-type diffuse glioma patients. J Neurol Sci 2023; 452:120762. [PMID: 37562166 DOI: 10.1016/j.jns.2023.120762] [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: 02/10/2023] [Revised: 07/03/2023] [Accepted: 07/31/2023] [Indexed: 08/12/2023]
Abstract
Diffuse gliomas are tumors that arise from glial or glial progenitor cells. They are currently classified as astrocytoma isocitrate dehydrogenase (IDH)-mutant or oligodendroglioma IDH-mutant, and 1p/19q-codeleted, both slower-growing tumors, or glioblastoma (GBM), a more aggressive tumor. Despite advances in the diagnosis and treatment of gliomas, the median survival time after diagnosis of GBM remains low, approximately 15 months, with a 5-year overall survival rate of only 6.8%. Therefore, new biomarkers that could support the earlier diagnosis and prognosis of these tumors would be of great value. MUC17, a membrane-bound mucin, has been identified as a potential biomarker for several tumors. However, the role of this mucin in adult gliomas has not yet been explored. Here, we show for the first time, in a retrospective study and by in silico analysis that MUC17 is one of the relevant mutant genes in adult gliomas. Moreover, that an increase in MUC17 methylation correlates with an increase in glioma malignancy grade. Patients with MUC17 mutations had a poorer prognosis than their wild-type counterparts in both GBM and non-GBM glioma cohorts. We also analyzed mutational profiles that correlated strongly with poor survival. Therefore, in this study, we present a new potential biomarker for further investigation, especially for the prognosis of adult diffuse gliomas.
Collapse
Affiliation(s)
- Gabriel Cardoso Machado
- Laboratory of Cell and Molecular Biology of Tumors, Department of Cell and Molecular Biology, Biology Institute, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil; Graduate Program in Pathological Anatomy, Faculty of Medicine, Rio de Janeiro Federal University, Rio de Janeiro, Brazil
| | - Valéria Pereira Ferrer
- Laboratory of Cell and Molecular Biology of Tumors, Department of Cell and Molecular Biology, Biology Institute, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil; Graduate Program in Pathological Anatomy, Faculty of Medicine, Rio de Janeiro Federal University, Rio de Janeiro, Brazil.
| |
Collapse
|
11
|
Chamignon C, Mallaret G, Rivière J, Vilotte M, Chadi S, de Moreno de LeBlanc A, LeBlanc JG, Carvalho FA, Pane M, Mousset PY, Langella P, Lafay S, Bermúdez-Humarán LG. Beneficial Effects of Lactobacilli Species on Intestinal Homeostasis in Low-Grade Inflammation and Stress Rodent Models and Their Implication in the Modulation of the Adhesive Junctional Complex. Biomolecules 2023; 13:1295. [PMID: 37759696 PMCID: PMC10527021 DOI: 10.3390/biom13091295] [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: 07/04/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Intestinal barrier integrity is essential in order to maintain the homeostasis of mucosal functions and efficient defensive reactions against chemical and microbial challenges. An impairment of the intestinal barrier has been observed in several chronic diseases. The gut microbiota and its impact on intestinal homeostasis is well described and numerous studies suggest the ability of some probiotic strains to protect the intestinal epithelial integrity and host homeostasis. In this work, we aimed to assess the beneficial effects of three Lactobacillus strains (Lacticaseibacillus rhamnosus LR04, Lacticaseibacillus casei LC03, and Lactiplantibacillus plantarum CNCM I-4459) and their mechanism of action in low-grade inflammation or neonatal maternal separation models in mice. We compared the impact of these strains to that of the well-known probiotic Lacticaseibacillus rhamnosus GG. Our results demonstrated that the three strains have the potential to restore the barrier functions by (i) increasing mucus production, (ii) restoring normal permeability, and (iii) modulating colonic hypersensitivity. Moreover, gene expression analysis of junctional proteins revealed the implication of Claudin 2 and Cingulin in the mechanisms that underlie the interactions between the strains and the host. Taken together, our data suggest that LR04, CNCM I-4459, and LC03 restore the functions of an impaired intestinal barrier.
Collapse
Affiliation(s)
- Célia Chamignon
- Institut National de Recherche pour l’Agriculture et l’Environnement (INRAE), Micalis Institut, AgroParisTech, University of Paris-Saclay, 78350 Jouy-en-Josas, France; (C.C.); (J.R.); (S.C.); (P.L.)
- INDIGO Therapeutics, 33000 Bordeaux, France (S.L.)
| | - Geoffroy Mallaret
- INSERM U1107 NeuroDol, University of Clermont Auvergne, 63001 Clermont-Ferrand, France; (G.M.); (F.A.C.)
| | - Julie Rivière
- Institut National de Recherche pour l’Agriculture et l’Environnement (INRAE), Micalis Institut, AgroParisTech, University of Paris-Saclay, 78350 Jouy-en-Josas, France; (C.C.); (J.R.); (S.C.); (P.L.)
| | - Marthe Vilotte
- INRAE, GABI, AgroParisTech, University of Paris-Saclay, 78350 Jouy-en-Josas, France;
| | - Sead Chadi
- Institut National de Recherche pour l’Agriculture et l’Environnement (INRAE), Micalis Institut, AgroParisTech, University of Paris-Saclay, 78350 Jouy-en-Josas, France; (C.C.); (J.R.); (S.C.); (P.L.)
| | | | - Jean Guy LeBlanc
- CERELA-CONICET, San Miguel de Tucumán T4000ILC, Tucumán, Argentina; (A.d.M.d.L.); (J.G.L.)
| | - Frédéric Antonio Carvalho
- INSERM U1107 NeuroDol, University of Clermont Auvergne, 63001 Clermont-Ferrand, France; (G.M.); (F.A.C.)
| | - Marco Pane
- Probiotical Research, 28100 Novara, Italy;
| | | | - Philippe Langella
- Institut National de Recherche pour l’Agriculture et l’Environnement (INRAE), Micalis Institut, AgroParisTech, University of Paris-Saclay, 78350 Jouy-en-Josas, France; (C.C.); (J.R.); (S.C.); (P.L.)
| | - Sophie Lafay
- INDIGO Therapeutics, 33000 Bordeaux, France (S.L.)
| | - Luis G. Bermúdez-Humarán
- Institut National de Recherche pour l’Agriculture et l’Environnement (INRAE), Micalis Institut, AgroParisTech, University of Paris-Saclay, 78350 Jouy-en-Josas, France; (C.C.); (J.R.); (S.C.); (P.L.)
| |
Collapse
|
12
|
Chen K, McCulloch J, Das Neves R, Rodrigues G, Hsieh WT, Gong W, Yoshimura T, Huang J, O'hUigin C, Difilippantonio S, McCollum M, Jones G, Durum SK, Trinchieri G, Wang JM. The beneficial effects of commensal E. coli for colon epithelial cell recovery are related with Formyl peptide receptor 2 (Fpr2) in epithelial cells. Gut Pathog 2023; 15:28. [PMID: 37322488 PMCID: PMC10268441 DOI: 10.1186/s13099-023-00557-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/06/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Formyl peptide receptor 2 (Fpr2) plays a crucial role in colon homeostasis and microbiota balance. Commensal E. coli is known to promote the regeneration of damaged colon epithelial cells. The aim of the study was to investigate the connection between E. coli and Fpr2 in the recovery of colon epithelial cells. RESULTS The deficiency of Fpr2 was associated with impaired integrity of the colon mucosa and an imbalance of microbiota, characterized by the enrichment of Proteobacteria in the colon. Two serotypes of E. coli, O22:H8 and O91:H21, were identified in the mouse colon through complete genome sequencing. E. coli O22:H8 was found to be prevalent in the gut of mice and exhibited lower virulence compared to O91:H21. Germ-free (GF) mice that were pre-orally inoculated with E. coli O22:H8 showed reduced susceptibility to chemically induced colitis, increased proliferation of epithelial cells, and improved mouse survival. Following infection with E. coli O22:H8, the expression of Fpr2 in colon epithelial cells was upregulated, and the products derived from E. coli O22:H8 induced migration and proliferation of colon epithelial cells through Fpr2. Fpr2 deficiency increased susceptibility to chemically induced colitis, delayed the repair of damaged colon epithelial cells, and heightened inflammatory responses. Additionally, the population of E. coli was observed to increase in the colons of Fpr2-/- mice with colitis. CONCLUSION Commensal E. coli O22:H8 stimulated the upregulation of Fpr2 expression in colon epithelial cells, and the products from E. coli induced migration and proliferation of colon epithelial cells through Fpr2. Fpr2 deficiency led to an increased E. coli population in the colon and delayed recovery of damaged colon epithelial cells in mice with colitis. Therefore, Fpr2 is essential for the effects of commensal E. coli on colon epithelial cell recovery.
Collapse
Affiliation(s)
- Keqiang Chen
- Laboratory of Cancer Innovation, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA.
| | - John McCulloch
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Rodrigo Das Neves
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Gisele Rodrigues
- Laboratory of Cancer Innovation, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA
| | - Wang-Ting Hsieh
- Animal Health Diagnostic Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Wanghua Gong
- Basic Research Program, Leidos Biomedical Research, Inc, Frederick, MD, 21702, USA
| | - Teizo Yoshimura
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan
| | - Jiaqiang Huang
- Laboratory of Cancer Innovation, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA
- College of Life Sciences, Beijing Jiaotong University, Beijing, 100044, People's Republic of China
| | - Colm O'hUigin
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Simone Difilippantonio
- Gnotobiotics Facility, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Matthew McCollum
- Gnotobiotics Facility, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Georgette Jones
- Gnotobiotics Facility, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Scott K Durum
- Laboratory of Cancer Innovation, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA
| | - Giorgio Trinchieri
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Ji Ming Wang
- Laboratory of Cancer Innovation, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA
| |
Collapse
|
13
|
Sinha A, Vaggu RG, Swain R, Patnaik S. Repurposing of RAS-Pathway Mediated Drugs for Intestinal Inflammation Related Diseases for Treating SARS-CoV-2 Infection. Curr Microbiol 2023; 80:194. [PMID: 37106165 PMCID: PMC10136399 DOI: 10.1007/s00284-023-03304-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/16/2023] [Indexed: 04/29/2023]
Abstract
Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) is an emerging zoonotic virus, which causes Coronavirus Disease 2019 (COVID-19). Entry of coronaviruses into the cell depends on binding of the viral spike (S) proteins to cellular receptors Angiotensin-converting enzyme 2 (ACE2). The virus-mediated reduction of ACE2/Ang1-7 causes flooding of inflammatory cytokines. A similar scenario of hyper immunologic reaction has been witnessed in the context of Intestinal Inflammatory Diseases (IIDs) with the deregulation of ACE2. This review summarizes several IIDs that lead to such susceptible conditions. It discusses suitable mechanisms of how ACE2, being a crucial regulator of the Renin-Angiotensin System (RAS) signaling pathway, can affect the physiology of intestine as well as lungs, the primary site of SARS-CoV-2 infection. ACE2, as a SARS-CoV-2 receptor, establishes a critical link between COVID-19 and IIDs. Intercessional studies targeting the RAS signaling pathway in patients may provide a novel strategy for addressing the COVID-19 crisis. Hence, the modulation of these key RAS pathway members can be beneficial in both instances. However, it's difficult to say how beneficial are the ACE inhibitors (ACEI)/ Angiotensin II type-1 receptor blockers (ARBs) during COVID-19. As a result, much more research is needed to better understand the relationship between the RAS and SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Anupriya Sinha
- School of Biotechnology, KIIT University, Campus-XI, Bhubaneswar, Odisha, 751024, India
| | | | - Ramakrushna Swain
- School of Biotechnology, KIIT University, Campus-XI, Bhubaneswar, Odisha, 751024, India
| | - Srinivas Patnaik
- School of Biotechnology, KIIT University, Campus-XI, Bhubaneswar, Odisha, 751024, India.
