1
|
Chieppa M, De Santis S, Verna G. Winnie Mice: A Chronic and Progressive Model of Ulcerative Colitis. Inflamm Bowel Dis 2025; 31:1158-1167. [PMID: 39912845 PMCID: PMC11985403 DOI: 10.1093/ibd/izaf006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Indexed: 02/07/2025]
Abstract
Recent trends show a continuous worldwide rise in the incidence of ulcerative colitis (UC), leading to increased interest in its etiology and pathogenesis, which is currently unknown. To gain a better mechanistic understanding of this disease, many mouse models have been developed over the last several years, with variations of dextran sodium sulfate administration representing the most widely employed. The Winnie mouse strain was created through elicited random mutations in Muc2, resulting in a progressive, chronic intestinal inflammation localized to the colon that worsens over time. Moreover, Winnie mice display immunologic and microbiota features that are similar to those that can be found in UC patients. Phenotypically, the presence, albeit rare, of rectal prolapse and other complications impacting quality of life can be observed in Winnie mice, as well as extraintestinal manifestations that are often associated with UC. While Winnie mice are currently less studied compared to other more established models of colitis, much has been discovered in the initial years of its use as a UC-like model. In summary, the use of Winnie mice adds to the growing armamentarium that is required to develop precision-based medicine for its future application in treating complex multifactorial diseases, such as UC.
Collapse
Affiliation(s)
- Marcello Chieppa
- Department of Experimental Medicine, University of Salento, 73100 Lecce, Italy
| | - Stefania De Santis
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Giulio Verna
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
2
|
Kumar A, Rahul, Kanika, Kumar J, Mahajan S, Ali A, Ali N, Bishnoi M, Son YO, Khan R. Multifunctional chrysin-loaded gallic acid-glycerol monostearate conjugate-based injectable hydrogel for targeted inhibition of hypoxia-induced NLRP3 inflammasome in ulcerative colitis. Biomater Sci 2025; 13:1801-1817. [PMID: 39998176 DOI: 10.1039/d4bm01700e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory condition affecting the colon part of the large intestine. Since there is no cure for this disease, conventional therapies only provide symptomatic relief. Recently, phytomolecules have shown promising treatment results in various diseases. However, short half-life, hydrophobicity, and poor bioavailability limit their therapeutic potential. To overcome all these challenges, we have earlier conjugated a phytomolecule (gallic acid) (GA) with the FDA-approved generally recognized as safe (GRAS) material that is glycerol monostearate (GMS). This GA-GMS conjugate self-assembles as a hydrogel via the heating-cooling method and acts as a pro-drug of GA. The in vivo imaging results suggest that the GA-GMS hydrogel more efficiently adheres to the inflamed colon than a therapeutic enema. Additionally, it is known that the gut microbiota exaggerates UC by creating a hypoxic environment in the colon. This hypoxia is linked with NLRP3 inflammasome activation that triggers the release of IL-1β and IL-18 that downregulates MUC2 protein expression in the colon, responsible for mucin secretion in the colon. Therefore, chrysin (CR) (HIF-1α inhibitor) is encapsulated into the GA-GMS hydrogel to target hypoxia. The CR@GA-GMS hydrogel follows the enzyme-responsive release of the CR and restores DSS-induced damage to colonic tissue. Furthermore, the CR@GA-GMS hydrogel downregulates HIF-1α mediated NLRP3 inflammasome signalling while upregulating MUC2 production.
Collapse
Affiliation(s)
- Ajay Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar) 140306, Punjab, India.
| | - Rahul
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar) 140306, Punjab, India.
| | - Kanika
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar) 140306, Punjab, India.
| | - Jattin Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar) 140306, Punjab, India.
| | - Shubham Mahajan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar) 140306, Punjab, India.
| | - Aneesh Ali
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar) 140306, Punjab, India.
| | - Nemat Ali
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Mahendra Bishnoi
- National Agri-Food Biotechnology Institute, Mohali, Punjab 140306, India
| | - Young-Ok Son
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si, 63243, Republic of Korea
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar) 140306, Punjab, India.
| |
Collapse
|
3
|
Yu X, Li G, Zhou S, Liu X, Yin F, Li D, Zhou D. Glycosylation of oyster peptides by COS ameliorates zinc deficiency-induced syndromes: intestinal inflammation and imbalance of the gut microbiota in vivo. Food Funct 2025; 16:640-656. [PMID: 39714215 DOI: 10.1039/d4fo04200j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Zinc is essential for maintaining the integrity and repair of small intestinal epithelial cells while zinc deficiency could induce the inflammatory infiltration and imbalance of intestinal flora in the intestine. In this study, glycosylation between oyster protein hydrolysate (OPH) and chitosan oligosaccharide (COS) was conducted and used as the carrier of zinc ions (OCZn). The results of zeta potential and particle size distribution showed that the OPH-COS successfully bound to zinc ions to form OCZn with a surface zinc content of 0.56% (scanning electron microscopy). In addition, OCZn was found to exhibit good intestinal digestion by in vitro simulated digestion microscopy, while TSQ fluorescence staining revealed the presence of free zinc ions released from OCZn in the intestinal cells. In the zinc deficiency-induced mouse model, a moderate dose of OCZn (zinc: 6.96 mg kg-1) showed significant restorative effects on colonic inflammation (IL-1β: 28.20 pg per mg·protein, IL-6: 27.73 pg per mg·protein), protein expressions of HO-1 and ZO-1, oxidative stress (the liver and kidneys), and imbalance of the gut microbiota, increasing microbial diversity and abundance (ratio of Firmicutes/Bacteroides). Zinc deficiency triggered the abundance of Proteobacteria (risk of diseases), while the dominant bacteria were mainly restored to Bacteroides, Parabacteroides, Alistipes, Alloprevotella, and Muribaculaceae following the administration of OCZn. This study provided a theoretical basis for improving the inflammatory infiltration of the colon and the imbalance of intestinal flora caused by zinc deficiency.
Collapse
Affiliation(s)
- Xuening Yu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Liaoning Province Key Laboratory for Marine Food Science and Technology, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China.
| | - Guang Li
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Liaoning Province Key Laboratory for Marine Food Science and Technology, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China.
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 23060, China
| | - Shuling Zhou
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Liaoning Province Key Laboratory for Marine Food Science and Technology, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China.
| | - Xiaoyang Liu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Liaoning Province Key Laboratory for Marine Food Science and Technology, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China.
| | - Fawen Yin
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Liaoning Province Key Laboratory for Marine Food Science and Technology, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China.
| | - Deyang Li
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Liaoning Province Key Laboratory for Marine Food Science and Technology, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China.
| | - Dayong Zhou
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Liaoning Province Key Laboratory for Marine Food Science and Technology, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China.
| |
Collapse
|
4
|
Matos MS, Ávila-Gálvez MÁ, González-Sarrías A, Silva NV, Crespo CL, Jacinto A, Serra AT, Matias AA, Nunes Dos Santos C. Unveiling the anti-inflammatory potential of 11β,13-dihydrolactucin for application in inflammatory bowel disease management. Food Funct 2024; 15:9254-9271. [PMID: 39162124 DOI: 10.1039/d4fo01446d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Management of inflammatory bowel disease (IBD) poses significant challenges, and there is a need for innovative therapeutic approaches. This study investigates the anti-inflammatory properties of the dietary sesquiterpene lactone (SL) 11β,13-dihydrolactucin, which can be found in chicory, in three distinct complementary models of intestinal inflammation (two cell models and a zebrafish model), offering comprehensive insights into its potential application for IBD treatment alternatives. In a triple cell co-culture composed of Caco-2, HT29-MTX-E12, and Raji B, 11β,13-dihydrolactucin demonstrated remarkable anti-inflammatory activity at several levels of the cellular inflammatory response. Notably, 11β,13-dihydrolactucin prevented the activation of critical signalling pathways associated with inflammation, namely NF-κB and MAPK p38. This SL also decreased the release of the neutrophil-recruiting chemokine IL-8. Additionally, the compound reduced the gene expression of IL-6 and TNF-α, as well as the gene and protein expression of the inflammatory inducible enzymes iNOS and COX-2. In a myofibroblast-like human cell model, 11β,13-dihydrolactucin decreased the release of the cytokine TNF-α and the COX-2-derived inflammation mediator PGE2. Finally, in a zebrafish model of gut inflammation, 11β,13-dihydrolactucin effectively reduced neutrophil infiltration, further supporting its anti-inflammatory efficacy in a physiological context. Collectively, our findings highlight the promising anti-inflammatory potential of 11β,13-dihydrolactucin across various facets of intestinal inflammation, providing a foundation for the consideration of chicory as a promising candidate for incorporation in food or nutraceutical products for the potential prevention of IBD.
Collapse
Affiliation(s)
- Melanie S Matos
- Instituto de Biologia Experimental e Tecnológica (iBET), 2780-157 Oeiras, Portugal.
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB-NOVA), 2780-157 Oeiras, Portugal
| | - María Ángeles Ávila-Gálvez
- Instituto de Biologia Experimental e Tecnológica (iBET), 2780-157 Oeiras, Portugal.
- NOVA Medical School, Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - Antonio González-Sarrías
- Laboratory of Food & Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, CEBAS-CSIC, 30100 Campus de Espinardo, Murcia, Spain
| | - Nuno-Valério Silva
- NOVA Medical School, Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - Carolina Lage Crespo
- NOVA Medical School, Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - António Jacinto
- NOVA Medical School, Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
- NOVA Institute for Medical Systems Biology, NIMSB, Universidade Nova de Lisboa, 1099-085 Lisboa, Portugal
| | - Ana Teresa Serra
- Instituto de Biologia Experimental e Tecnológica (iBET), 2780-157 Oeiras, Portugal.
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB-NOVA), 2780-157 Oeiras, Portugal
| | - Ana A Matias
- Instituto de Biologia Experimental e Tecnológica (iBET), 2780-157 Oeiras, Portugal.
| | - Cláudia Nunes Dos Santos
- Instituto de Biologia Experimental e Tecnológica (iBET), 2780-157 Oeiras, Portugal.