| |
Collapse
|
14
|
Fang X, Kang L, Qiu YF, Li ZS, Bai Y. Yersinia enterocolitica in Crohn’s disease. Front Cell Infect Microbiol 2023; 13:1129996. [PMID: 36968108 PMCID: PMC10031030 DOI: 10.3389/fcimb.2023.1129996] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/13/2023] [Indexed: 03/11/2023] Open
Abstract
Increasing attention is being paid to the unique roles gut microbes play in both physiological and pathological processes. Crohn’s disease (CD) is a chronic, relapsing, inflammatory disease of the gastrointestinal tract with unknown etiology. Currently, gastrointestinal infection has been proposed as one initiating factor of CD. Yersinia enterocolitica, a zoonotic pathogen that exists widely in nature, is one of the most common bacteria causing acute infectious gastroenteritis, which displays clinical manifestations similar to CD. However, the specific role of Y. enterocolitica in CD is controversial. In this Review, we discuss the current knowledge on how Y. enterocolitica and derived microbial compounds may link to the pathogenesis of CD. We highlight examples of Y. enterocolitica-targeted interventions in the diagnosis and treatment of CD, and provide perspectives for future basic and translational investigations on this topic.
Collapse
Affiliation(s)
| | | | | | | | - Yu Bai
- *Correspondence: Zhao-Shen Li, ; Yu Bai,
| |
Collapse
|
15
|
Akinrinde AS, Oyewole SO, Ola-Davies OE. Supplementation with sesame oil suppresses genotoxicity, hepatotoxicity and enterotoxicity induced by sodium arsenite in rats. Lipids Health Dis 2023; 22:14. [PMID: 36707815 PMCID: PMC9881342 DOI: 10.1186/s12944-022-01760-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/19/2022] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Sesame oil, an edible essential oil, is known to be rich in unsaturated fatty acids, vitamins and lignans with several reported health-promoting benefits. Acute arsenic poisoning produces toxic hepatitis, bone marrow depression and adverse gastrointestinal responses. In this study, we investigated the protective effect of sesame seed oil (SSO) against genotoxicity, hepatotoxicity and colonic toxicity induced by sodium arsenite (SA) in Wistar rats. METHODS Twenty-eight male Wistar albino rats were randomly allocated into four groups: control, SA only (2.5 mg/kg), SA + SSO (4 ml/kg) and SSO alone for eight consecutive days. Liver function and morphology, bone marrow micronuclei induction, colonic histopathology, mucus production and immune expression of Bcl-2, carcinoembryonic antigen (CEA), MUC1 and cytokeratins AE1/AE3 were evaluated. RESULTS SA provoked increased serum activities of liver enzymes, including alanine aminotransferase (ALT) and aspartate aminotransferase (AST), and caused severely altered morphology of hepatic and colonic tissues with increased frequency of micronucleated polychromatic erythrocytes (MnPCEs/1000PCE) in the bone marrow. In addition, SA triggered increased expression of colonic CEA and MUC1 but weak Bcl-2 immunoexpression. However, cotreatment with SSO demonstrated protective activities against SA-induced damage, as indicated by significantly reduced serum ALT and AST, fewer micronucleated bone marrow erythrocytes and well-preserved hepatic and colonic morphologies compared to the SA-treated rats. Furthermore, SSO protected the colonic mucosa by boosting mucus production, elevating anti-apoptotic Bcl-2 expression and reducing CEA expression. GC-MS analysis of SSO revealed that it was predominated by linoleic acid, an omega-3 fatty acid, and tocopherols. CONCLUSIONS Our data indicated that SSO protected the liver, colon and bone marrow potentially via anti-inflammatory and anti-apoptotic activities. The data suggest that sesame oil has potential therapeutic applications against chemical toxicities induced by arsenic.
Collapse
Affiliation(s)
- Akinleye Stephen Akinrinde
- grid.9582.60000 0004 1794 5983Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Stephen Oluwasemilore Oyewole
- grid.9582.60000 0004 1794 5983Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olufunke Eunice Ola-Davies
- grid.9582.60000 0004 1794 5983Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
16
|
Abdel-Gaber R, Hawsah MA, Al-Otaibi T, Alojayri G, Al-Shaebi EM, Mohammed OB, Elkhadragy MF, Al-Quraishy S, Dkhil MA. Biosynthesized selenium nanoparticles to rescue coccidiosis-mediated oxidative stress, apoptosis and inflammation in the jejunum of mice. Front Immunol 2023; 14:1139899. [PMID: 36875142 PMCID: PMC9982015 DOI: 10.3389/fimmu.2023.1139899] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 02/08/2023] [Indexed: 02/19/2023] Open
Abstract
One of the most crucial approaches for treating human diseases, particularly parasite infections, is nanomedicine. One of the most significant protozoan diseases that impact farm and domestic animals is coccidiosis. While, amprolium is one of the traditional anticoccidial medication, the advent of drug-resistant strains of Eimeria necessitates the development of novel treatments. The goal of the current investigation was to determine whether biosynthesized selenium nanoparticles (Bio-SeNPs) using Azadirachta indica leaves extract might treat mice with Eimeria papillata infection in the jejunal tissue. Five groups of seven mice each were used, as follows: Group 1: Non-infected-non-treated (negative control). Group 2: Non-infected treated group with Bio-SeNPs (0.5 mg/kg of body weight). Groups 3-5 were orally inoculated with 1×103 sporulated oocysts of E. papillata. Group 3: Infected-non-treated (positive control). Group 4: Infected and treated group with Bio-SeNPs (0.5 mg/kg). Group 5: Infected and treated group with the Amprolium. Groups 4 and 5 daily received oral administration (for 5 days) of Bio-SeNPs and anticoccidial medication, respectively, after infection. Bio-SeNPs caused a considerable reduction in oocyst output in mice feces (97.21%). This was also accompanied by a significant reduction in the number of developmental parasitic stages in the jejunal tissues. Glutathione reduced (GSH), glutathione peroxidase (GPx), and superoxide dismutase (SOD) levels were dramatically reduced by the Eimeria parasite, whereas, nitric oxide (NO) and malonaldehyde (MDA) levels were markedly elevated. The amount of goblet cells and MUC2 gene expression were used as apoptotic indicators, and both were considerably downregulated by infection. However, infection markedly increased the expression of inflammatory cytokines (IL-6 and TNF-α) and the apoptotic genes (Caspase-3 and BCL2). Bio-SeNPs were administrated to mice to drastically lower body weight, oxidative stress, and inflammatory and apoptotic indicators in the jejunal tissue. Our research thus showed the involvement of Bio-SeNPs in protecting mice with E. papillata infections against jejunal damage.
Collapse
Affiliation(s)
- Rewaida Abdel-Gaber
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Maysar Abu Hawsah
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Tahani Al-Otaibi
- Department of Science and Technology, Al-Nairiyah University College, University of Hafr Al-Batin, Hafr Al-Batin, Saudi Arabia
| | - Ghada Alojayri
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Esam M Al-Shaebi
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Osama B Mohammed
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Manal F Elkhadragy
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Saleh Al-Quraishy
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed A Dkhil
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt.,Applied Science Research Center, Applied Science Private University, Amman, Jordan
| |
Collapse
|
17
|
Zhang Z, Dong M, Zallot R, Blackburn GM, Wang N, Wang C, Chen L, Baumann P, Wu Z, Wang Z, Fan H, Roth C, Jin Y, He Y. Mechanistic and Structural Insights into the Specificity and Biological Functions of Bacterial Sulfoglycosidases. ACS Catal 2022. [DOI: 10.1021/acscatal.2c05405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Zhen Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule, College of Chemistry and Materials Science, Northwest University, Xi’an 710127, P. R. China
- School of Chemistry, Cardiff University, Cardiff CF10 3AT, U.K
| | - Mochen Dong
- School of Chemistry, Cardiff University, Cardiff CF10 3AT, U.K
| | - Rémi Zallot
- Institute of Life Sciences, Swansea University Medical School, Swansea SA2 8PP, U.K
| | - George Michael Blackburn
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield S10 2TN, U.K
| | - Nini Wang
- Key Laboratory of Synthetic and Natural Functional Molecule, College of Chemistry and Materials Science, Northwest University, Xi’an 710127, P. R. China
| | - Chengjian Wang
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi’an 710069, P. R. China
| | - Long Chen
- School of Chemistry, Cardiff University, Cardiff CF10 3AT, U.K
| | - Patrick Baumann
- School of Chemistry, Cardiff University, Cardiff CF10 3AT, U.K
| | - Zuyan Wu
- School of Chemistry, Cardiff University, Cardiff CF10 3AT, U.K
| | - Zhongfu Wang
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi’an 710069, P. R. China
| | - Haiming Fan
- Key Laboratory of Synthetic and Natural Functional Molecule, College of Chemistry and Materials Science, Northwest University, Xi’an 710127, P. R. China
| | - Christian Roth
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Arnimallee 22, 14195 Berlin, German
| | - Yi Jin
- School of Chemistry, Cardiff University, Cardiff CF10 3AT, U.K
| | - Yuan He
- Key Laboratory of Synthetic and Natural Functional Molecule, College of Chemistry and Materials Science, Northwest University, Xi’an 710127, P. R. China
| |
Collapse
|
18
|
Yeom JE, Kim SK, Park SY. Regulation of the Gut Microbiota and Inflammation by β-Caryophyllene Extracted from Cloves in a Dextran Sulfate Sodium-Induced Colitis Mouse Model. Molecules 2022; 27:7782. [PMID: 36431883 PMCID: PMC9695579 DOI: 10.3390/molecules27227782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Ulcerative colitis is an inflammatory bowel disease characterized by symptoms such as abdominal pain, diarrhea, bleeding, and weight loss. Ulcerative colitis is typically treated with anti-inflammatory drugs; however, these drugs are associated with various side effects, limiting their use. β-Caryophyllene (BCP), a natural compound derived from cloves, has antioxidant, antibacterial, and anti-inflammatory activities. In this study, we aimed to investigate the effects of BCP on colitis in a dextran sulfate sodium (DSS)-induced colitis mouse model. BCP was administered for seven days, followed by 2.5% DSS for additional seven days to induce colitis. Changes in stool weight, recovery of gut motility, colon length, colon histology, myeloperoxidase activity, inflammatory cytokines (TNF-α, IL-1β, IL-6, IgA, and IgG), and the gut microbiota were observed. Administration of BCP increased stool weight, restored gut motility, and considerably increased colon length compared to those in the untreated colitis mouse model. In addition, the amount of mucin and myeloperoxidase activity in the colon increased, whereas the concentrations of IL-1β, IL-6, and TNF-α decreased following the administration of BCP. Furthermore, BCP reduced the abundance of Proteobacteria which can cause intestinal immune imbalance. These results suggest that BCP has a potential to be developed as a preventive agent for colitis.