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB-NOVA), 2780-157 Oeiras, Portugal
- NOVA Medical School, Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
- NOVA Institute for Medical Systems Biology, NIMSB, Universidade Nova de Lisboa, 1099-085 Lisboa, Portugal
| |
Collapse
|
5
|
Wang Q, Zhang G, An C, Hambly BD, Bao S. The role of IL-37 in gastrointestinal diseases. Front Immunol 2024; 15:1431495. [PMID: 39206201 PMCID: PMC11349528 DOI: 10.3389/fimmu.2024.1431495] [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: 05/12/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Gastrointestinal mucosal surface is frequently under challenge due to it's the large surface area and most common entry of microbes. IL-37, an anti-inflammatory cytokine, regulates local and systemic host immunity. H. pylori infection leads to the inhibition of IL-37 in the gastric mucosa, contributing to heightened mucosal inflammation and destruction, thereby facilitating increased proliferation of H. pylori. Food allergy, due to immune dysregulation, also contribute to GI injury. On the other hand, elevated levels of IL-37 observed in gastric cancer patients align with reduced host immunity at the cellular and humoral levels, indicating that IL-37 may contribute to the development of gastric cancer via suppressing pro-inflammatory responses. While IL-37 provides protection in an IBD animal model, the detection of highly produced IL-37 in IBD patients suggests a stage-dependent role, being protective in acute inflammation but potentially exacerbates the development of IBD in chronic conditions. Moreover, elevated colonic IL-37 in CRC correlates with overall survival time and disease time, indicating a protective role for IL-37 in CRC. The differential regulation and expression of IL-37 between upper- and lower-GI organs may be attributed to variations in the microbial flora. This information suggests that IL-37 could be a potential therapeutic agent, depending on the stage and location.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Anatomy, School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Guangrun Zhang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Caiping An
- Department of Nephrology, Gansu Provincial Hospital, Lanzhou, China
| | - Brett D. Hambly
- Centre for Healthy Futures, Torrens University Australia, Sydney, NSW, Australia
| | - Shisan Bao
- Foreign Affairs Office, The Third Affiliated Hospital of Gansu University of Chinese Medicine, Baiyin, China
- Foreign Affairs Office, The First People’s Hospital of Baiyin, Baiyin, China
| |
Collapse
|
6
|
Ryu HM, Islam SMS, Riaz B, Sayeed HM, Choi B, Sohn S. Immunomodulatory Effects of a Probiotic Mixture: Alleviating Colitis in a Mouse Model through Modulation of Cell Activation Markers and the Gut Microbiota. Int J Mol Sci 2024; 25:8571. [PMID: 39201260 PMCID: PMC11354276 DOI: 10.3390/ijms25168571] [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: 07/12/2024] [Revised: 08/02/2024] [Accepted: 08/02/2024] [Indexed: 09/02/2024] Open
Abstract
Ulcerative colitis (UC) is a persistent inflammatory intestinal disease that consistently affects the colon and rectum. Its exact cause remains unknown. UC causes a considerable challenge in healthcare, prompting research for novel therapeutic strategies. Although probiotics have gained popularity as possible candidates for managing UC, studies are still ongoing to identify the best probiotics or probiotic mixtures for clinical applications. This study aimed to determine the efficacy of a multi-strain probiotic mixture in mitigating intestinal inflammation in a colitis mouse model induced by dextran sulfate sodium. Specifically, a multi-strain probiotic mixture consisting of Tetragenococcus halophilus and Eubacterium rectale was used to study its impact on colitis symptoms. Anti-inflammatory effects were evaluated using ELISA and flow cytometry. The configuration of gut microbial communities was determined using 16S rRNA metagenomic analysis. According to this study, colitis mice treated with the probiotic mixture experienced reduced weight loss and significantly less colonic shortening compared to untreated mice. Additionally, the treated mice exhibited increased levels of forkhead box P3 (Foxp3) and interleukin 10, along with decreased expression of dendritic cell activation markers, such as CD40+, CD80+, and CD83+, in peripheral blood leukocytes and intraepithelial lymphocytes. Furthermore, there was a significant decrease in the frequencies of CD8+N.K1.1+ cells and CD11b+Ly6G+ cells. In terms of the gut microbiota, probiotic-mixture treatment of colitis mice significantly increased the abundance of the phyla Actinobacteria and Verrucomicrobia (p < 0.05). These results provide valuable insights into the therapeutic promise of multi-strain probiotics, shedding light on their potential to alleviate colitis symptoms. This research contributes to the ongoing exploration of effective probiotic interventions for managing inflammatory bowel disease.
Collapse
Affiliation(s)
- Hye-Myung Ryu
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea;
| | - S. M. Shamsul Islam
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (S.M.S.I.); (B.R.); (H.M.S.)
| | - Bushra Riaz
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (S.M.S.I.); (B.R.); (H.M.S.)
| | - Hasan M. Sayeed
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (S.M.S.I.); (B.R.); (H.M.S.)
| | - Bunsoon Choi
- Institute of Medical Science, Ajou University School of Medicine, Suwon 16499, Republic of Korea;
| | - Seonghyang Sohn
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea;
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (S.M.S.I.); (B.R.); (H.M.S.)
| |
Collapse
|
7
|
Lee Y, Yoon B, Son S, Cho E, Kim KB, Choi EY, Kim DE. Inhibition of Immunoproteasome Attenuates NLRP3 Inflammasome Response by Regulating E3 Ubiquitin Ligase TRIM31. Cells 2024; 13:675. [PMID: 38667290 PMCID: PMC11048918 DOI: 10.3390/cells13080675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Excessive secretion of pro-inflammatory cytokines leads to the disruption of intestinal barrier in inflammatory bowel disease (IBD). The inflammatory cytokine tumor necrosis factor alpha (TNFα) induces the assembly of the NLRP3 inflammasome, resulting in the augmented secretion of inflammatory cytokines implicated in the pathogenesis of inflammatory bowel disease (IBD). TNFα has also been known to induce the formation of immunoproteasome (IP), which incorporates immunosubunits LMP2, LMP7, and MECL-1. Inhibition of IP activity using the IP subunit LMP2-specific inhibitor YU102, a peptide epoxyketone, decreased the protein levels of NLRP3 and increased the K48-linked polyubiquitination levels of NLRP3 in TNFα-stimulated intestinal epithelial cells. We observed that inhibition of IP activity caused an increase in the protein level of the ubiquitin E3 ligase, tripartite motif-containing protein 31 (TRIM31). TRIM31 facilitated K48-linked polyubiquitination and proteasomal degradation of NLRP3 with an enhanced interaction between NLRP3 and TRIM31 in intestinal epithelial cells. In addition, IP inhibition using YU102 ameliorated the symptoms of colitis in the model mice inflicted with dextran sodium sulfate (DSS). Administration of YU102 in the DSS-treated colitis model mice caused suppression of the NLRP3 protein levels and accompanied inflammatory cytokine release in the intestinal epithelium. Taken together, we demonstrated that inhibiting IP under inflammatory conditions induces E3 ligase TRIM31-mediated NLRP3 degradation, leading to attenuation of the NLRP3 inflammatory response that triggers disruption of intestinal barrier.
Collapse
Affiliation(s)
- Yubin Lee
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (Y.L.); (B.Y.); (S.S.); (E.C.)
| | - Boran Yoon
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (Y.L.); (B.Y.); (S.S.); (E.C.)
| | - Sumin Son
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (Y.L.); (B.Y.); (S.S.); (E.C.)
| | - Eunbin Cho
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (Y.L.); (B.Y.); (S.S.); (E.C.)
| | - Kyung Bo Kim
- Department of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Center for Translational Science at Port St. Lucie, Florida International University, 11350 SW Village Pkwy, Port St. Lucie, FL 34987, USA;
| | - Eun Young Choi
- Department of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Center for Translational Science at Port St. Lucie, Florida International University, 11350 SW Village Pkwy, Port St. Lucie, FL 34987, USA;
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (Y.L.); (B.Y.); (S.S.); (E.C.)
| |
Collapse
|
8
|
Liu H, Feng X, Wang D, Liu L, Liu Y, Liu B, Zhu L, Zhang C, Yang W. Altered metabolome and microbiome features provide clues in predicting recurrence of ulcerative colitis. J Pharm Biomed Anal 2024; 239:115864. [PMID: 38029703 DOI: 10.1016/j.jpba.2023.115864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023]
Abstract
PURPOSE Many studies have shown that the imbalance of the intestinal flora and metabolite can lead to the development of ulcerative colitis (UC), but their role in recurrent-UC is still unclear. We studied the intestinal flora and metabolites associated with recurrent-UC to elucidate the mechanism and biomarkers of recurrent-UC. METHODS Ulcerative colitis (UC) models in active, remission, and recurrence stages were established, and the abundance of intestinal flora was determined by 16 S rRNA sequencing. The changes in the metabolites present in feces and serum were analyzed by UPLC-MS/MS. RESULTS We identified 24 metabolites in feces and serum, which might be used as diagnostic and predictive biomarkers of recurrent-UC. The dominant flora of recurrent-UC included Romboutsia, UCG-005, etc. The results of a network analysis found that long-chain fatty acids and phenylalanine were strongly correlated with Firmicutes and Proteobacteria, which indicated that the recurrence of UC might be closely related to metabolites and microorganisms. CONCLUSION The changes in intestinal microbiota and metabolites are closely related to the development of UC. Microbiota is an important inducer of UC, which can regulate metabolites through the 'microorganism-gut-metabolite' axis. It may provide a new method for the prediction and treatment of UC.