Collapse
Affiliation(s)
- Ji Eun Yeom
- Laboratory of Pharmacognosy, College of Pharmacy, Dankook University, 119, Dandae-ro, Dongnam-gu, Cheonan-si 31116, Korea
- SFC Bio Co., Ltd., 119, Dandae-ro, Dongnam-gu, Cheonan-si 31116, Korea
| | - Sung-Kyu Kim
- SFC Bio Co., Ltd., 119, Dandae-ro, Dongnam-gu, Cheonan-si 31116, Korea
| | - So-Young Park
- Laboratory of Pharmacognosy, College of Pharmacy, Dankook University, 119, Dandae-ro, Dongnam-gu, Cheonan-si 31116, Korea
| |
Collapse
|
19
|
Wu X, Fu S, Jiang M, Wang J, Tang H, Fang C, Li W, Fu C. Sanhuang Xiexin decoction ameliorates DSS-induced colitis in mice by regulating intestinal inflammation, intestinal barrier, and intestinal flora. JOURNAL OF ETHNOPHARMACOLOGY 2022; 297:115537. [PMID: 35843414 DOI: 10.1016/j.jep.2022.115537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 06/30/2022] [Accepted: 07/11/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sanhuang Xiexin decoction (SXD) is a widely applicated traditional Chinese medicine (TCM) with a significant intestinal anti-inflammatory effect. AIM OF THE STUDY To evaluate the therapeutic effect and elucidate the possible underlying mechanisms of SXD on dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) in mice. METHODS To model UC, 3% DSS was added to the drinking water for 7 days. The UC mice were grouped and treated with three doses of SXD (1.3, 2.6, and 6 g/kg) orally for 7 days. Mice body weight and disease activity index (DAI) scores were recorded daily. After treatment with SXD, the colon was removed, and the colon length and histopathological changes were recorded. Blood cells were counted and colonic inflammatory cytokines and oxidative stress indicators were examined. The key proteins in TLR4-MyD88-NF-κB signaling and the colonic barrier were determined by Western blot analysis. The restorative effect of SXD on intestinal flora was determined. RESULTS Treatment with SXD reduced DAI scores, increased body weight, improved colon shortening, and decreased colonic damage. SXD decreased the numbers of white blood cells (WBCs), increased the numbers of red blood cells (RBCs), and inhibited the expression of inflammatory cytokines and oxidative stress indicators. In addition, SXD displayed an effective anti-inflammatory effect by inhibiting the expression levels of p-IκBα, TLR4, MyD88, and p65. Furthermore, SXD significantly restored the integrity of the colonic barrier and the abundance of beneficial flora. CONCLUSIONS SXD significantly reduced DSS-induced colon damage when the dose was higher than 1.3 g/kg, and the middle dose group (2.6 g/kg) indicated the best effect. SXD effectively ameliorated DSS-induced UC in mice, possibly by inhibiting oxidative stress, protecting the mucosal barrier, inhibiting the TLR4-MyD88-NF-κB signaling pathway, and regulating the intestinal flora.
Collapse
Affiliation(s)
- Xueyuan Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Chengdu Agricultural College, Chengdu, 611130, China
| | - Shu Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Miao Jiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jing Wang
- Wenjiang Traditional Chinese Medicine Hospital of Chengdu, Chengdu, 611130, China
| | - Huaqiao Tang
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Chunlin Fang
- Chengdu Agricultural College, Chengdu, 611130, China
| | - Wen Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
20
|
Wang L, Xu H, Yang H, Zhou J, Zhao L, Zhang F. Glucose metabolism and glycosylation link the gut microbiota to autoimmune diseases. Front Immunol 2022; 13:952398. [PMID: 36203617 PMCID: PMC9530352 DOI: 10.3389/fimmu.2022.952398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/19/2022] [Indexed: 11/21/2022] Open
Abstract
Carbohydrates serve as important energy sources and structural substances for human body as well as for gut microbes. As evidenced by the advances in immunometabolism, glucose metabolism and adenosine triphosphate (ATP) generation are deeply involved in immune cell activation, proliferation, and signaling transduction as well as trafficking and effector functions, thus contributing to immune response programming and assisting in host adaption to microenvironment changes. Increased glucose uptake, aberrant expression of glucose transporter 1 (e.g., GLU1), and abnormal glycosylation patterns have been identified in autoimmunity and are suggested as partially responsible for the dysregulated immune response and the modification of gut microbiome composition in the autoimmune pathogenesis. The interaction between gut microbiota and host carbohydrate metabolism is complex and bidirectional. Their impact on host immune homeostasis and the development of autoimmune diseases remains to be elucidated. This review summarized the current knowledge on the crosstalk of glucose metabolism and glycosylation in the host with intestinal microbiota and discussed their possible role in the development and progression of autoimmune diseases. Potential therapeutic strategies targeting glucose metabolism and glycosylation in modulating gut ecosystem and treating autoimmune diseases were discussed as well.
Collapse
Affiliation(s)
- Lu Wang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Rheumatology, Ministry of Education, Beijing, China
| | - Haojie Xu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Rheumatology, Ministry of Education, Beijing, China
| | - Huaxia Yang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Rheumatology, Ministry of Education, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
| | - Jiaxin Zhou
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Rheumatology, Ministry of Education, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- *Correspondence: Jiaxin Zhou, ; Lidan Zhao,
| | - Lidan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Rheumatology, Ministry of Education, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- *Correspondence: Jiaxin Zhou, ; Lidan Zhao,
| | - Fengchun Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Rheumatology, Ministry of Education, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
21
|
Jatrorrhizine Alleviates DSS-Induced Ulcerative Colitis by Regulating the Intestinal Barrier Function and Inhibiting TLR4/MyD88/NF-κB Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:3498310. [PMID: 36193153 PMCID: PMC9526656 DOI: 10.1155/2022/3498310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/28/2022] [Accepted: 08/10/2022] [Indexed: 11/18/2022]
Abstract
Background Ulcerative colitis (UC), a kind of autoimmune disease with unknown etiology, has been troubling human physical and mental health. Jatrorrhizine (Jat) is a natural isoquinoline alkaloid isolated from Coptis Chinensis, which has been proved to have antibacterial, anti-inflammatory, and antitumor effects. Purpose The purpose is to explore the therapeutic effect of Jat on DSS-induced UC and the mechanism of action. Study Design. The UC mice model was induced by 3% DSS in drinking water. The mice were orally administered with Jat (40, 80, 160 mg/kg) for 10 days. Methods The changes in body weight, colon length, spleen wet weight index, disease activity index (DAI), colonic histopathology, and inflammatory factors of serum and colon tissue were analyzed to evaluate the severity of colitis mice. The colon mucus secretion capacity was analyzed by Alcian blue periodic acid Schiff (AB-PAS) staining. Furthermore, protein expressions such as TLR4, MyD88, p–NF–κB-p65, NF-κB-p65, COX-2, ZO-1, and Occludin were detected to elucidate the molecular mechanism of Jat on DSS-induced colitis model. Results The results showed that Jat could significantly alleviate the symptoms, colon shortening, spleen index, and histological damage and restore the body weight in DSS-induced colitis mice. Jat also suppressed the levels of inflammatory cytokines and upregulated the levels of anti-inflammatory cytokines. In addition, Jat repaired the intestinal barrier function by upregulating the level of colonic tight junction (TJ) proteins and enhancing the secretion of mucin produced by goblet cells. Furthermore, Jat could significantly suppress the expression of TLR4, MyD88, p–NF–κB-p65/NF-κB-p65, and COX-2 in colon tissue. Conclusion The results suggested that Jat plays a protective role in DSS-induced colitis by regulating the intestinal barrier function and inhibiting the TLR4/MyD88/NF-κB signaling pathway. This study, for the first time, demonstrates the therapeutic and protective effects of Jat on UC.
Collapse
|
22
|
Ma S, Yeom J, Lim YH. Specific activation of hypoxia-inducible factor-2α by propionate metabolism via a β-oxidation-like pathway stimulates MUC2 production in intestinal goblet cells. Biomed Pharmacother 2022; 155:113672. [PMID: 36095963 DOI: 10.1016/j.biopha.2022.113672] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 12/20/2022] Open
Abstract
Microbiota-derived short-chain fatty acids (SCFAs) are known to stimulate mucin expression in the intestine, which contributes to the gut mucosal immune responses, and the gut mucosal immune system extends to the brain and other organs through several axes. Hypoxia-inducible factors (HIFs), especially HIF-1α, are known to act as the master regulator of mucin expression, however, underlying mechanism of mucin expression during hypoxia by SCFAs remains unclear. In this study, we investigated the mechanism of MUC2 expression by propionate, an SCFA, in intestinal goblet cells. The real time oxygen consumption rate (OCR) and ATPase activity were measured to investigate the induction of hypoxia by propionate. Using 2-dimensional electrophoresis (2-DE), microarray analysis, and siRNA-induced gene silencing, we found that propionate is metabolized via a β-oxidation-like pathway instead of the vitamin B12-dependent carboxylation pathway (also known as the methylmalonyl pathway). We verified the results by analyzing several intermediates in the pathway using LC-MS and GC-MS. Propionate metabolism via the β-oxidation-like pathway leads to the depletion of oxygen and thereby induces hypoxia. Analysis of HIFs revealed that HIF-2α is the primary HIF whose activation is induced by propionate metabolism in a hypoxic environment and that HIF-2α regulates the expression of MUC2. Thus, hypoxia induced during propionate metabolism via a β-oxidation-like pathway specifically activates HIF-2α, stimulating MUC2 production in LS 174 T goblet cells. Our findings show that propionate-induced selective HIF-2α stimulation contributes to intestinal mucosal defense.