Collapse
Affiliation(s)
- Haifan Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xue Feng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dunfang Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yaqing Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bin Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lin Zhu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Caijuan Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Weipeng Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
9
|
E L, Li W, Hu Y, Deng L, Yao J, Zhou X. Methyl cinnamate protects against dextran sulfate sodium-induced colitis in mice by inhibiting the MAPK signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1806-1818. [PMID: 37654075 PMCID: PMC10686792 DOI: 10.3724/abbs.2023124] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/19/2023] [Indexed: 09/02/2023] Open
Abstract
Effective and non-toxic therapeutic agents are lacking for the prevention and treatment of colitis. Previous studies found that methyl cinnamate (MC), extracted from galangal ( Alpinia officinarum Hance), has anti-inflammatory properties. However, whether MC is effective as anti-colitis therapy remains unknown. In this study, we investigate the therapeutic effects of MC on dextran sulfate sodium (DSS)-induced colitis in mice and further explore its potential mechanism of action. MC treatment relieves symptoms associated with DSS-induced colitis, including the recovery of DSS-induced weight loss, decreases the disease activity index score, and increases the colon length without toxic side effects. MC treatment protects the integrity of the intestinal barrier in mice with DSS-induced colitis and inhibits the overexpression of pro-inflammatory cytokines in vivo and in vitro. Moreover, the MAPK signaling pathway is found to be closely related to the treatment with MC of colitis. Western blot analysis show that phosphorylation of the p38 protein in colon tissues treated with MC is markedly reduced and phosphorylation levels of the p38, JNK and ERK proteins are significantly decreased in RAW 264.7 cells treated with MC, indicating that the mechanism of MC in treating DSS-induced colitis could be achieved by inhibiting the MAPK signaling pathway. Furthermore, 16S RNA sequencing analysis show that MC can improve intestinal microbial dysbiosis in mice with DSS-induced colitis. Altogether, these findings suggest that MC may be a novel therapeutic candidate with anti-colitis efficacy. Furthermore, MC treatment relieves the symptoms of colitis by inhibiting the MAPK signaling pathway and improving the intestinal microbiota.
Collapse
Affiliation(s)
- Lilin E
- Department of Biochemistry and Molecular BiologySun Yat-sen University Zhongshan School of MedicineSun Yat-sen UniversityGuangzhou510080China
| | - Wenjie Li
- The First Affiliated HospitalSun Yat-Sen UniversityGuangzhou510080China
| | - Yuanjia Hu
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauMacao SAR 999078China
| | - Lijuan Deng
- Formula-Pattern Research CenterSchool of Traditional Chinese MedicineJinan UniversityGuangzhou510632China
| | - Jianping Yao
- The First Affiliated HospitalSun Yat-Sen UniversityGuangzhou510080China
| | - Xingwang Zhou
- Department of Biochemistry and Molecular BiologySun Yat-sen University Zhongshan School of MedicineSun Yat-sen UniversityGuangzhou510080China
| |
Collapse
|
10
|
Ekhtiar M, Ghasemi-Dehnoo M, Mirzaei Y, Azadegan-Dehkordi F, Amini-Khoei H, Lorigooini Z, Samiei-Sefat A, Bagheri N. The coumaric acid and syringic acid ameliorate acetic acid-induced ulcerative colitis in rats via modulator of Nrf2/HO-1 and pro-inflammatory cytokines. Int Immunopharmacol 2023; 120:110309. [PMID: 37182450 DOI: 10.1016/j.intimp.2023.110309] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/27/2023] [Accepted: 05/05/2023] [Indexed: 05/16/2023]
Abstract
BACKGROUND Ulcerative colitis (UC) is an inflammatory bowel disease (IBD) that causes uncontrolled inflammation and ulcers in your digestive tract. The coumaric acid and syringic acid are phenolic derivative found in many fruits and vegetables and is widely recognized for the ability of anti-parasitic, anti-microbial, anti-viral, anti-inflammatory, and antioxidant. The purpose of this study was to investigate the anti-inflammatory and antioxidant properties of coumaric acid and syringic acid on acetic acid-induced colitis in rats. METHODS A total of 64 male Wistar rats were divided into eight equal groups (n = 8). Colitis was induced by intrarectal administration of acetic acid, and rats orally received coumaric acid (100 and 150 mg/kg), syringic acid (10, 25, and 50 mg/kg), and dexamethasone (2 mg/kg) once per day for four days after colitis induction. Then, HO-1, Nrf2, and NQO1 mRNA expression were quantified by real time-PCR. Finally, the tissue levels of TNF-α and IL-1β protein were measured by ELISA. RESULTS Colitis led to a decrease in HO-1, Nrf2, and NQO1 mRNA expression and an increase in the tissue levels of TNF-α and IL-1β protein in the colon tissue. Treatment with dexamethasone significantly increased HO-1, Nrf2, and NQO1 mRNA expression and decreased the tissue levels of TNF-α and IL-1β protein compared to the UC group. Treatment with 150 mg/kg of coumaric acid and 50 mg/kg of syringic acid significantly increased HO-1, Nrf2, and NQO1 mRNA expression compared to the UC group. Also, treatment with 100 and 150 mg/kg of coumaric acid and 10, 25, and 50 mg/kg of syringic acid significantly decreased the tissue levels of TNF-α and IL-1β protein compared to the UC group. CONCLUSION The coumaric acid and syringic acid, especially at high doses, may be an alternative strategy for the treatment of UC by the reduction of TNF-α and IL-1β levels and upregulation of the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Mahsa Ekhtiar
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Maryam Ghasemi-Dehnoo
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Yousef Mirzaei
- Department of Medical Biochemical Analysis, Cihan University-Erbil, Kurdistan Region, Iraq
| | - Fatemeh Azadegan-Dehkordi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Hossein Amini-Khoei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Lorigooini
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Azadeh Samiei-Sefat
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran; Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
11
|
Maleitzke T, Dietrich T, Hildebrandt A, Weber J, Appelt J, Jahn D, Otto E, Zocholl D, Jiang S, Baranowsky A, Duda GN, Tsitsilonis S, Keller J. Inactivation of the gene encoding procalcitonin prevents antibody-mediated arthritis. Inflamm Res 2023; 72:1069-1081. [PMID: 37039837 DOI: 10.1007/s00011-023-01719-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/25/2023] [Accepted: 03/09/2023] [Indexed: 04/12/2023] Open
Abstract
BACKGROUND Procalcitonin (PCT) is applied as a sensitive biomarker to exclude bacterial infections in patients with rheumatoid arthritis (RA) flare-ups. Beyond its diagnostic value, little is known about the pathophysiological role of PCT in RA. METHODS Collagen antibody-induced arthritis (CAIA) was induced in Calca-deficient mice (Calca-/-), lacking PCT (n = 15), and wild-type (WT) mice (n = 13), while control (CTRL) animals (n = 8 for each genotype) received phosphate-buffered saline. Arthritis severity and grip strength were assessed daily for 10 or 48 days. Articular inflammation, cartilage degradation, and bone lesions were assessed by histology, gene expression analysis, and µ-computed tomography. RESULTS Serum PCT levels and intra-articular PCT expression increased following CAIA induction. While WT animals developed a full arthritic phenotype, Calca-deficient mice were protected from clinical and histological signs of arthritis and grip strength was preserved. Cartilage turnover markers and Tnfa were exclusively elevated in WT mice. Calca-deficient animals expressed increased levels of Il1b. Decreased bone surface and increased subchondral bone porosity were observed in WT mice, while Calca-deficiency preserved bone integrity. CONCLUSION The inactivation of Calca and thereby PCT provided full protection from joint inflammation and arthritic bone loss in mice exposed to CAIA. Together with our previous findings on the pathophysiological function of Calca-derived peptides, these data indicate an independent pro-inflammatory role of PCT in RA.
Collapse
Affiliation(s)
- Tazio Maleitzke
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin, Germany
| | - Tamara Dietrich
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
| | - Alexander Hildebrandt
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
| | - Jérôme Weber
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
| | - Jessika Appelt
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
| | - Denise Jahn
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
| | - Ellen Otto
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
| | - Dario Zocholl
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Biometry and Clinical Epidemiology, Berlin, Germany
| | - Shan Jiang
- University Medical Center Hamburg-Eppendorf, Department of Trauma and Orthopedic Surgery, Hamburg, Germany
| | - Anke Baranowsky
- University Medical Center Hamburg-Eppendorf, Department of Trauma and Orthopedic Surgery, Hamburg, Germany
| | - Georg N Duda
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
| | - Serafeim Tsitsilonis
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
| | - Johannes Keller
- University Medical Center Hamburg-Eppendorf, Department of Trauma and Orthopedic Surgery, Hamburg, Germany.
| |
Collapse
|
12
|
Patton EA, Cunningham P, Noneman M, Helms HP, Martinez-Muniz G, Sumal AS, Dhameja MK, Unger CA, Alahdami AK, Enos RT, Chatzistamou I, Velázquez KT. Acute Administration of Ojeok-san Ameliorates Pain-like Behaviors in Pre-Clinical Models of Inflammatory Bowel Diseases. Nutrients 2023; 15:nu15071559. [PMID: 37049400 PMCID: PMC10096710 DOI: 10.3390/nu15071559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
(1) Background: Gastrointestinal pain and fatigue are the most reported concerns of patients with inflammatory bowel disease (IBD). Commonly prescribed drugs focus on decreasing excessive inflammation. However, up to 20% of IBD patients in an "inactive" state experience abdominal pain. The medicinal herb Ojeok-san (OJS) has shown promise in the amelioration of visceral pain. However, no research on OJS has been conducted in preclinical models of IBD. The mechanism by which OJS promotes analgesia is still elusive, and it is unclear if OJS possesses addictive properties. (2) Aims: In this study, we examined the potential of OJS to promote analgesic effects and rewarding behavior. Additionally, we investigated if tumor necrosis factor alpha (TNFα) from macrophages is a primary culprit of IBD-induced nociception. (3) Methods: Multiple animal models of IBD were used to determine if OJS can reduce visceral nociception. TNFα-macrophage deficient mice were used to investigate the mechanism of action by which OJS reduces nociceptive behavior. Mechanical sensitivity and operant conditioning tests were used to determine the analgesic and rewarding effects of OJS. Body weight, colon length/weight, blood in stool, colonic inflammation, and complete blood count were assessed to determine disease progression. (4) Results: OJS reduced the evoked mechanical nociception in the dextran sulphate sodium model of colitis and IL-10 knockout (KO) mice and delayed aversion to colorectal distension in C57BL/6 mice. No rewarding behavior was observed in OJS-treated IL-10 KO and mdr1a KO mice. The analgesic effects of OJS are independent of macrophage TNFα levels and IBD progression. (5) Conclusions: OJS ameliorated elicited mechanical and visceral nociception without producing rewarding effects. The analgesic effects of OJS are not mediated by macrophage TNFα.