Collapse
Affiliation(s)
- Seongho Ma
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Jiah Yeom
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Young-Hee Lim
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea; School of Biosystems and Biomedical Sciences, College of Health Science, Korea University, Seoul 02841, Republic of Korea; Department of Laboratory Medicine, Korea University Guro Hospital, Seoul 08308, Republic of Korea.
| |
Collapse
|
23
|
Quinoa bran soluble dietary fiber ameliorates dextran sodium sulfate induced ulcerative colitis in BALB/c mice by maintaining intestinal barrier function and modulating gut microbiota. Int J Biol Macromol 2022; 216:75-85. [DOI: 10.1016/j.ijbiomac.2022.06.194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 06/25/2022] [Accepted: 06/28/2022] [Indexed: 12/27/2022]
|
24
|
Wang H, Fan C, Zhao Z, Zhai Z, Hao Y. Anti-inflammatory effect of Bifidobacterium animalis subsp. lactis A6 on DSS-induced colitis in mice. J Appl Microbiol 2022; 133:2063-2073. [PMID: 35737740 DOI: 10.1111/jam.15681] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/02/2022] [Accepted: 06/20/2022] [Indexed: 11/30/2022]
Abstract
AIMS This study aimed to investigate the protective effect of Bifidobacterium animalis subsp. lactis A6 on dextran sodium sulphate (DSS)-induced colitis in C57BL/6J mice. METHODS AND RESULTS Mice were randomly divided into three groups (n = 8 per group). Each group was administered with PBS (Control and DSS group) or B. lactis A6 with a dosage of ~4.0 × 109 CFU day-1 (DSS + A6 group) for 21 consecutive days. The DSS and DSS + A6 group mice were ad libitum drinking 2.5% DSS water during day 15-21, while the Control group mice were given normal water. The administration of B. lactis A6 significantly inhibited DSS-induced bodyweight loss and colon shortening (p < 0.001), but showed no significant influence on the spleen enlargement (p > 0.05). The intestinal barrier integrity was improved by reducing colonic damage, recovering mucus layer loss and enhancing tight junction expression including ZO-1, occludin and claudin-1. In addition, B. lactis A6 attenuated the oxidative stress by decreasing MDA and increasing SOD and GSH levels in colon tissues. Moreover, B. lactis A6 suppressed DSS-induced inflammatory responses via downregulating TNF-α, IL-1β and IL-6 levels and upregulating IL-10 level in colon tissues. CONCLUSION B. lactis A6 effectively alleviated DSS-induced colitis by maintaining intestinal barrier integrity, reducing oxidative stress and inhibiting inflammatory responses. SIGNIFICANCE AND IMPACT OF THE STUDY This study suggests that B. lactis A6 could act as a candidate probiotic for UC treatment.
Collapse
Affiliation(s)
- Hui Wang
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Chengfei Fan
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Zhaoer Zhao
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Zhengyuan Zhai
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yanling Hao
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| |
Collapse
|
25
|
Si-Wu Water Extracts Protect against Colonic Mucus Barrier Damage by Regulating Muc2 Mucin Expression in Mice Fed a High-Fat Diet. Foods 2022; 11:foods11162499. [PMID: 36010498 PMCID: PMC9407452 DOI: 10.3390/foods11162499] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/09/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022] Open
Abstract
A high-fat diet (HFD) could cause gut barrier damage. The herbs in si-wu (SW) include dang gui (Angelica sinensis (Oliv.) Diels), shu di huang (the processed root of Rehmannia glutinosa Libosch.), chuan xiong (rhizome of Ligusticum chuanxiong Hort.), and bai shao (the root of Paeonia lactiflora f. pilosella (Nakai) Kitag.). Si-wu water extracts (SWE) have been used to treat blood deficiency. Components of one herb from SW have been reported to have anti-inflammatory and anti-obesity activities. However, there have been no reports about the effects of SWE on gut barrier damage. Therefore, the aim of the study was to explore the effect of SWE on gut barrier damage. In this study, we found that SWE effectively controlled body weight, liver weight, and feed efficiency, as well as decreased the serum TC level in HFD-fed mice. Moreover, SWE and rosiglitazone (Ros, positive control) increased the colonic alkaline phosphatase (ALP) level, down-regulated serum pro-inflammatory cytokine levels, and reduced intestinal permeability. In addition, SWE increased goblet cell numbers and mucus layer thickness to strengthen the mucus barrier. After supplementation with SWE and rosiglitazone, the protein expression of CHOP and GRP78 displayed a decrease, which improved the endoplasmic reticulum (ER) stress condition. Meanwhile, the increase in Cosmc and C1GALT1 improved the O-glycosylation process for correct protein folding. These results collectively demonstrated that SWE improved the mucus barrier, focusing on Muc2 mucin expression, in a prolonged high-fat diet, and provides evidence for the potential of SWE in the treatment of intestinal disease-associated mucus barrier damage.
Collapse
|
26
|
Pathological features-based targeted delivery strategies in IBD therapy: A mini review. Biomed Pharmacother 2022; 151:113079. [PMID: 35605297 DOI: 10.1016/j.biopha.2022.113079] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 11/22/2022] Open
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is characterized by a complex and dysfunctional immune response. Currently, IBD is incurable, and patients with IBD often need to take drugs for life. However, as the traditional systemic treatment strategies for IBD do not target the site of inflammation, only limited efficacy can be obtained from them. Moreover, the possibility of serious side effects stemming from the systemic administration or redistribution of drugs in the body is high when conventional drug formulations are used. Therefore, a targeted drug-delivery system for IBD should be considered. Based on the pathological features related to IBD, the new targeted drug-delivery strategy can directly transfer the drug to the inflammatory site, thus enhancing the accumulation of the drugs and reducing side effects. This article reviews the pathological features of IBD and the application of the IBD-targeted delivery system based on different pathological features, and discusses the challenges and new prospects in this field.
Collapse
|
27
|
Tao Y, Qiao S, Lv C, Yun X, Yue M, Fang Y, Wei Z, Dai Y, Xia Y. Phytoestrogen arctigenin preserves the mucus barrier in inflammatory bowel diseases by inhibiting goblet cell apoptosis via the
ERβ
/
TRIM21
/
PHB1
pathway. Phytother Res 2022; 36:3248-3264. [DOI: 10.1002/ptr.7495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 04/26/2022] [Accepted: 04/30/2022] [Indexed: 01/17/2023]
Affiliation(s)
- Yu Tao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
- Jiangsu Province Hospital of Chinese Medicine The Affiliated Hospital of Nanjing University of Chinese Medicine Nanjing China
| | - Si‐miao Qiao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| | - Chang‐jun Lv
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| | - Xin‐ming Yun
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| | - Meng‐fan Yue
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| | - Yu‐lai Fang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| | - Zhi‐feng Wei
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| | - Yue Dai
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| | - Yu‐feng Xia
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| |
Collapse
|
28
|
Jin S, Guan T, Wang S, Hu M, Liu X, Huang S, Liu Y. Dioscin Alleviates Cisplatin-Induced Mucositis in Rats by Modulating Gut Microbiota, Enhancing Intestinal Barrier Function and Attenuating TLR4/NF-κB Signaling Cascade. Int J Mol Sci 2022; 23:ijms23084431. [PMID: 35457248 PMCID: PMC9025408 DOI: 10.3390/ijms23084431] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/15/2022] [Accepted: 04/16/2022] [Indexed: 12/12/2022] Open
Abstract
Cisplatin-based chemotherapy causes intestinal mucositis, which causes patients immense suffering and hinders the process of cancer treatment. Dioscin is a natural steroid saponin that exhibits strong anti-inflammatory and immunomodulatory properties. Herein, we investigate the protective effect of dioscin on cisplatin induced mucositis in rats from the perspective of gut microbiota and intestinal barrier. We established a rat model of intestinal mucositis by tail vein injection of cisplatin, and concurrently treated with dioscin oral administration. Parameters, such as body weight, diarrheal incidence, and D-Lactate levels, were assessed in order to evaluate the effects of dioscin on intestinal mucositis in rats. Furthermore, biological samples were collected for microscopic gut microbiota, intestinal integrity, and immune inflammation analyses to elucidate the protective mechanisms of dioscin on intestinal mucositis. The results revealed that administration of dioscin significantly attenuated clinical manifestations, histological injury and inflammation in mucositis rats. Besides this, dioscin markedly inhibited the gut microbiota dysbiosis induced by cisplatin. Meanwhile, dioscin partially alleviated junctions between ileum epithelial cells and increased mucus secretion. Moreover, dioscin effectively inhibited the TLR4-MyD88-NF-κB signal transduction pathway and reduced the secretion of subsequent inflammatory mediators. These results suggested that dioscin effectively attenuated cisplatin-induced mucositis in part by modulating the gut microflora profile, maintaining ileum integrity and inhibiting the inflammatory response through the TLR4-MyD88-NF-κB pathway.
Collapse
Affiliation(s)
- Shengzi Jin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (S.J.); (T.G.); (S.W.); (M.H.); (X.L.); (S.H.)
| | - Tongxu Guan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (S.J.); (T.G.); (S.W.); (M.H.); (X.L.); (S.H.)
| | - Shuang Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (S.J.); (T.G.); (S.W.); (M.H.); (X.L.); (S.H.)
| | - Mengxin Hu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (S.J.); (T.G.); (S.W.); (M.H.); (X.L.); (S.H.)
| | - Xingyao Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (S.J.); (T.G.); (S.W.); (M.H.); (X.L.); (S.H.)
| | - Siqi Huang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (S.J.); (T.G.); (S.W.); (M.H.); (X.L.); (S.H.)
| | - Yun Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (S.J.); (T.G.); (S.W.); (M.H.); (X.L.); (S.H.)
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, China
- Correspondence:
| |
Collapse
|
29
|
Livzan MA, Bicbavova GR, Romanyuk AE. Ulcerative colitis: focus on colonic mucosal resistance. BULLETIN OF SIBERIAN MEDICINE 2022; 21:121-132. [DOI: 10.20538/1682-0363-2022-1-121-132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
In recent decades, following cooperation between scientists in various specialties, new unique data on the pathogenesis of ulcerative colitis have been obtained. The role of an impaired immune response to antigens of gut microbiota in genetically predisposed individuals under the effect of certain environmental factors was proven. Assessing the interaction between the colonic mucosa and gut microbiota will help to understand the mechanisms of ulcerative colitis and develop new treatment strategies for the disease.This review presents modern views on the pathogenesis of ulcerative colitis with a focus on the imbalance between local protective and aggressive factors of the gastric and intestinal mucosa. The structure and role of the epithelial barrier both under normal conditions and in ulcerative colitis are considered in detail.The aim of this review was to summarize the data on resistance of the colonic mucosa and its damage in ulcerative colitis.
Collapse
|
30
|
Biological Clock and Inflammatory Bowel Disease Review: From the Standpoint of the Intestinal Barrier. Gastroenterol Res Pract 2022; 2022:2939921. [PMID: 35320972 PMCID: PMC8938076 DOI: 10.1155/2022/2939921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 02/27/2022] [Accepted: 03/01/2022] [Indexed: 11/29/2022] Open
Abstract
Inflammatory bowel disease is a group of chronic, recurrent, nonspecific inflammatory diseases of the intestine that severely affect the quality of life of patients. The pathogenesis of this disease is caused by complex and interactive neural networks composed of factors such as genetic susceptibility, external environment, immune disorders, and intestinal barrier dysfunction. It is well known that there is a strong link between environmental stressors (also known as circadian clocks) that can influence circadian changes and inflammatory bowel disease. Among them, the biological clock is involved in the pathogenesis of inflammatory bowel disease by affecting the function of the intestinal barrier. Therefore, this review is aimed at systematically summarizing the latest research progress on the role of the circadian clock in the pathogenesis of inflammatory bowel disease by affecting intestinal barrier functions (intestinal mechanical barrier, intestinal immune barrier, intestinal microecological barrier, and intestinal chemical barrier) and the potential clinical value of clock genes in the management of inflammatory bowel disease, for the application of circadian clock therapy in the management of inflammatory bowel disease and then the benefit to the majority of patients.