Collapse
Affiliation(s)
- Emma A Patton
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Patrice Cunningham
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Matthew Noneman
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Henry P Helms
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Gustavo Martinez-Muniz
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Aman S Sumal
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Milan K Dhameja
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Christian A Unger
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Ahmed K Alahdami
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Reilly T Enos
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Ioulia Chatzistamou
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Kandy T Velázquez
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| |
Collapse
|
13
|
Wang Q, Xu K, Cai X, Wang C, Cao Y, Xiao J. Rosmarinic Acid Restores Colonic Mucus Secretion in Colitis Mice by Regulating Gut Microbiota-Derived Metabolites and the Activation of Inflammasomes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:4571-4585. [PMID: 36883243 DOI: 10.1021/acs.jafc.2c08444] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Maintaining a steady state of mucus barrier is an important potential target for polyphenol to exert its anticolitis activity. This study elucidates the pivotal role of polyphenol rosmaric acid (RA) in regulating the mucus barrier function and alleviating inflammation by identifying its gut microbiota-derived metabolites and evaluating its inhibitory effect on inflammasomes in colitis mice. Results demonstrated that RA treatment promoted the proliferation of goblet cells and restored the level of mucus secretion, especially Muc2. RA reshaped the microbiota of colitis mice, particularly the boost of core probiotics, such as p. Bacteroidaceae, f. Muribaculaceae, g. Muribaculaceae, g. Alistipes, and g. Clostridia_UCG-014. Nontargeted metabonomics and targeted metabonomics confirmed a significant increase in the bile acids and their metabolites (7-sulfocholic acid, stercobilin, chenodeoxycholic acid 3-sulfate, chenodeoxycholic acid sulfate, and ursodeoxycholic acid 3-sulfate), indole metabolites ((R)-2,3-dihydro-3,5-dihydroxy-2-oxo-3-indoleacetic acid, frovatriptan, 3-formyl-6-hydroxyindole, and brassicanal A), and short-chain fatty acids (SCFAs) (acetic acid, butyric acid, isobutyric acid, isovaleric acid, and valeric acid) that contributed to the strengthened mucus barrier function. In addition, being absorbed mainly in the lower digestive tract, RA inhibited the overexpression of inflammasomes (especially NLRP6) that occurred in colitis mice to promote the mucus secretion of goblet cells. These data confirmed that RA, as a promising candidate to enhance gut health, restored colonic mucus secretion in colitis mice by mediating the production of gut microbiota-derived metabolites and the overexpression of inflammasomes. The presented study provides scientific evidence explaining the apparent paradox of low bioavailability and high bioactivity in polyphenols.
Collapse
Affiliation(s)
- Qun Wang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Kangjie Xu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Xu Cai
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Chujing Wang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
14
|
Deletion of TNF in Winnie- APCMin/+ Mice Reveals Its Dual Role in the Onset and Progression of Colitis-Associated Colorectal Cancer. Int J Mol Sci 2022; 23:ijms232315145. [PMID: 36499472 PMCID: PMC9737576 DOI: 10.3390/ijms232315145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
Colorectal cancer (CRC) is among the best examples for depicting the relationship between inflammation and cancer. The introduction of new therapeutics targeting inflammatory mediators showed a marked decrease in the overall risk of CRC, although their chemopreventive potential is still debated. Specifically, a monoclonal antibody that blocks tumor necrosis factor (TNF), infliximab, increases CRC risk in inflammatory bowel disease patients. To address the axis between TNF and CRC development and progression, we depleted the Tnf from our previously established murine model of colitis-associated cancer (CAC), the Winnie-ApcMin/+ line. We characterized the new Winnie-APCMin/+-TNF-KO line through macroscopical and microscopical analyses. Surprisingly, the latter demonstrated that the deletion of Tnf in Winnie-ApcMin/+ mice resulted in an initial reduction in dysplastic lesion incidence in 5-week-old mice followed by a faster disease progression at 8 weeks. Histological data were confirmed by the molecular profiling obtained from both the real-time PCR analysis of the whole tissue and the RNA sequencing of the macrodissected tumoral lesions from Winnie-APCMin/+-TNF-KO distal colon at 8 weeks. Our results highlight that TNF could exert a dual role in CAC, supporting the promotion of neoplastic lesions onset in the early stage of the disease while inducing their reduction during disease progression.
Collapse
|
15
|
Yoon B, Yun Y, Kim KB, Kim DE. Inhibition of immunoproteasome attenuates NLRP3 inflammasome formation in tumor necrosis factor α-stimulated intestinal epithelial cell. Biochem Biophys Res Commun 2022; 624:157-163. [PMID: 35944388 PMCID: PMC10913474 DOI: 10.1016/j.bbrc.2022.07.120] [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: 07/27/2022] [Revised: 07/29/2022] [Accepted: 07/31/2022] [Indexed: 11/26/2022]
Abstract
Excessive release of inflammatory cytokines has been considered as a major cause of chronic inflammation, resulting in intestinal barrier disruption that leads to inflammatory bowel disease (IBD). Tumor necrosis factor α (TNFα) is one of the well-known inflammatory cytokines that activates formation of NLRP3 inflammasome, thus resulting in excessive secretion of inflammatory cytokines causing IBD. Although immunoproteasome inhibitors have been reported to inhibit inflammatory cytokine release, immunoproteasome inhibition has not yet been addressed for attenuation of NLRP3 inflammasome activity in intestinal epithelial cell. Here, we observed that NLRP3 inflammasome assembly was attenuated by peptide epoxyketone YU102, a LMP2 subunit immunoproteasome inhibitor, in intestinal epithelial cell. YU102 also inhibited maturation of active caspase-1 and secretion of IL-1β, which are subsequent inflammatory cascade after the formation of NLRP3 inflammasome. Progression of epithelial-mesenchymal transition and increase of cellular permeability, which were induced by TNFα, were also suppressed through inhibition of immunoproteasome. Furthermore, we found that YU102 does not inhibit degradation of IкBα and its following NF-кB activation that leads to transcription of NLRP3. These findings suggest that inhibition of immunoproteasome with YU102 offers a potential therapeutic premise for prevention of TNFα-induced chronic inflammation through attenuation of NLRP3 inflammasome assembly.
Collapse
Affiliation(s)
- Boran Yoon
- Department of Bioscience and Biotechnology, Konkuk University, Neundong-ro 120, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Yewon Yun
- Department of Bioscience and Biotechnology, Konkuk University, Neundong-ro 120, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Kyung Bo Kim
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, 40536-0596, United States
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University, Neundong-ro 120, Gwangjin-gu, Seoul, 05029, Republic of Korea.
| |
Collapse
|
16
|
Sun Y, Cai D, Hu W, Fang T. Identifying hub genes and miRNAs in Crohn’s disease by bioinformatics analysis. Front Genet 2022; 13:950136. [PMID: 36118873 PMCID: PMC9471261 DOI: 10.3389/fgene.2022.950136] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/08/2022] [Indexed: 12/04/2022] Open
Abstract
Introduction: Crohn’s disease (CD) is a disease that manifests mainly as chronic inflammation of the gastrointestinal tract, which is still not well understood in terms of its pathogenesis. The aim of this study was to use bioinformatics analysis to identify differentially expressed genes (DEGs) and miRNAs with diagnostic and therapeutic potential in CD. Materials and methods: Three CD datasets (GSE179285, GSE102133, GSE75214) were downloaded from the Gene Expression Omnibus (GEO) database. DEGs between normal and CD tissues were identified using the GEO2R online tool. The Gene Ontology (GO) term and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses of the DEGs were conducted using the clusterProfiler function in the R package. Protein-protein interaction network (PPI) analysis and visualization were performed with STRING and Cytoscape. Ten hub genes were identified using cytoHubba’s MCC algorithm and validated with datasets GSE6731 and GSE52746. Finally, the miRNA gene regulatory network was constructed by Cytoscape and NetworkAnalyst to predict potential microRNAs (miRNAs) associated with DEGs. Results: A total of 97 DEGs were identified, consisting of 88 downregulated genes and 9 upregulated genes. The enriched functions and pathways of the DEGs include immune system process, response to stress, response to cytokine and extracellular region. KEGG pathway analysis indicates that the genes were significantly enriched in Cytokine-cytokine receptor interaction, IL-17 signaling pathway, Rheumatoid arthritis and TNF signaling pathway. In combination with the results of the protein-protein interaction (PPI) network and CytoHubba, 10 hub genes including IL1B, CXCL8, CXCL10, CXCL1, CXCL2, CXCL5, ICAM1, IL1RN, TIMP1 and MMP3 were selected. Based on the DEG-miRNAs network construction, 5 miRNAs including hsa-mir-21-5p, hsa-mir-93-5p, hsa-mir-98-5p, hsa-mir-1-3p and hsa-mir-335-5p were identified as potential critical miRNAs. Conclusion: In conclusion, a total of 97 DEGs, 10 hub genes and 5 miRNAs that may be involved in the progression or occurrence of CD were identified in this study, which could be regarded as biomarkers of CD.
Collapse
Affiliation(s)
- Yuxin Sun
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Daxing Cai
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Weitao Hu
- Department of Rheumatology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Taiyong Fang
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- *Correspondence: Taiyong Fang,
| |
Collapse
|
17
|
Ahmed O, Farid A, Elamir A. Dual role of melatonin as an anti-colitis and anti-extra intestinal alterations against acetic acid-induced colitis model in rats. Sci Rep 2022; 12:6344. [PMID: 35428860 PMCID: PMC9012815 DOI: 10.1038/s41598-022-10400-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/06/2022] [Indexed: 12/14/2022] Open
Abstract
The available ulcerative colitis drugs exhibit limited outcomes and adverse side effects. Therefore, our study aimed to investigate the therapeutic efficacy of melatonin in acetic acid (AA)-induced colitis to establish a possible treatment for colitis and its impacts on vital organs. Following colitis induction (2 ml 5% AA, rectally), rats were orally received melatonin (5 mg/kg) once per day for 6 days after colitis induction. Then, histopathological examination of colon, kidney, liver, and spleen was conducted, interleukin-1 beta (IL-1β), tumor necrosis factor-alpha (TNF-α), myeloperoxidase (MPO), malondialdehyde (MDA), glutathione (GSH), and total antioxidant capacity (TAC) levels were assessed in colon tissue. Colitis induction in untreated rats caused necrotic effects in colon tissues, a significant increase in colonic IL-1β, TNF-α, MPO, and MDA levels, and a remarkable decrease in GSH and TAC levels in colon tissue in comparison to the control group. Meanwhile, melatonin treatment reversed these parameters by improving the microscopic and macroscopic colitis features and extra-intestinal (kidney, liver, and spleen) changes in all treated rats compared to the colitis control group. These results denote a reduction in colitis severity due to the anti-inflammatory and anti-oxidative effects of melatonin and its positive impact on the vital organs.
Collapse
Affiliation(s)
- Osama Ahmed
- Zoology Department, Faculty of Science, Cairo University, Giza, 12613, Egypt.
| | - Alyaa Farid
- Zoology Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Azza Elamir
- Zoology Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| |
Collapse
|
18
|
Liso M, Verna G, Cavalcanti E, De Santis S, Armentano R, Tafaro A, Lippolis A, Campiglia P, Gasbarrini A, Mastronardi M, Pizarro TT, Cominelli F, Lopetuso LR, Chieppa M. Interleukin 1β Blockade Reduces Intestinal Inflammation in a Murine Model of Tumor Necrosis Factor-Independent Ulcerative Colitis. Cell Mol Gastroenterol Hepatol 2022; 14:151-171. [PMID: 35314399 PMCID: PMC9120241 DOI: 10.1016/j.jcmgh.2022.03.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND & AIMS Inflammatory bowel diseases are multifactorial diseases commonly treated with either immunomodulatory drugs or anti-tumor necrosis factor (TNF). Currently, failure to respond to anti-TNF therapy (assessed no earlier than 8-12 weeks after starting treatment) occurs in 20%-40% of patients enrolled in clinical trials and in 10%-20% in clinical practice. Murine models of inflammatory bowel disease provide important tools to better understand disease mechanism(s). In this context and among the numerous models available, Winnie-TNF-knockout (KO) mice recently were reported to show characteristics of ulcerative colitis (UC) that are independent of TNF, and with increased interleukin (IL)1β production. METHODS Herein, the efficacy of recombinant IL1-receptor antagonist (anakinra) administration was evaluated in Winnie-TNF-KO mice, used as a UC model of primary anti-TNF nonresponders. RESULTS We analyzed gut mucosal biopsy specimens and circulating cytokine profiles of a cohort of 30 UC patients; approximately 75% of primary nonresponders were characterized by abundant IL1β in both the serum and local intestinal tissues. In Winnie-TNF-KO mice, administration of anakinra efficiently reduced the histologic score of the distal colon, which represents the most common site of inflammation in Winnie mice. Furthermore, among lamina propria and mesenteric lymph node-derived T cells, interferon γ-expressing CD8+ T cells were reduced significantly after anakinra administration. CONCLUSIONS Our study provides new insight and alternative approaches to treat UC patients, and points to anti-IL1 strategies (ie, anakinra) that may be a more effective therapeutic option for primary nonresponders to anti-TNF therapy.
Collapse
Affiliation(s)
- Marina Liso
- National Institute of Gastroenterology “S. de Bellis,” Research Hospital, Castellana Grotte (BA), Italy
| | - Giulio Verna
- Department of Pharmacy, University of Salerno, Fisciano (SA), Italy,Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Elisabetta Cavalcanti
- National Institute of Gastroenterology “S. de Bellis,” Research Hospital, Castellana Grotte (BA), Italy
| | - Stefania De Santis
- Department of Pharmacy-Drug Science, University of Bari Aldo Moro, Bari, Italy
| | - Raffaele Armentano
- National Institute of Gastroenterology “S. de Bellis,” Research Hospital, Castellana Grotte (BA), Italy
| | - Angela Tafaro
- National Institute of Gastroenterology “S. de Bellis,” Research Hospital, Castellana Grotte (BA), Italy
| | - Antonio Lippolis
- National Institute of Gastroenterology “S. de Bellis,” Research Hospital, Castellana Grotte (BA), Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Fisciano (SA), Italy
| | - Antonio Gasbarrini
- Digestive Disease Center–Unità Operativa Complessa di Medicina Interna e Gastroenterologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario “A. Gemelli” Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Roma, Italy
| | - Mauro Mastronardi
- National Institute of Gastroenterology “S. de Bellis,” Research Hospital, Castellana Grotte (BA), Italy
| | - Theresa Torres Pizarro
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Fabio Cominelli
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Loris Riccardo Lopetuso
- Digestive Disease Center–Unità Operativa Complessa di Medicina Interna e Gastroenterologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario “A. Gemelli” Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Roma, Italy,Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy,Center for Advanced Studies and Technology, “G. d'Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Marcello Chieppa
- National Institute of Gastroenterology “S. de Bellis,” Research Hospital, Castellana Grotte (BA), Italy,Dietetics and Clinical Nutrition Laboratory, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy,Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, via Monteroni, Lecce, Italy,Correspondence Address correspondence to: Marcello Chieppa, PhD, Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, via Monteroni, 73100 Lecce, Italy.
| |
Collapse
|
19
|
Ahmed O, Abdel-Halim M, Farid A, Elamir A. Taurine loaded chitosan-pectin nanoparticle shows curative effect against acetic acid-induced colitis in rats. Chem Biol Interact 2022; 351:109715. [PMID: 34695389 DOI: 10.1016/j.cbi.2021.109715] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/29/2021] [Accepted: 10/20/2021] [Indexed: 12/22/2022]
Abstract
Owing to the poor outcomes and adverse side effects of existing ulcerative colitis drugs, the study aimed to develop an alternative nano-based treatment approach. The study was designed to characterize the in vitro and in vivo properties of taurine, taurine-loaded chitosan pectin nanoparticles (Tau-CS-PT-NPs) and chitosan pectin nanoparticles (CS-PT-NPs) in the therapy of acetic acid (AA)-induced colitis in rats. CS-PT-NPs and Tau-CS-PT-NPs were prepared by ionic gelation method then in vitro characterized, including transmission electron microscopy (TEM), polydispersity index (PDI), zeta potential, Fourier transform infrared (FTIR) spectroscopy, encapsulation efficiency (EE), and drug release profile. Following colitis induction, rats were orally administrated with free taurine, Tau-CS-PT-NPs, and CS-PT-NPs once per day for six days. The sizes of Tau-CS-PT-NPs and CS-PT-NPs were 74.17 ± 2.88 nm and 42.22 ± 2.41 nm, respectively. EE was about 69.09 ± 1.58%; furthermore, 60% of taurine was released in 4 h in simulated colon content. AA-induced colitis in untreated rats led to necrosis of colon tissues and a significant increase in interleukin-1beta (IL-1β), Tumor Necrosis Factor-alpha (TNF-α), myeloperoxidase (MPO), and malondialdehyde (MDA) levels associated with a remarkable reduction in glutathione (GSH) level in colon tissue in comparison to control group. Treatment with taurine, Tau-CS-PT-NPs, and CS-PT-NPs partly reversed these effects. The present study demonstrated that the administration of free taurine, CS-PT-NPs, and Tau-CS-PT-NPs exerted beneficial effects in acetic acid-induced colitis by their anti-inflammatory and antioxidant activities. The best therapeutic effect was observed in animals treated with taurine-loaded chitosan pectin nanoparticles.
Collapse
Affiliation(s)
- Osama Ahmed
- Zoology Dep., Faculty of Science, Cairo University, Giza, 12613, Egypt.
| | - Mohammad Abdel-Halim
- Pharmaceutical Chemistry Department, Faculty of Pharmacy and Biotechnology, German University, Cairo, 11835, Egypt
| | - Alyaa Farid
- Zoology Dep., Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Azza Elamir
- Zoology Dep., Faculty of Science, Cairo University, Giza, 12613, Egypt
| |
Collapse
|
20
|
Yao H, Shi Y, Yuan J, Sa R, Chen W, Wan X. Matrine protects against DSS-induced murine colitis by improving gut barrier integrity, inhibiting the PPAR-α signaling pathway, and modulating gut microbiota. Int Immunopharmacol 2021; 100:108091. [PMID: 34474274 DOI: 10.1016/j.intimp.2021.108091] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 08/16/2021] [Accepted: 08/22/2021] [Indexed: 12/30/2022]
Abstract
Matrine is a naturally occurring quinolizidine alkaloid with various bioactivities. However, little is known of its function on ulcerative colitis (UC). Here, we investigated the effect and underlying mechanisms of matrine on dextran sulfate sodium (DSS)-induced UC mice. In this study, different concentrations of matrine were given to mice with DSS-induced colitis for a week. The symptoms of colitis, colonic pathology, inflammation-related indicators, and intestinal mucosal barrier function were detected and analyzed. Moreover, RNA-seq analysis in colon tissues was conducted, and 16S rDNA sequencing was carried out to evaluate the gut microbiota of colon contents. The results showed that matrine significantly alleviated clinical activity and histological changes of UC mice, inhibited the production of the pro-inflammatory cytokines, and improved gut barrier integrity. Moreover, RNA-seq analysis and experimental verification showed that matrine significantly inhibited the peroxisome proliferator-activated receptor-α (PPAR-α) signaling pathway. 16S rDNA sequencing revealed that matrine altered the composition and functions of gut microbiota, increased the abundance of Barnesiella intestinihominis and decreased the abundance of Helicobacter ganmani at the species level. In conclusion, matrine ameliorated DSS-induced colitis by improving gut barrier integrity, inhibiting the PPAR-α signaling pathway, and modulating gut microbiota. These suggested that matrine may be a therapeutic agent for UC treatment.