Collapse
|
31
|
Hu W, Duan H, Zhong S, Zeng J, Mou Y. High frequency of PDGFRA and MUC family gene mutations in diffuse hemispheric glioma, H3 G34-mutant: a glimmer of hope? J Transl Med 2022; 20:64. [PMID: 35109850 PMCID: PMC8812218 DOI: 10.1186/s12967-022-03258-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 01/17/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Diffuse hemispheric glioma H3 G34-mutant (G34-DHG) is a new type of pediatric-type diffuse high-grade glioma in the fifth edition of the WHO Classification of Tumors of the Central Nervous System. The current treatment for G34-DHG involves a combination of surgery and conventional radiotherapy or chemotherapy; however, the therapeutic efficacy of this approach is not satisfactory. In recent years, molecular targeted therapy and immunotherapy have achieved significant benefits in a variety of tumors. In-depth understanding of molecular changes and immune infiltration in G34-DHGs will help to establish personalized tumor treatment strategies. Here, we report the clinicopathological, molecular and immune infiltration characteristics of G34-DHG cases from our center along with cases from the HERBY Trial and the Chinese Glioma Genome Atlas database (CGGA). METHODS Hematoxylin-eosin (HE) and immunohistochemistry (IHC) staining were used to present the clinicopathological characteristics of 10 Chinese G34-DHG patients treated at our institution. To address the molecular characteristics of G34-DHG, we performed whole-exome sequencing (WES) and RNA sequencing (RNA-seq) analyses of 5 patients from our center and 3 Chinese patients from the Chinese Glioma Genome Atlas (CGGA) database. Additionally, 7 European G34-DHG patients from the HERBY Trail were also subjected to analyses, with 7 cases of WES data and 2 cases of RNA-seq data. Six G34-DHG patients from another organization were used as external validation. RESULTS WES showed a high frequency of PDGFRA mutation in G34-DHGs (12/15). We further identified frequent mutations in MUC family genes in G34-DHGs, including MUC16 (8/15) and MUC17 (8/15). Although no statistical difference was found, PDGFRA mutation tended to be an indicator for worse prognosis whereas MUC16/MUC17 mutation indicated a favorable prognosis in G34-DHGs. RNA sequencing results revealed that most G34-DHG are considered to be immune cold tumors. However, one patient in our cohort with MUC16 mutation showed significant immune infiltration, and the total overall survival of this patient reached 75 months. CONCLUSIONS Our results demonstrate that G34-DHG is a new high-grade glioma with high frequency of PDGFRA and MUC gene family mutations. PDGFRA may serve as an indicator of poor prognosis and an effective therapeutic target. Moreover, MUC16 tends to be a favorable prognostic factor and indicates high immune infiltration in certain patients, and these findings may provide a new direction for targeted therapy and immunotherapy of patients with G34-DHGs.
Collapse
Affiliation(s)
- Wanming Hu
- Department of Pathology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Hao Duan
- Department of Neurosurgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Sheng Zhong
- Department of Neurosurgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jing Zeng
- Department of Pathology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| | - Yonggao Mou
- Department of Neurosurgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
32
|
Mucins Dynamics in Physiological and Pathological Conditions. Int J Mol Sci 2021; 22:ijms222413642. [PMID: 34948435 PMCID: PMC8707880 DOI: 10.3390/ijms222413642] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/10/2021] [Accepted: 12/16/2021] [Indexed: 12/14/2022] Open
Abstract
Maintaining intestinal health requires clear segregation between epithelial cells and luminal microbes. The intestinal mucus layer, produced by goblet cells (GCs), is a key element in maintaining the functional protection of the epithelium. The importance of the gut mucus barrier is highlighted in mice lacking Muc2, the major form of secreted mucins. These mice show closer bacterial residence to epithelial cells, develop spontaneous colitis and became moribund when infected with the attaching and effacing pathogen, Citrobacter rodentium. Furthermore, numerous observations have associated GCs and mucus layer dysfunction to the pathogenesis of inflammatory bowel disease (IBD). However, the molecular mechanisms that regulate the physiology of GCs and the mucus layer remain obscured. In this review, we consider novel findings describing divergent functionality and expression profiles of GCs subtypes within intestinal crypts. We also discuss internal (host) and external (diets and bacteria) factors that modulate different aspects of the mucus layer as well as the contribution of an altered mucus barrier to the onset of IBD.
Collapse
|
33
|
Meng Z, Huang S, Sun W, Yan S, Chen X, Diao J, Zhou Z, Zhu W. A Typical Fungicide and Its Main Metabolite Promote Liver Damage in Mice through Impacting Gut Microbiota and Intestinal Barrier Function. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:13436-13447. [PMID: 34735141 DOI: 10.1021/acs.jafc.1c05508] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The environmental risks of prothioconazole (PTC), a popular agricultural fungicide, and its main metabolite, prothioconazole-desthio (PTCd), have attracted more and more attention recently. In this study, the adverse effects of PTC and PTCd on liver function in mice and their underlying mechanisms have been systematically studied from the perspective of gut microbiota. Combining the results of physiological, biochemical, and histopathological analysis showed that PTC and PTCd exposure could cause lipid accumulation and inflammation in the liver of mice. In addition, exposure to PTC and PTCd could also significantly affect the transcriptome of liver tissue, leading to disorders of lipid metabolism of the liver. Particularly, the abundances of bacteria in liver tissues were significantly increased with PTC and PTCd exposure. Further results show that PTC and PTCd could affect the expression of genes related to inflammation and the barrier function in colon tissue, leading to intestinal dysfunction in mice. Last but not least, the results based on 16S rRNA gene sequencing and 1H NMR metabolomics analysis showed that exposure to PTC and PTCd could cause gut microbiota imbalances and cecal content metabolic profile disorders. In short, this study found that PTC and PTCd exposure could cause liver damage in mice by changing the gut microbiota, disrupting the intestinal barrier function and promoting bacterial translocation. These results clarified the key role of gut microbiota in liver damage induced by PTC and PTCd in mice and proposed a new insight into the mechanisms of liver toxicity induced by pesticides through the dialogue of the gut-liver axis.
Collapse
Affiliation(s)
- Zhiyuan Meng
- School of Horticulture and Plant Protection, Yangzhou University/Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou, Jiangsu 225009, China
| | - Shiran Huang
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Science, China Agricultural University, Beijing 100193, China
| | - Wei Sun
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Science, China Agricultural University, Beijing 100193, China
| | - Sen Yan
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Science, China Agricultural University, Beijing 100193, China
| | - Xiaojun Chen
- School of Horticulture and Plant Protection, Yangzhou University/Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou, Jiangsu 225009, China
| | - Jinling Diao
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Science, China Agricultural University, Beijing 100193, China
| | - Zhiqiang Zhou
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Science, China Agricultural University, Beijing 100193, China
| | - Wentao Zhu
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Science, China Agricultural University, Beijing 100193, China
| |
Collapse
|
34
|
Mucolytic bacteria: prevalence in various pathological diseases. World J Microbiol Biotechnol 2021; 37:176. [PMID: 34519941 DOI: 10.1007/s11274-021-03145-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/07/2021] [Indexed: 02/07/2023]
Abstract
All mucins are highly glycosylated and a key constituent of the mucus layer that is vigilant against pathogens in many organ systems of animals and humans. The viscous layer is organized in bilayers, i.e., an outer layer that is loosely arranged, variable in thickness, home to the commensal microbiota that grows in the complex environment, and an innermost layer that is stratified, non-aspirated, firmly adherent to the epithelial cells and devoid of any microorganisms. The O-glycosylation moiety represents the site of adhesion for pathogens and due to the increase of motility, mucolytic activity, and upregulation of virulence factors, some microorganisms can circumvent the component of the mucus layer and cause disruption in organ homeostasis. A dysbiotic microbiome, defective mucus barrier, and altered immune response often result in various diseases. In this review, paramount emphasis is given to the role played by the bacterial species directly or indirectly involved in mucin degradation, alteration in mucus secretion or its composition or mucin gene expression, which instigates many diseases in the digestive, respiratory, and other organ systems. A systematic view can help better understand the etiology of some complex disorders such as cystic fibrosis, ulcerative colitis and expand our knowledge about mucin degraders to develop new therapeutic approaches to correct ill effects caused by these mucin-dwelling pathogens.
Collapse
|
35
|
Gu Z, Zhu Y, Mei F, Dong X, Xia G, Shen X. Tilapia head glycolipids protect mice against dextran sulfate sodium-induced colitis by ameliorating the gut barrier and suppressing NF-kappa B signaling pathway. Int Immunopharmacol 2021; 96:107802. [PMID: 34162163 DOI: 10.1016/j.intimp.2021.107802] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/10/2021] [Accepted: 05/17/2021] [Indexed: 12/26/2022]
Abstract
The purpose of this study was to evaluate the relieving effect of tilapia head glycolipids (TH-GLs) on dextran sulfate sodium (DSS)-induced colitis in mice and to further explore its mechanism. Mice were orally administered 3% (w/v) DSS to establish a model of ulcerative colitis (UC), and subsequently treated with TH-GLs or sulfasalazine. In addition, the expression of key targets in the intestinal mucosal barrier and the inflammatory signal pathway were studied by combining immunochemical analysis techniques. The results showed that varying doses of TH-GLs can significantly improve colon lesions caused by DSS, reduce histological scores, increase mucus secretion, extend colon length, increase weight, and inhibit the occurrence of inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), Interleukin-1β (IL-1β), and Interleukin- 6 (IL-6). Further, studies have shown that TH-GLs increase the secretion of MUC2 and up-regulate the expression of tight junction related proteins, such as ZO-1 and Occludin. In addition, TH-GLs significantly down-regulated the protein expression levels of TNF-α, IKK-β, and nuclear factor-κB (NF-κB). Here, we have elucidated the potential mechanism of TH-GLs in protecting mice with colitis. In general, this study shows that TH-GLs could improve the symptoms of UC by improving the gut barrier and inhibiting inflammatory signals, which provides a scientific basis for future clinical applications.