Collapse
Affiliation(s)
- Huixiang Yao
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yan Shi
- Department of GI Endoscopy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Junqing Yuan
- Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Ri Sa
- Department of Nuclear Medicine, the First Hospital of Jilin University, Changchun, China.
| | - Wei Chen
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xinjian Wan
- Department of GI Endoscopy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
21
|
De Santis S, Scarano A, Liso M, Calabrese FM, Verna G, Cavalcanti E, Sila A, Lippolis A, De Angelis M, Santino A, Chieppa M. Polyphenol Enriched Diet Administration During Pregnancy and Lactation Prevents Dysbiosis in Ulcerative Colitis Predisposed Littermates. Front Cell Infect Microbiol 2021; 11:622327. [PMID: 34178715 PMCID: PMC8221423 DOI: 10.3389/fcimb.2021.622327] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
Neonatal colonization of the gastrointestinal tract depends on mother microbiome, thus mother microbiota dysbiosis is transmitted to the offspring during the delivery and shaped by breastmilk characteristics. Here we used a murine model of UC predisposition (Winnie-/-) to evaluate the effects of maternal diet during pregnancy and lactation. Using heterozygous breeders, we obtained both Winnie-/- and C57BL/6 littermates from the same mother and compared their microbiota at weaning and adult age, using a diet enriched with 1% tomato fruit of a line – named Bronze – highly enriched in bioactive polyphenols, or Control tomato. Females received enriched diets two weeks before the beginning of the breeding and never stopped for the following six months. No significant effect was observed in regard to the percentage of Winnie-/- offspring, as with both diets the percentage was about 25% as expected. Winnie littermates from breeders fed with the Bronze-enriched diet showed reduced dysbiosis at 4 weeks of age if compared with Winnie under the Control tomato diet. This effect was then reduced when mice reached adult age. Conversely, the microbiota of C57BL/6 does not change significantly, indicating that fortified mothers-diet significantly contribute to preventing dysbiosis in genetically predisposed offspring, but has mild effects on healthy littermates and adult mice. An overall tendency towards reduced inflammation was underlined by the colon weight and the percentage of Foxp3+ cells reduction in Winnie mice fed with Bronze diet. Control diet did not show similar tendency.
Collapse
Affiliation(s)
- Stefania De Santis
- Department of Pharmacy-Drug Science, University of Bari Aldo Moro, Bari, Italy
| | - Aurelia Scarano
- Institute of Sciences of Food Production National Research Council (CNR), Unit of Lecce, Lecce, Italy
| | - Marina Liso
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Italy
| | | | - Giulio Verna
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | - Elisabetta Cavalcanti
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Italy
| | - Annamaria Sila
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Italy
| | - Antonio Lippolis
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Italy
| | - Maria De Angelis
- Department of Soil, Plant and Food Sciences, University of Bari, Bari, Italy
| | - Angelo Santino
- Institute of Sciences of Food Production National Research Council (CNR), Unit of Lecce, Lecce, Italy
| | - Marcello Chieppa
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Italy
| |
Collapse
|
22
|
Cao F, Cheng YS, Yu L, Xu YY, Wang Y. Bioinformatics Analysis of Differentially Expressed Genes and Protein-Protein Interaction Networks Associated with Functional Pathways in Ulcerative Colitis. Med Sci Monit 2021; 27:e927917. [PMID: 33462173 PMCID: PMC7824989 DOI: 10.12659/msm.927917] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background This bioinformatics study aimed to identify differentially expressed genes (DEGs) and protein–protein interaction (PPI) networks associated with functional pathways in ulcerative colitis based on 3 Gene Expression Omnibus (GEO) datasets. Material/Methods The GSE87466, GSE75214, and GSE48958 MINiML formatted family files were downloaded from the GEO database. DEGs were identified from the 3 datasets, and volcano maps and heat maps were drawn after R language standardization and analysis, respectively. Venn diagram software was used to identify common DEGs. PPI analysis of common DEGs was performed using the Search Tool for the Retrieval of Interacting Genes. Gene modules and hub genes were visualized in the PPI network using Cytoscape. Enrichment analysis was performed for all common DEGs, module genes, and hub genes. Results A total of 90 DEGs were selected, which included 3 functional modules and 1 hub gene module. CXCL8 module genes were mainly enriched in cytokine-mediated signaling pathways and interleukin (IL)-10 signaling. CCL20 module genes were mainly enriched in the IL-17 signaling pathway and cellular response to IL-1. Hub gene modules mainly involved IL-10, IL-4, and IL-13 signaling pathways. CXCL8, CXCL1, and IL-1β were the top 3 hub genes and were mainly involved in IL-10 signaling. Conclusions Bioinformatics analysis using 3 GEO datasets identified CXCL8, CXCL1, and IL-1β, which are involved in IL-10 signaling, as the top 3 hub genes in ulcerative colitis. The findings from this study remain to be validated, but they may contribute to the further understanding of the pathogenesis of ulcerative colitis.
Collapse
Affiliation(s)
- Feng Cao
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, Anhui, China (mainland)
| | - Yun-Sheng Cheng
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, Anhui, China (mainland)
| | - Liang Yu
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, Anhui, China (mainland)
| | - Yan-Yan Xu
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, China (mainland)
| | - Yong Wang
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, Anhui, China (mainland)
| |
Collapse
|
23
|
Jiaweishaoyao Decoction Alleviates DSS-Induced Ulcerative Colitis via Inhibiting Inflammation. Gastroenterol Res Pract 2020; 2020:7182874. [PMID: 32565784 PMCID: PMC7273494 DOI: 10.1155/2020/7182874] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/09/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023] Open
Abstract
Purpose Jiaweishaoyao decoction (JWSYD) is a traditional prescription of Chinese medicine that is initially used for the treatment of diarrhea. This study is aimed at investigating the effects of JWSYD on DSS-induced ulcerative colitis (UC). Methods DSS-induced UC mice and LPS-induced RAW264.7 cells were used as the UC model in vivo and in vitro. UC was assessed by body weight, disease activity index (DAI), colon length, spleen weight, and histopathological score (HE staining). The levels of TNF-α, IL-1β, and IL-6 were analyzed by ELISA and qRT-PCR. The levels of NLRP3 inflammasome- and NF-κB pathway-associated proteins were measured by western blot. Results JWSYD alleviated DSS-induced UC in respect to body weight, DAI, colon length, spleen weight, and histopathological score. JWSYD reduced the levels of TNF-α, IL-1β, and IL-6 in DSS-induced UC mice and the supernatants of LPS-induced RAW264.7 cells. JWSYD suppressed the protein levels of inflammasome-associated proteins, including NLRP3, ASC1, Procaspase-1, Cleaved caspase-1, and Cleaved IL-1β in DSS-induced UC mice and LPS-induced RAW264.7 cells. In addition, JWSYD suppressed the NF-κB pathway in vitro and in vivo. Conclusion JWSYD alleviated DSS-induced UC via inhibiting the NLRP3 inflammasome and NF-κB pathway.
Collapse
|
24
|
De Santis S, Verna G, Serino G, Armentano R, Cavalcanti E, Liso M, Dicarlo M, Coletta S, Mastronardi M, Lippolis A, Tafaro A, Santino A, Pinto A, Campiglia P, Huang AY, Cominelli F, Pizarro TT, Chieppa M. Winnie- APCMin/+ Mice: A Spontaneous Model of Colitis-Associated Colorectal Cancer Combining Genetics and Inflammation. Int J Mol Sci 2020; 21:ijms21082972. [PMID: 32340123 PMCID: PMC7215554 DOI: 10.3390/ijms21082972] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 12/19/2022] Open
Abstract
(1) Background: Colorectal cancer (CRC) is among the best examples of the relationship between inflammation and increased cancer risk. (2) Methods: To examine the effects of spontaneous low-grade chronic inflammation on the pathogenesis of CRC, we developed a new murine model of colitis-associated cancer (CAC) by crossing Mucin 2 mutated mice (Winnie) with ApcMin/+ mice. (3) Results: The resulting Winnie-ApcMin/+ model combines an inflammatory background with a genetic predisposition to small intestinal polyposis. Winnie-ApcMin/+ mice show an early occurrence of inflammatory signs and dysplastic lesions in the distal colon with a specific molecular signature. (4) Conclusion: The Winnie-ApcMin/+ model is a perfect model to demonstrate that chronic inflammation represents a crucial risk factor for the onset and progression of tumoral lesions in individuals genetically predisposed to CRC.
Collapse
Affiliation(s)
- Stefania De Santis
- Department of Pharmacy-Drug Science, University of Bari Aldo Moro, 70126 Bari, Italy
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy; (A.P.); (P.C.)
- Correspondence: (S.D.S.); (M.C.); Tel.: +39-080-544-2763 (S.D.S.); +39-080-499-4628 (M.C.)
| | - Giulio Verna
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
| | - Grazia Serino
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
| | - Raffaele Armentano
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
| | - Elisabetta Cavalcanti
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
| | - Marina Liso
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
| | - Manuela Dicarlo
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
| | - Sergio Coletta
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
| | - Mauro Mastronardi
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
| | - Antonio Lippolis
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
| | - Angela Tafaro
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
| | - Angelo Santino
- Institute of Sciences of Food Production C.N.R., Unit of Lecce, 73100 Lecce, Italy;
| | - Aldo Pinto
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy; (A.P.); (P.C.)
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy; (A.P.); (P.C.)
| | - Alex Y. Huang
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Angie Fowler AYA Cancer Institute, Cleveland, OH 44106, USA;
| | - Fabio Cominelli
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (F.C.); (T.T.P.)