Collapse
Affiliation(s)
- Zhipeng Gu
- Hainan Engineering Research Center of Aquatic Resources Efficient Utilization in South China Sea, Hainan University, Hainan 570228, China; Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou 570228, China; Engineering Research Center of Utilization of Tropical Polysaccharide Resources, Ministry of Education, Haikou 570228, China; Key Laboratory of Seafood Processing of Haikou, Hainan University, Haikou 570228, Hainan, China; College of Food Science and Technology, Hainan University, Hainan 570228, China
| | - Yujie Zhu
- Hainan Engineering Research Center of Aquatic Resources Efficient Utilization in South China Sea, Hainan University, Hainan 570228, China; Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou 570228, China; Engineering Research Center of Utilization of Tropical Polysaccharide Resources, Ministry of Education, Haikou 570228, China; Key Laboratory of Seafood Processing of Haikou, Hainan University, Haikou 570228, Hainan, China; College of Food Science and Technology, Hainan University, Hainan 570228, China
| | - Fengfeng Mei
- Hainan Engineering Research Center of Aquatic Resources Efficient Utilization in South China Sea, Hainan University, Hainan 570228, China; Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou 570228, China; Engineering Research Center of Utilization of Tropical Polysaccharide Resources, Ministry of Education, Haikou 570228, China
| | - Xiuping Dong
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116000, Liaoning, China
| | - Guanghua Xia
- Hainan Engineering Research Center of Aquatic Resources Efficient Utilization in South China Sea, Hainan University, Hainan 570228, China; Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou 570228, China; Engineering Research Center of Utilization of Tropical Polysaccharide Resources, Ministry of Education, Haikou 570228, China; Key Laboratory of Seafood Processing of Haikou, Hainan University, Haikou 570228, Hainan, China; College of Food Science and Technology, Hainan University, Hainan 570228, China.
| | - Xuanri Shen
- Hainan Engineering Research Center of Aquatic Resources Efficient Utilization in South China Sea, Hainan University, Hainan 570228, China; Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou 570228, China; Engineering Research Center of Utilization of Tropical Polysaccharide Resources, Ministry of Education, Haikou 570228, China; Key Laboratory of Seafood Processing of Haikou, Hainan University, Haikou 570228, Hainan, China; College of Food Science and Technology, Hainan University, Hainan 570228, China.
| |
Collapse
|
36
|
Tran DN, Go SM, Park SM, Jung EM, Jeung EB. Loss of Nckx3 Exacerbates Experimental DSS-Induced Colitis in Mice through p53/NF-κB Pathway. Int J Mol Sci 2021; 22:ijms22052645. [PMID: 33807999 PMCID: PMC7961925 DOI: 10.3390/ijms22052645] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 12/20/2022] Open
Abstract
Inflammatory bowel diseases (IBDs) comprises a range of chronic inflammatory conditions of the intestinal tract. The incidence and prevalence of IBDs are increasing worldwide, but the precise etiology of these diseases is not completely understood. Calcium signaling plays a regulatory role in cellular proliferation. Nckx3, a potassium-dependent Na+/Ca2+ exchanger, is not only expressed in the brain but also in the aortic, uterine, and intestinal tissues, which contain abundant smooth muscle cells. This study investigated the role of Nckx3 in intestinal inflammation. Microarray analyses revealed the upregulation of the innate immune response-associated genes in the duodenum of Nckx3 knockout (KO) mice. The Nckx3 KO mice also showed an increase in IBD- and tumorigenesis-related genes. Using dextran sodium sulfate (DSS)-induced experimental colitis mice models, the Nckx3 KO mice showed severe colitis. Furthermore, the pathways involving p53 and NF-κB signaling were significantly upregulated by the absence of Nckx3. Overall, Nckx3 plays a critical role in the innate immune and immune response and may be central to the pathogenesis of IBD.
Collapse
Affiliation(s)
- Dinh Nam Tran
- Laboratory of Veterinary Biochemistry and Molecular Biology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Korea; (D.N.T.); (S.M.G.); (S.-M.P.)
| | - Seon Myeong Go
- Laboratory of Veterinary Biochemistry and Molecular Biology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Korea; (D.N.T.); (S.M.G.); (S.-M.P.)
| | - Seon-Mi Park
- Laboratory of Veterinary Biochemistry and Molecular Biology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Korea; (D.N.T.); (S.M.G.); (S.-M.P.)
| | - Eui-Man Jung
- Laboratory of Molecular Developmental Biology, Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busandaehang-ro, 63beon-gil 2, Geumjeong-gu, Busan 46241, Korea;
| | - Eui-Bae Jeung
- Laboratory of Veterinary Biochemistry and Molecular Biology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Korea; (D.N.T.); (S.M.G.); (S.-M.P.)
- Correspondence: ; Tel.: +82-43-261-2397; Fax: +82-43-267-3150
| |
Collapse
|
37
|
Tsuzuno T, Takahashi N, Yamada-Hara M, Yokoji-Takeuchi M, Sulijaya B, Aoki-Nonaka Y, Matsugishi A, Katakura K, Tabeta K, Yamazaki K. Ingestion of Porphyromonas gingivalis exacerbates colitis via intestinal epithelial barrier disruption in mice. J Periodontal Res 2021; 56:275-288. [PMID: 33512709 DOI: 10.1111/jre.12816] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 09/07/2020] [Accepted: 10/18/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVE This study aimed to evaluate the effects of ingested periodontal pathogens on experimental colitis in mice and to elucidate its underlying mechanisms. BACKGROUND Inflammatory bowel disease (IBD) is defined as a chronic intestinal inflammation that results in damage to the gastrointestinal tract. Epidemiological studies have shown an association between IBD and periodontitis. Although a large number of ingested oral bacteria reach gastrointestinal tract constantly, the effect of ingested periodontal pathogens on intestinal inflammation is still unknown. METHODS Experimental colitis was induced by inclusion of dextran sodium sulfate solution in drinking water of the mice. Major periodontal pathogens (Porphyromonas gingivalis, Prevotella intermedia, and Fusobacterium nucleatum) were administered orally every day during the experiment. The severity of colitis between the groups was compared. In vitro studies of the intestinal epithelial cell line were conducted to explore the molecular mechanisms by which periodontal pathogens affect the development of colitis. RESULTS The oral administration of P. gingivalis significantly increased the severity of colitis when compared to other pathogens in the DSS-induced colitis model. The ingested P. gingivalis disrupted the colonic epithelial barrier by decreasing the expression of tight junction proteins in vivo. In vitro permeability assays using the intestinal epithelial cell line suggested the P. gingivalis-specific epithelial barrier disruption. The possible involvement of gingipains in the exacerbation of colitis was implied by using P. gingivalis lacking gingipains. CONCLUSION Porphyromonas gingivalis exacerbates gastrointestinal inflammation by directly interacting with the intestinal epithelial barrier in a susceptible host.
Collapse
Affiliation(s)
- Takahiro Tsuzuno
- Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Naoki Takahashi
- Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Miki Yamada-Hara
- Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Research Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Mai Yokoji-Takeuchi
- Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Benso Sulijaya
- Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Department of Periodontology, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Yukari Aoki-Nonaka
- Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Aoi Matsugishi
- Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kyoko Katakura
- Department of Gastroenterology, Iwase general hospital, Fukushima, Japan
| | - Koichi Tabeta
- Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kazuhisa Yamazaki
- Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
38
|
Yao G, Zhang H, Luo G, Wang Z, Zhao H, Zhang J, Dong Y, Gao Y, Wu S. Effect of Senecio scandens ethanol extract on gut microbiota composition in mice. Arch Microbiol 2021; 203:1477-1488. [PMID: 33394080 DOI: 10.1007/s00203-020-02144-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 11/20/2020] [Accepted: 12/03/2020] [Indexed: 01/07/2023]
Abstract
The gut microbiota inhabits the animal intestinal tract, and dysbiosis of the gut microbiota may result in disease. Senecio scandens has pharmaceutical antibacterial activities and is regarded as a broad-spectrum antibiotic in traditional Chinese medicine. Extracts of S. scandens are reported to show strong antimicrobial activity, and quercetin significantly decreases some species in the caecal microflora. However, the bactericidal effects of the extracts on the gut microbiota remain obscure. Here, we supplied ethanol extract of S. scandens, which might possibly be used as an alternative for chemical antibiotics, to mice to investigate the state of the intestinal microbiota. Our studies included a control group, low-, moderate-, and high-dose ethanol extract groups, and cefixime capsule group. The ethanol extract groups did not present reduced diversity or differences in the gut microbiota balance. There were significant differences between the ethanol extract and cefixime capsule groups in terms of the gut microbiota. The control and ethanol extract groups contained similar bacteria, which suggested that the ethanol extract has no inhibitory effect on the gut microbiota in vivo. Bifidobacteriales and Lactobacillus acidophilus were significantly increased in the high-dose group. Both secretory immunoglobulin A and mucin 2 concentrations increased as the dose of ethanol extract increased. The functional prediction differences between the control and ethanol extract groups decreased with increasing extract doses, which indicated that the low-dose and high-dose extract treatments might regulate different pathways and functions of the gut microbiota. The results also highlighted the prevention of bacterial drug resistance in the ethanol extract groups.
Collapse
Affiliation(s)
- Gang Yao
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Huaxi University Town, Dongqing South Road, Guiyang, 550025, China.
| | - Hui Zhang
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Huaxi University Town, Dongqing South Road, Guiyang, 550025, China
| | - Guoyong Luo
- College of Pharmaceutical Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Zuhua Wang
- College of Pharmaceutical Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Hai Zhao
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Huaxi University Town, Dongqing South Road, Guiyang, 550025, China
| | - Jian Zhang
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Huaxi University Town, Dongqing South Road, Guiyang, 550025, China
| | - Yuanqiu Dong
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Huaxi University Town, Dongqing South Road, Guiyang, 550025, China
| | - Yuan Gao
- College of Pharmaceutical Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Shuguang Wu
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Huaxi University Town, Dongqing South Road, Guiyang, 550025, China
| |
Collapse
|
39
|
Fenton CG, Taman H, Florholmen J, Sørbye SW, Paulssen RH. Transcriptional Signatures That Define Ulcerative Colitis in Remission. Inflamm Bowel Dis 2021; 27:94-105. [PMID: 32322884 PMCID: PMC7737162 DOI: 10.1093/ibd/izaa075] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND This study addresses whether existing specific transcriptional profiles can improve and support the current status of the definition of ulcerative colitis (UC) remission apart from the existing endoscopic, histologic, and laboratory scoring systems. For that purpose, a well-stratified UC patient population in remission was compared to active UC and control patients and was investigated by applying the next-generation technology RNA-Seq. METHODS Mucosal biopsies from patients in remission (n = 14), patients with active UC (n = 14), and healthy control patientss (n = 16) underwent whole-transcriptome RNA-Seq. Principal component analysis, cell deconvolution methods, gene profile enrichment, and pathway enrichment methods were applied to define a specific transcriptional signature of UC in remission. RESULTS Analyses revealed specific transcriptional signatures for UC in remission with increased expression of genes involved in O-glycosylation (MUC17, MUC3A, MUC5AC, MUC12, SPON1, B3GNT3), ephrin-mediated repulsion of cells (EFNB2E, EFNA3, EPHA10, EPHA1), GAP junction trafficking (TUBA1C, TUBA4A, TUBB4B, GJB3, CLTB), and decreased expression of several toll-like receptors (TLR1, TLR3, TLR5, TLR6). CONCLUSIONS This study reveals specific transcriptional signatures for remission. Partial restoration and improvement of homeostasis in the epithelial mucus layer and revival of immunological functions were observed. A clear role for bacterial gut flora composition can be implied. The results can be useful for the development of treatment strategies for UC in remission and may be useful targets for further investigations aiming to predict the outcome of UC in the future.
Collapse
Affiliation(s)
- Christopher G Fenton
- Department of Clinical Medicine,Genomics Support Centre Tromsø, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Hagar Taman
- Department of Clinical Medicine,Genomics Support Centre Tromsø, UiT-The Arctic University of Norway, Tromsø, Norway
- Department of Clinical Medicine, Gastroenterology and Nutrition Research Group, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Jon Florholmen
- Department of Clinical Medicine, Gastroenterology and Nutrition Research Group, UiT-The Arctic University of Norway, Tromsø, Norway
- Department of Medical Gastroenterology, University Hospital of North Norway, Tromsø, Norway
| | - Sveinung W Sørbye
- §Department of Clinical Pathology, University Hospital of North Norway, Tromsø, Norway
| | - Ruth H Paulssen
- Department of Clinical Medicine,Genomics Support Centre Tromsø, UiT-The Arctic University of Norway, Tromsø, Norway
- Department of Clinical Medicine, Gastroenterology and Nutrition Research Group, UiT-The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
40
|
Leaky Gut and Autoimmunity: An Intricate Balance in Individuals Health and the Diseased State. Int J Mol Sci 2020; 21:ijms21249770. [PMID: 33371435 PMCID: PMC7767453 DOI: 10.3390/ijms21249770] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/08/2020] [Accepted: 12/17/2020] [Indexed: 02/07/2023] Open
Abstract
Damage to the tissue and the ruining of functions characterize autoimmune syndromes. This review centers around leaky gut syndromes and how they stimulate autoimmune pathogenesis. Lymphoid tissue commonly associated with the gut, together with the neuroendocrine network, collaborates with the intestinal epithelial wall, with its paracellular tight junctions, to maintain the balance, tolerance, and resistance to foreign/neo-antigens. The physiological regulator of paracellular tight junctions plays a vital role in transferring macromolecules across the intestinal barrier and thereby maintains immune response equilibrium. A new paradigm has explained the intricacies of disease development and proposed that the processes can be prevented if the interaction between the genetic factor and environmental causes is barred by re-instituting the intestinal wall function. The latest clinical evidence and animal models reinforce this current thought and offer the basis for innovative methodologies to thwart and treat autoimmune syndromes.
Collapse
|
41
|
Kudelka MR, Stowell SR, Cummings RD, Neish AS. Intestinal epithelial glycosylation in homeostasis and gut microbiota interactions in IBD. Nat Rev Gastroenterol Hepatol 2020; 17:597-617. [PMID: 32710014 PMCID: PMC8211394 DOI: 10.1038/s41575-020-0331-7] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/05/2020] [Indexed: 02/08/2023]
Abstract
Inflammatory bowel disease (IBD) affects 6.8 million people globally. A variety of factors have been implicated in IBD pathogenesis, including host genetics, immune dysregulation and gut microbiota alterations. Emerging evidence implicates intestinal epithelial glycosylation as an underappreciated process that interfaces with these three factors. IBD is associated with increased expression of truncated O-glycans as well as altered expression of terminal glycan structures. IBD genes, glycosyltransferase mislocalization, altered glycosyltransferase and glycosidase expression and dysbiosis drive changes in the glycome. These glycan changes disrupt the mucus layer, glycan-lectin interactions, host-microorganism interactions and mucosal immunity, and ultimately contribute to IBD pathogenesis. Epithelial glycans are especially critical in regulating the gut microbiota through providing bacterial ligands and nutrients and ultimately determining the spatial organization of the gut microbiota. In this Review, we discuss the regulation of intestinal epithelial glycosylation, altered epithelial glycosylation in IBD and mechanisms for how these alterations contribute to disease pathobiology. We hope that this Review provides a foundation for future studies on IBD glycosylation and the emergence of glycan-inspired therapies for IBD.
Collapse
Affiliation(s)
- Matthew R Kudelka
- Medical Scientist Training Program, Emory University School of Medicine, Atlanta, GA, USA
- Department of Internal Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sean R Stowell
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Andrew S Neish
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
42
|
Dietary Supplementation with Spray-Dried Porcine Plasma Attenuates Colon Inflammation in a Genetic Mouse Model of Inflammatory Bowel Disease. Int J Mol Sci 2020; 21:ijms21186760. [PMID: 32942624 PMCID: PMC7555992 DOI: 10.3390/ijms21186760] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/12/2020] [Accepted: 09/11/2020] [Indexed: 12/12/2022] Open
Abstract
Dietary supplementation with spray-dried porcine plasma (SDP) can modulate the immune response of gut-associated lymphoid tissue. SDP supplementation reduces acute mucosal inflammation, as well as chronic inflammation associated with aging. The aim of this study was to analyze if SDP supplementation could ameliorate colitis in a genetic mouse model of inflammatory bowel disease (IBD). Wild-type mice and Mdr1a knockout (KO) mice were administered a control diet or an SDP-supplemented diet from day 21 (weaning) until day 56. The histopathological index, epithelial barrier, and intestinal immune system were analyzed in the colonic mucosa. KO mice had higher epithelial permeability, increased Muc1 and Muc4 expression, and lower abundance of E-cadherin and Muc2 (all p < 0.001). SDP prevented these effects (all p < 0.05) and decreased the colonic inflammation observed in KO mice, reducing neutrophil and monocyte infiltration and activation and the percentage of activated T helper lymphocytes in the colonic mucosa (all p < 0.05). SDP also diminished proinflammatory cytokine expression and increased the anti-inflammatory IL-10 concentration in the colonic mucosa (all p < 0.05). In conclusion, dietary supplementation with SDP enhances colon barrier function and reduces mucosal inflammation in a mouse model of IBD.
Collapse
|
43
|
Li C, Ma D, Zhou H, Zhang M, An L, Wang Y, Wu C. Effects of different doses lipopolysaccharides on the mucosal barrier in mouse intestine. Res Vet Sci 2020; 133:75-84. [PMID: 32947071 DOI: 10.1016/j.rvsc.2020.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/26/2020] [Accepted: 09/07/2020] [Indexed: 12/22/2022]
Abstract
Lipopolysaccharide (LPS), an important component in the outer membrane of the cell wall of Gram-negative bacteria, can induce a systemic inflammatory response and play an important role in bacterial infection and disease evolution. The thick layer of mucus covering the small intestinal villus acts primarily to the first barrier from damage by toxic substances. We aimed to study the effects of LPS on the intestinal mucus layer barrier. The results showed that the thickness of the mucus layer was significantly increased by a low dose of LPS. Further, LPS can cross this barrier into the blood, put the body in a state of chronic low-grade inflammation, and activate the body's immune response. However, after a long-term high dose of LPS exposure, a large number of lysosomes in goblet cells caused a loss of function, and mucus layer thickness was significantly decreased. A large amount of LPS stuck to the mucus, leading to normal LPS and inflammatory cytokines level of plasma. The intestinal tissue morphology was damaged, and many immune cells died through necrosis in the intestine. Collectively, the function of the goblet cell was normal, a low dose of LPS cannot be stuck to the mucus layer. However, a high dose of LPS stuck to the mucus when goblet cells caused a loss of function, which can be directly linked to the severity of the immunosuppression in the body.
Collapse
Affiliation(s)
- Chao Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - DeXue Ma
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Hongchao Zhou
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Mingming Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Liyan An
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yi Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Chenchen Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
44
|
Grondin JA, Kwon YH, Far PM, Haq S, Khan WI. Mucins in Intestinal Mucosal Defense and Inflammation: Learning From Clinical and Experimental Studies. Front Immunol 2020; 11:2054. [PMID: 33013869 PMCID: PMC7500085 DOI: 10.3389/fimmu.2020.02054] [Citation(s) in RCA: 209] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/28/2020] [Indexed: 12/24/2022] Open
Abstract
Throughout the gastrointestinal (GI) tract, a distinct mucus layer composed of highly glycosylated proteins called mucins plays an essential role in providing lubrication for the passage of food, participating in cell signaling pathways and protecting the host epithelium from commensal microorganisms and invading pathogens, as well as toxins and other environmental irritants. These mucins can be broadly classified into either secreted gel-forming mucins, those that provide the structural backbone for the mucus barrier, or transmembrane mucins, those that form the glycocalyx layer covering the underlying epithelial cells. Goblet cells dispersed among the intestinal epithelial cells are chiefly responsible for the synthesis and secretion of mucins within the gut and are heavily influenced by interactions with the immune system. Evidence from both clinical and animal studies have indicated that several GI conditions, including inflammatory bowel disease (IBD), colorectal cancer, and numerous enteric infections are accompanied by considerable changes in mucin quality and quantity. These changes include, but are not limited to, impaired goblet cell function, synthesis dysregulation, and altered post-translational modifications. The current review aims to highlight the structural and functional features as well as the production and immunological regulation of mucins and the impact these key elements have within the context of barrier function and host defense in intestinal inflammation.
Collapse
Affiliation(s)
- Jensine A Grondin
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Yun Han Kwon
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Parsa Mehraban Far
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Sabah Haq
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Waliul I Khan
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
45
|
Duan L, Huang H, Zhao X, Zhou M, Chen S, Wang C, Han Z, Han ZC, Guo Z, Li Z, Cao X. Extracellular vesicles derived from human placental mesenchymal stem cells alleviate experimental colitis in mice by inhibiting inflammation and oxidative stress. Int J Mol Med 2020; 46:1551-1561. [PMID: 32945344 PMCID: PMC7447323 DOI: 10.3892/ijmm.2020.4679] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are pluripotent cells that can be applied to the treatment of immune disorders, including inflammatory bowel disease (IBD). The therapeutic effects of MSCs have been mostly attributed to the secretion of soluble factors with paracrine actions, such as extracellular vesicles (EVs), which may play a relevant role in the repair of damaged tissues. In the present study, a mouse model of colitis was induced with the use of trinitrobenzene sulfonic acid (TNBS). EVs derived from human placental mesenchymal stem cells (hP‑MSCs) were used for the treatment of colitis by in situ injection. Clinical scores were applied to verify the therapeutic effects of EVs on mice with colitis. Inflammation in the colon was evaluated by measuring the levels of various inflammatory cytokines. The content of reactive oxygen species (ROS) was detected by the use of molecular imaging methods for real‑time tracking and the therapeutic effects of EVs on mucosal healing in mice with colitis were evaluated. The results revealed that the injection of EVs regulated the balance of pro‑inflammatory and anti‑inflammatory cytokines in colon tissue. Treatment with EVs also suppressed oxidative stress by decreasing the activity of myeloperoxidase (MPO) and ROS. Histological analysis further confirmed that the EVs significantly promoted mucosal healing, as reflected by the promotion of the proliferation of colonic epithelial cells and the maintenance of tight junctions. Taken together, the findings of the present study demonstrated that EVs derived from hP‑MSCs alleviated TNBS‑induced colitis by inhibiting inflammation and oxidative stress. These findings may provide a novel theoretical basis for the EV‑based treatment of IBD.