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Theresa T. Pizarro
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (F.C.); (T.T.P.)
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Marcello Chieppa
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
- European Biomedical Research Institute of Salerno (EBRIS), 84125 Salerno, Italy
- Correspondence: (S.D.S.); (M.C.); Tel.: +39-080-544-2763 (S.D.S.); +39-080-499-4628 (M.C.)
| |
Collapse
|
25
|
Liso M, De Santis S, Verna G, Dicarlo M, Calasso M, Santino A, Gigante I, Eri R, Raveenthiraraj S, Sobolewski A, Palmitessa V, Lippolis A, Mastronardi M, Armentano R, Serino G, De Angelis M, Chieppa M. A Specific Mutation in Muc2 Determines Early Dysbiosis in Colitis-Prone Winnie Mice. Inflamm Bowel Dis 2020; 26:546-556. [PMID: 31748792 PMCID: PMC7054774 DOI: 10.1093/ibd/izz279] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Inflammatory bowel disease (IBD), including Crohn disease (CD) and ulcerative colitis (UC), is a multifactorial disorder characterized by chronic inflammation and altered gut barrier function. Dysbiosis, a condition defined by dysregulation of the gut microbiome, has been reported in patients with IBD and in experimental models of colitis. Although several factors have been implicated in directly affecting gut microbial composition, the genetic determinants impacting intestinal dysbiosis in IBD remain relatively unknown. METHODS We compared the microbiome of normal, uninflamed wild-type (WT) mice with that of a murine model of UC (ie, Winnie strain). Winnie mice possess a missense mutation in Muc2 that manifests in altered mucus production as early as 4 weeks of age, with ensuing colonic inflammation. To better address the potential role of mutant Muc2 in promoting dysbiosis in Winnie mice, we evaluated homozygous mutant mice (Winnie-/-) with their WT littermates that, after weaning from common mothers, were caged separately according to genotype. Histologic and inflammatory status were assessed over time, along with changes in their respective microbiome compositions. RESULTS Dysbiosis in Winnie mice was already established at 4 weeks of age, before histologic evidence of gut inflammatory changes, in which microbial communities diverged from that derived from their mothers. Furthermore, dysbiosis persisted until 12 weeks of age, with peak differences in microbiome composition observed between Winnie and WT mice at 8 weeks of age. The relative abundance of Bacteroidetes was greater in Winnie compared with WT mice. Verrucomicrobia was detected at the highest relative levels in 4-week-old Winnie mice; in particular, Akkermansia muciniphila was among the most abundant species found at 4 weeks of age. CONCLUSIONS Our results demonstrate that mutant genetic determinants involved in the complex regulation of intestinal homeostasis, such as that observed in Winnie mice, are able to promote early gut dysbiosis that is independent from maternal microbial transfer, including breastfeeding. Our data provide evidence for intestinal dysbiosis attributed to a Muc2-driven mucus defect that leads to colonic inflammation and may represent an important target for the design of future interventional studies.
Collapse
Affiliation(s)
- Marina Liso
- National Institute of Gastroenterology “S. de Bellis,” Institute of Research, Castellana Grotte (BA), Italy
| | - Stefania De Santis
- Department of Pharmacy, School of Pharmacy, University of Salerno, Fisciano (SA), Italy
| | - Giulio Verna
- National Institute of Gastroenterology “S. de Bellis,” Institute of Research, Castellana Grotte (BA), Italy
| | - Manuela Dicarlo
- National Institute of Gastroenterology “S. de Bellis,” Institute of Research, Castellana Grotte (BA), Italy
| | - Maria Calasso
- Department of Soil, Plant and Food Sciences, University of Bari, Bari, Italy
| | - Angelo Santino
- Institute of Sciences of Food Production C.N.R., Unit of Lecce, Lecce, Italy
| | - Isabella Gigante
- National Institute of Gastroenterology “S. de Bellis,” Institute of Research, Castellana Grotte (BA), Italy
| | - Rajaraman Eri
- Mucosal Biology, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | | | - Anastasia Sobolewski
- School of Pharmacy University of East Anglia, Norwich Research Park, Norwich, UK
| | - Valeria Palmitessa
- National Institute of Gastroenterology “S. de Bellis,” Institute of Research, Castellana Grotte (BA), Italy
| | - Antonio Lippolis
- National Institute of Gastroenterology “S. de Bellis,” Institute of Research, Castellana Grotte (BA), Italy
| | - Mauro Mastronardi
- National Institute of Gastroenterology “S. de Bellis,” Institute of Research, Castellana Grotte (BA), Italy
| | - Raffaele Armentano
- National Institute of Gastroenterology “S. de Bellis,” Institute of Research, Castellana Grotte (BA), Italy
| | - Grazia Serino
- National Institute of Gastroenterology “S. de Bellis,” Institute of Research, Castellana Grotte (BA), Italy
| | - Maria De Angelis
- Department of Soil, Plant and Food Sciences, University of Bari, Bari, Italy
| | - Marcello Chieppa
- National Institute of Gastroenterology “S. de Bellis,” Institute of Research, Castellana Grotte (BA), Italy,Department of Immunology and Cell Biology, European Biomedical ResearchInstitute of Salerno (EBRIS), Salerno, Italy,Address correspondence to: Marcello Chieppa, PhD, National Institute of Gastroenterology “S. de Bellis,” Institute of Research, Castellana Grotte (BA), Italy ()
| |
Collapse
|
26
|
Mastronardi M, Curlo M, Cavalcanti E, Burattini O, Cuppone R, Tauro R, De Santis S, Serino G, Pesole PL, Stasi E, Caruso ML, Donghia R, Guerra V, Giorgio P, Chieppa M. Administration Timing Is the Best Clinical Outcome Predictor for Adalimumab Administration in Crohn's Disease. Front Med (Lausanne) 2019; 6:234. [PMID: 31737635 PMCID: PMC6838026 DOI: 10.3389/fmed.2019.00234] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 10/03/2019] [Indexed: 01/01/2023] Open
Abstract
Biological intervention for Crohn's Disease (CDs) patients, mainly using anti-TNF antibodies, is often an efficient therapeutic solution. Nonetheless, data defining the administration timing to maximize the chances of clinical remission are lacking. The objective of this “real-life” retrospective study was to evaluate if early Adalimumab (ADA) administration (<12 months) was an efficient strategy to improve patients' clinical outcome. This single center study included 157 CD patients, of which 80 received the first ADA administration within the first 12 months from the diagnosis. After 1 year of therapy, clinical remission was observed in 50.32% of patients, mucosal healing in 37.58%. Clinical remission was observed in 66.25% of the early ADA administration patients vs. 33.77% of the late (>12 months) (p < 0.001); mucosal healing was observed in 53.75% of the early vs. 20.78% of the late (p < 0.001). Dose escalation was required for 30.00% of the early vs. 66.23% of the late (<0.01). In the early ADA administration group, 7.50% patients were considered non-responders at the end of the follow-up vs. 22.08% patients in the late administration group. These findings highlighted that early ADA administration (within 1 year of diagnosis) improves the clinical response and mucosal healing, and reduces the loss of response rate and need for dose escalation.
Collapse
Affiliation(s)
- Mauro Mastronardi
- Department of Research, National Institute of Gastroenterology "S. de Bellis", Bari, Italy
| | - Margherita Curlo
- Department of Research, National Institute of Gastroenterology "S. de Bellis", Bari, Italy
| | - Elisabetta Cavalcanti
- Department of Research, National Institute of Gastroenterology "S. de Bellis", Bari, Italy
| | - Osvaldo Burattini
- Department of Research, National Institute of Gastroenterology "S. de Bellis", Bari, Italy
| | - Renato Cuppone
- Department of Research, National Institute of Gastroenterology "S. de Bellis", Bari, Italy
| | - Romina Tauro
- Department of Research, National Institute of Gastroenterology "S. de Bellis", Bari, Italy
| | - Stefania De Santis
- Department of Research, National Institute of Gastroenterology "S. de Bellis", Bari, Italy.,Department of Pharmacy, Faculty of Pharmacy and Medicine, University of Salerno, Fisciano, Italy
| | - Grazia Serino
- Department of Research, National Institute of Gastroenterology "S. de Bellis", Bari, Italy
| | - Pasqua Letizia Pesole
- Department of Research, National Institute of Gastroenterology "S. de Bellis", Bari, Italy
| | - Elisa Stasi
- Department of Research, National Institute of Gastroenterology "S. de Bellis", Bari, Italy
| | - Maria Lucia Caruso
- Department of Research, National Institute of Gastroenterology "S. de Bellis", Bari, Italy
| | - Rossella Donghia
- Department of Research, National Institute of Gastroenterology "S. de Bellis", Bari, Italy
| | - Vito Guerra
- Department of Research, National Institute of Gastroenterology "S. de Bellis", Bari, Italy
| | - Pietro Giorgio
- Department of Research, National Institute of Gastroenterology "S. de Bellis", Bari, Italy
| | - Marcello Chieppa
- Department of Research, National Institute of Gastroenterology "S. de Bellis", Bari, Italy
| |
Collapse
|
27
|
Geesala R, Issuree PD, Maretzky T. Novel functions of inactive rhomboid proteins in immunity and disease. J Leukoc Biol 2019; 106:823-835. [PMID: 31369701 DOI: 10.1002/jlb.3vmr0219-069r] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/03/2019] [Accepted: 06/20/2019] [Indexed: 12/11/2022] Open
Abstract
iRhoms are related to a family of intramembrane serine proteinases called rhomboids but lack proteolytic activity. In mammals, there are two iRhoms, iRhom1 and iRhom2, which have similar domain structures and overlapping specificities as well as distinctive functions. These catalytically inactive rhomboids are essential regulators for the maturation and trafficking of the disintegrin metalloprotease ADAM17 from the endoplasmic reticulum to the cell surface, and are required for the cleavage and release of a variety of membrane-associated proteins, including the IL-6 receptor, l-selectin, TNF, and EGFR ligands. iRhom2-dependent regulation of ADAM17 function has been recently implicated in the development and progression of several autoimmune diseases including rheumatoid arthritis, lupus nephritis, as well as hemophilic arthropathy. In this review, we discuss our current understanding of iRhom biology, their implications in autoimmune pathologies, and their potential as therapeutic targets.