Collapse
Affiliation(s)
- Liyun Duan
- Department of Hepato-Gastroenterology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300071, P.R. China
| | - Haoyan Huang
- School of Medicine, Nankai University, Tianjin 300071, P.R. China
| | - Xiaotong Zhao
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Manqian Zhou
- Department of Radiation Oncology, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| | - Shang Chen
- School of Medicine, Nankai University, Tianjin 300071, P.R. China
| | - Chen Wang
- School of Medicine, Nankai University, Tianjin 300071, P.R. China
| | - Zhibo Han
- Beijing Engineering Laboratory of Perinatal Stem Cells, Beijing Institute of Health and Stem Cells, Health and Biotech Co., Beijing 100176, P.R. China
| | - Zhong-Chao Han
- Beijing Engineering Laboratory of Perinatal Stem Cells, Beijing Institute of Health and Stem Cells, Health and Biotech Co., Beijing 100176, P.R. China
| | - Zhikun Guo
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Zongjin Li
- School of Medicine, Nankai University, Tianjin 300071, P.R. China
| | - Xiaocang Cao
- Department of Hepato-Gastroenterology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300071, P.R. China
| |
Collapse
|
46
|
Vacca M, Celano G, Calabrese FM, Portincasa P, Gobbetti M, De Angelis M. The Controversial Role of Human Gut Lachnospiraceae. Microorganisms 2020; 8:E573. [PMID: 32326636 PMCID: PMC7232163 DOI: 10.3390/microorganisms8040573] [Citation(s) in RCA: 823] [Impact Index Per Article: 205.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/05/2020] [Accepted: 04/13/2020] [Indexed: 02/06/2023] Open
Abstract
The complex polymicrobial composition of human gut microbiota plays a key role in health and disease. Lachnospiraceae belong to the core of gut microbiota, colonizing the intestinal lumen from birth and increasing, in terms of species richness and their relative abundances during the host's life. Although, members of Lachnospiraceae are among the main producers of short-chain fatty acids, different taxa of Lachnospiraceae are also associated with different intra- and extraintestinal diseases. Their impact on the host physiology is often inconsistent across different studies. Here, we discuss changes in Lachnospiraceae abundances according to health and disease. With the aim of harnessing Lachnospiraceae to promote human health, we also analyze how nutrients from the host diet can influence their growth and how their metabolites can, in turn, influence host physiology.
Collapse
Affiliation(s)
- Mirco Vacca
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.V.); (F.M.C.); (M.D.A.)
| | - Giuseppe Celano
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.V.); (F.M.C.); (M.D.A.)
| | - Francesco Maria Calabrese
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.V.); (F.M.C.); (M.D.A.)
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70121 Bari, Italy
| | - Marco Gobbetti
- Faculty of Science and Technology, Free University of Bozen, 39100 Bolzano, Italy;
| | - Maria De Angelis
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.V.); (F.M.C.); (M.D.A.)
| |
Collapse
|
47
|
Ma S, Yeom J, Lim YH. Dairy Propionibacterium freudenreichii ameliorates acute colitis by stimulating MUC2 expression in intestinal goblet cell in a DSS-induced colitis rat model. Sci Rep 2020; 10:5523. [PMID: 32218552 PMCID: PMC7099060 DOI: 10.1038/s41598-020-62497-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 03/09/2020] [Indexed: 12/21/2022] Open
Abstract
An intact mucus layer is important in managing inflammatory bowel disease (IBD). Dairy Propionibacterium freudenreichii has probiotic potential, produces propionic acid and is known to promote health. The aim of this study was to evaluate the effects of P. freudenreichii on the improvement of colitis. LS 174T goblet cells and a dextran sodium sulfate (DSS)-induced colitis rat model were used to investigate the P. freudenreichii-induced stimulation of mucin production in vitro and in vivo, respectively. The mRNA and protein expression levels of MUC2, a main component of intestinal mucus, increased in the supernatant of P. freudenreichii culture (SPFC)-treated LS 174 cells. The SPFC and live P. freudenreichii (LPF) reduced the disease activity index (DAI) in the rats with DSS-induced colitis. After treatment with SPFC or LPF, the mRNA levels of typical pro-inflammatory cytokines decreased and the inflammatory state was histologically improved in the rats with DSS-induced colitis. The SPFC and LPF treatments increased the gene and protein expression levels of MUC2 in the rats with DSS-induced colitis compared with the expression levels in the negative control rats, and immunohistochemistry (IHC) showed an increase of the intestinal MUC2 level. In addition, SPFC and LPF augmented the level of propionate in the faeces of the rats with DSS-induced colitis. In conclusion, P. freudenreichii might improve acute colitis by restoring goblet cell number and stimulating the expression of MUC2 in intestinal goblet cells.
Collapse
Affiliation(s)
- Seongho Ma
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul, 02841, Republic of Korea
| | - Jiah Yeom
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul, 02841, Republic of Korea
| | - Young-Hee Lim
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul, 02841, Republic of Korea. .,Department of Public Health Science (Brain Korea 21 PLUS program), Graduate School, Korea University, Seoul, 02841, Republic of Korea. .,Department of Laboratory Medicine, Korea University Guro Hospital, Seoul, 08308, Republic of Korea.
| |
Collapse
|
48
|
Pascoal C, Francisco R, Ferro T, Dos Reis Ferreira V, Jaeken J, Videira PA. CDG and immune response: From bedside to bench and back. J Inherit Metab Dis 2020; 43:90-124. [PMID: 31095764 DOI: 10.1002/jimd.12126] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/13/2019] [Accepted: 05/15/2019] [Indexed: 12/20/2022]
Abstract
Glycosylation is an essential biological process that adds structural and functional diversity to cells and molecules, participating in physiological processes such as immunity. The immune response is driven and modulated by protein-attached glycans that mediate cell-cell interactions, pathogen recognition and cell activation. Therefore, abnormal glycosylation can be associated with deranged immune responses. Within human diseases presenting immunological defects are congenital disorders of glycosylation (CDG), a family of around 130 rare and complex genetic diseases. In this review, we have identified 23 CDG with immunological involvement, characterized by an increased propensity to-often life-threatening-infection. Inflammatory and autoimmune complications were found in 7 CDG types. CDG natural history(ies) and the mechanisms behind the immunological anomalies are still poorly understood. However, in some cases, alterations in pathogen recognition and intracellular signaling (eg, TGF-β1, NFAT, and NF-κB) have been suggested. Targeted therapies to restore immune defects are only available for PGM3-CDG and SLC35C1-CDG. Fostering research on glycoimmunology may elucidate the involved pathophysiological mechanisms and open new therapeutic avenues, thus improving CDG patients' quality of life.
Collapse
Affiliation(s)
- Carlota Pascoal
- Portuguese Association for CDG, Lisbon, Portugal
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Rita Francisco
- Portuguese Association for CDG, Lisbon, Portugal
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Tiago Ferro
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Vanessa Dos Reis Ferreira
- Portuguese Association for CDG, Lisbon, Portugal
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
| | - Jaak Jaeken
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
- Center for Metabolic Diseases, Department of Development and Regeneration, UZ and KU Leuven, Leuven, Belgium
| | - Paula A Videira
- Portuguese Association for CDG, Lisbon, Portugal
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| |
Collapse
|
49
|
Jiang Z, Wang H, Li L, Hou Z, Liu W, Zhou T, Li Y, Chen S. Analysis of TGCA data reveals genetic and epigenetic changes and biological function of MUC family genes in colorectal cancer. Future Oncol 2019; 15:4031-4043. [PMID: 31773991 DOI: 10.2217/fon-2019-0363] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aim: Few studies focused on functions and regulatory networks of MUC family members in colorectal cancer based on comprehensive analysis of online database. Materials & methods: Copy number variation, methylation, pathway analysis and drug influence on MUC expression were analyzed based on The Cancer Genome Atlas and GTEx database. Results: Copy number variation analysis showed MUC heterozygous amplification and heterozygous deletion predominate. Methylation of MUC17, MUC12 and MUC4 were found related to gene expression. Function of MUC family genes mainly affects pathways such as apoptosis, cell cycle, DNA damage and EMT pathways. PLX4720, dabrafenib, gefitinib, afatinib and austocystin D can alter the expression of MUC gene. Conclusion: The genetic and epigenetic changes of MUC are related to the level of MUC expression in colorectal cancer.
Collapse
Affiliation(s)
- Zhipeng Jiang
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, 26 Yuancunerheng Road, Guangzhou 510655, Guangdong, PR China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal & Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou 510655, Guangdong, PR China
| | - Huashe Wang
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, 26 Yuancunerheng Road, Guangzhou 510655, Guangdong, PR China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal & Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou 510655, Guangdong, PR China
| | - Liang Li
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, 26 Yuancunerheng Road, Guangzhou 510655, Guangdong, PR China
| | - Zehui Hou
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, 26 Yuancunerheng Road, Guangzhou 510655, Guangdong, PR China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal & Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou 510655, Guangdong, PR China
| | - Wei Liu
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, 26 Yuancunerheng Road, Guangzhou 510655, Guangdong, PR China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal & Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou 510655, Guangdong, PR China
| | - Taicheng Zhou
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, 26 Yuancunerheng Road, Guangzhou 510655, Guangdong, PR China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal & Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou 510655, Guangdong, PR China
| | - Yingru Li
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, 26 Yuancunerheng Road, Guangzhou 510655, Guangdong, PR China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal & Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou 510655, Guangdong, PR China
| | - Shuang Chen
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, 26 Yuancunerheng Road, Guangzhou 510655, Guangdong, PR China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal & Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou 510655, Guangdong, PR China
| |
Collapse
|
50
|
Gut microbiota as an "invisible organ" that modulates the function of drugs. Biomed Pharmacother 2019; 121:109653. [PMID: 31810138 DOI: 10.1016/j.biopha.2019.109653] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 10/30/2019] [Accepted: 11/06/2019] [Indexed: 02/08/2023] Open
Abstract
Gut microbiota plays an important role in the gut and have become a hotspot of recent research interests. Commensal microbiota in gut exert a variety of effects on the host, from shaping the structure and function of the gut and the immune system to the modulation of nutrient status of the host and the treatment outcomes of some drugs. Gut microbiota and its enzyme product and subsequent products, such as short-chain fatty acid and bile acid, play important roles in the biotransformation of drugs via directly or indirectly affecting drug absorption, toxicity, metabolism and bioavailability. Drugs, especially antibiotics, also affect the homeostasis of probiotics and the integrity and function of the intestinal mucosa. These interplaying processes produce a variety of important metabolites of the host and drugs and affect the balance of microbiota and the mucosal barrier then modulate the function of drugs. Gut microbiota imbalance is associated with a broad range of disease mechanisms, and this association denotes a new drug-therapeutic avenue. The present review summarizes how gut microbiota acts as an "invisible organ" to directly or indirectly modulate the function of drugs, on the aspects of probiotic homeostasis, drugs and host nutritional metabolism, AJC, mucus layer and microfold cells.
Collapse
|