Collapse
Affiliation(s)
- Ramasatyaveni Geesala
- Inflammation Program, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa, USA.,Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa, USA
| | - Priya D Issuree
- Inflammation Program, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa, USA.,Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa, USA
| | - Thorsten Maretzky
- Inflammation Program, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa, USA.,Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa, USA.,Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
28
|
Desalegn G, Pabst O. Inflammation triggers immediate rather than progressive changes in monocyte differentiation in the small intestine. Nat Commun 2019; 10:3229. [PMID: 31324779 PMCID: PMC6642215 DOI: 10.1038/s41467-019-11148-2] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 06/17/2019] [Indexed: 12/14/2022] Open
Abstract
Bone marrow-derived circulating monocytes contribute to the replenishment and maintenance of the intestinal macrophage population. Intestinal monocytes undergo context-dependent phenotypic and functional adaptations to either maintain local immune balance or support intestinal inflammation. Here we use monocyte adoptive transfer to dissect the dynamics of monocyte-to-macrophage differentiation in normal and inflamed small intestine. We find that during homeostasis CCR2 and β7-integrin mediate constitutive homing of monocytes to the gut. By contrast, intestinal inflammation increases monocyte recruitment via CCR2, but not β7-integrin. In the non-inflamed intestine, monocytes gradually differentiate to express genes typically associated with tolerogenic macrophage functions. Conversely, immediately upon entry into the inflamed intestine, monocytes adapt a different expression pattern in a partly Trem-1-dependent manner. Our observations suggest that inflammation fundamentally changes the kinetics and modalities of monocyte differentiation in tissues. Bone marrow-derived monocytes are recruited to the gut to replenish the local macrophage pool. Here the authors show that, while such replenishment constitutively occur under homeostasis, gut inflammation induces an immediate, Trem1-related transcription change to recruited monocyte to enable a context-dependent modulation of macrophage functions.
Collapse
Affiliation(s)
- Girmay Desalegn
- Institute of Molecular Medicine, RWTH Aachen University, D-52074, Aachen, Germany.,Institute of Immunology, Hannover Medical School, D-30625, Hannover, Germany
| | - Oliver Pabst
- Institute of Molecular Medicine, RWTH Aachen University, D-52074, Aachen, Germany.
| |
Collapse
|
29
|
Zhang D, Ren YB, Wei K, Hong J, Yang YT, Wu LJ, Zhang J, Shi Z, Wu HG, Ma XP. Herb-partitioned moxibustion alleviates colon injuries in ulcerative colitis rats. World J Gastroenterol 2018; 24:3384-3397. [PMID: 30122878 PMCID: PMC6092579 DOI: 10.3748/wjg.v24.i30.3384] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 06/22/2018] [Accepted: 06/30/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To observe the effect of herb-partitioned moxibustion (HPM) on expression of colonic cytokines in ulcerative colitis (UC) rats. METHODS A UC rat model was established by protein immunization in combination with topical chemical stimulation. Rats in the HPM group (n = 8) received HPM at bilateral Tianshu (ST25) points. The gross injury and pathological scores of the colon were recorded. The expression profile of colonic cytokines was assayed using the protein microarray technique. Specific differential cytokines were selected and verified by ELISA. The corresponding UniProt Accessions of the differentially expressed cytokines were retrieved in the UniProt database. The pathways involved were analyzed with the help of the KEGG PATHWAY database. The DAVID database was used for functional cluster and pathway analysis. RESULTS HPM improved colon injuries in UC rats, manifested by accelerated repair of ulcers and alleviation of inflammation, and the gross injury and pathological scores both significantly decreased (P < 0.01). Fold change > 1.3 or < 0.77 was taken as the screening standard. There were 77 down-regulated and 9 up-regulated differentially expressed colonic cytokines in the HPM group compared with the model group, and expression of 20 differed significantly (P < 0.05). Twelve of the 20 significantly differentially expressed cytokines [β-catenin, interleukin-1 receptor 6 (IL-1R6), IL-1β, B7-1, nerve growth factor receptor, AMP-activated protein kinase-α1, neuropilin-2, orexin A, adipocyte differentiation-related protein, IL-2, Fas and FasL] were up-regulated in the model group (n = 3, compared with the normal group) but down-regulated in the HPM group (n = 3, compared with the model group). Functional cluster analysis showed that the differentially expressed colonic cytokines in the HPM group regulated apoptosis and protein phosphorylation. KEGG pathway analysis showed that 52 down-regulated and 7 up-regulated differentially expressed colonic cytokines in the HPM group had pathways. The pathways that interacted between the cytokines and their receptors accounted for the largest proportion (28 of the down-regulated and 5 of the up-regulated cytokines). CONCLUSION HPM promotes the repair of colon injuries in UC rats, which is related to the regulation of several abnormally expressed cytokines.
Collapse
Affiliation(s)
- Dan Zhang
- Laboratory of Acupuncture-moxibustion and Immunology, Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Yan-Bo Ren
- Department of Integrated Traditional Chinese Medicine and Western Medicine, North Branch of Huashan Hospital, Fudan University, Shanghai 201907, China
| | - Kai Wei
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jue Hong
- Laboratory of Acupuncture-moxibustion and Immunology, Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Yan-Ting Yang
- Yueyang Clinical Medicine School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Li-Jie Wu
- Yueyang Clinical Medicine School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ji Zhang
- Yueyang Clinical Medicine School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zheng Shi
- Laboratory of Acupuncture-moxibustion and Immunology, Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Huan-Gan Wu
- Laboratory of Acupuncture-moxibustion and Immunology, Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Xiao-Peng Ma
- Laboratory of Acupuncture-moxibustion and Immunology, Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
- Yueyang Clinical Medicine School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
30
|
Lambertucci F, Arboatti A, Sedlmeier MG, Motiño O, Alvarez MDL, Ceballos MP, Villar SR, Roggero E, Monti JA, Pisani G, Quiroga AD, Martín-Sanz P, Carnovale CE, Francés DE, Ronco MT. Disruption of tumor necrosis factor alpha receptor 1 signaling accelerates NAFLD progression in mice upon a high-fat diet. J Nutr Biochem 2018; 58:17-27. [PMID: 29860102 DOI: 10.1016/j.jnutbio.2018.04.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/12/2018] [Accepted: 04/25/2018] [Indexed: 02/07/2023]
Abstract
Obesity is accompanied by a low-grade inflammation state, characterized by increased proinflammatory cytokines levels such as tumor necrosis factor alpha (TNFα) and interleukin-1 beta (IL-1β). In this regard, there exists a lack of studies in hepatic tissue about the role of TNFα receptor 1 (TNFR1) in the context of obesity and insulin resistance during the progression of nonalcoholic fatty liver disease (NAFLD). The aim of this work was to evaluate the effects of high-caloric feeding (HFD) (40% fat, for 16 weeks) on liver inflammation-induced apoptosis, insulin resistance, hepatic lipid accumulation and its progression toward nonalcoholic steatohepatitis (NASH) in TNFR1 knock-out and wild-type mice. Mechanisms involved in HFD-derived IL-1β release and impairment of insulin signaling are still unknown, so we determined whether IL-1β affects liver insulin sensitivity and apoptosis through TNFα receptor 1 (TNFR1)-dependent pathways. We showed that knocking out TNFR1 induces an enhanced IL-1β plasmatic release upon HFD feed. This was correlated with higher hepatic and epididymal white adipose tissue mRNA levels. In vivo and in vitro assays confirmed an impairment in hepatic insulin signaling, in part due to IL-1β-induced decrease of AKT activation and diminution of IRS1 levels, followed by an increase in inflammation, macrophage (resident and recruited) accumulation, hepatocyte apoptotic process and finally hepatic damage. In addition, TNFR1 KO mice displayed higher levels of pro-fibrogenic markers. TNFR1 signaling disruption upon an HFD leads to an accelerated progression from simple steatosis to a more severe phenotype with many NASH features, pointing out a key role of TNFR1 in NAFLD progression.
Collapse
Affiliation(s)
- Flavia Lambertucci
- Instituto de Fisiología Experimental (IFISE-CONICET), Suipacha 570, 2000 Rosario, Argentina
| | - Ainelén Arboatti
- Instituto de Fisiología Experimental (IFISE-CONICET), Suipacha 570, 2000 Rosario, Argentina
| | | | - Omar Motiño
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain
| | - María de Luján Alvarez
- Instituto de Fisiología Experimental (IFISE-CONICET), Suipacha 570, 2000 Rosario, Argentina
| | - María Paula Ceballos
- Instituto de Fisiología Experimental (IFISE-CONICET), Suipacha 570, 2000 Rosario, Argentina
| | - Silvina R Villar
- Instituto de Inmunología, Facultad de Ciencias Médicas, UNR, Suipacha 531, 2000 Rosario, Argentina
| | - Eduardo Roggero
- Instituto de Inmunología, Facultad de Ciencias Médicas, UNR, Suipacha 531, 2000 Rosario, Argentina
| | - Juan A Monti
- Instituto de Fisiología Experimental (IFISE-CONICET), Suipacha 570, 2000 Rosario, Argentina
| | - Gerardo Pisani
- Cátedra de Morfología, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Ariel D Quiroga
- Instituto de Fisiología Experimental (IFISE-CONICET), Suipacha 570, 2000 Rosario, Argentina
| | - Paloma Martín-Sanz
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
| | | | - Daniel Eleazar Francés
- Instituto de Fisiología Experimental (IFISE-CONICET), Suipacha 570, 2000 Rosario, Argentina
| | - María Teresa Ronco
- Instituto de Fisiología Experimental (IFISE-CONICET), Suipacha 570, 2000 Rosario, Argentina.
| |
Collapse
|