1
|
Li Z, Guo H, He H, Wang S, Pei S, Xie L. The relationship between smoking cessation history and significant liver fibrosis among the population with metabolic dysfunction-associated steatotic liver disease in the United States. PLoS One 2025; 20:e0320573. [PMID: 40168280 PMCID: PMC11960941 DOI: 10.1371/journal.pone.0320573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/19/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Smoking was identified as a risk factor for the development of liver fibrosis in patients with metabolic dysfunction-associated steatotic liver disease (MASLD). However, the association between smoking cessation history and the development of liver fibrosis remains unclear. This study was intended to analyze the association between smoking cessation history and significant liver fibrosis in adult MASLD participants in the United States. METHODS This study utilized data from 2643 patients with MASLD from the National Health and Nutrition Examination Survey (NHANES). Significant liver fibrosis was detected based on transient elastography measurements. According to the smoking questionnaire data, patients were categorized as non-smokers, ex-smokers and current smokers. A multivariate logistic regression analysis, adjusted for weights, was performed to investigate the relationship between smoking cessation history and the presence of significant liver fibrosis in participants with MASLD. RESULTS A total of 2643 patients with MASLD were included in this study. Compared with non-smokers, ex-smokers had a slightly elevated risk of developing significant liver fibrosis (OR: 1.07, 95% CI: 1.02-1.13). Specifically, a positive correlation was observed between patients who quit smoking for < 20 years and significant liver fibrosis (OR: 1.07, 95% CI: 1.01-1.15). Furthermore, MASLD patients who started regularly smoking at an age of ≤ 20 years (OR: 1.09, 95% CI: 1.02-1.16) and had a smoking duration of ≥ 10 years before quitting (OR: 1.10, 95% CI: 1.02-1.18) were also highly correlated with an increased likelihood of developing significant liver fibrosis. CONCLUSIONS This study revealed that individuals with MASLD who have ceased smoking exhibit an elevated risk for significant liver fibrosis when compared to those who never smoked. It is highly emphasized that MASLD patients who quit smoking for < 20 years, started regularly smoking at an age of ≤ 20 years, and had a smoking duration of ≥ 10 years before quitting should be extremely vigilant regarding the risk of significant liver fibrosis.
Collapse
Affiliation(s)
- Zhongtao Li
- Department of General Surgery (Wenhua Road Campus), The Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Institute of Hepatobiliary Pancreatic and Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Hao Guo
- Department of General Surgery (Wenhua Road Campus), The Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Institute of Hepatobiliary Pancreatic and Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Hongyu He
- Department of General Surgery (Wenhua Road Campus), The Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Institute of Hepatobiliary Pancreatic and Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Shu Wang
- Department of Urology Surgery, The Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Shufen Pei
- Department of Otolaryngology-Head and Neck Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Liang Xie
- Department of General Surgery (Wenhua Road Campus), The Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Institute of Hepatobiliary Pancreatic and Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| |
Collapse
|
2
|
Otake S, Chubachi S, Miyamoto J, Haneishi Y, Arai T, Iizuka H, Shimada T, Sakurai K, Okuzumi S, Kabata H, Asakura T, Miyata J, Irie J, Asano K, Nakamura H, Kimura I, Fukunaga K. Impact of smoking on gut microbiota and short-chain fatty acids in human and mice: Implications for COPD. Mucosal Immunol 2025; 18:353-365. [PMID: 39675727 DOI: 10.1016/j.mucimm.2024.12.006] [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: 07/10/2024] [Revised: 12/02/2024] [Accepted: 12/08/2024] [Indexed: 12/17/2024]
Abstract
We aimed to elucidate the dynamic changes in short-chain fatty acids (SCFA) produced by the gut microbiota following smoking exposure and their role in chronic obstructive pulmonary disease (COPD) pathogenesis. SCFA concentrations were measured in human plasma, comparing non-smokers (n = 6) and smokers (n = 12). Using a mouse COPD model induced by cigarette smoke exposure or elastase-induced emphysema, we modulated SCFA levels through dietary interventions and antibiotics to evaluate their effects on inflammation and alveolar destruction. Human smokers showed lower plasma SCFA concentrations than non-smokers, with plasma propionic acid positively correlating with forced expiratory volume in 1 s/forced vital capacity. Three-month smoking-exposed mice demonstrated altered gut microbiota and significantly reduced fecal SCFA concentrations compared to air-exposed controls. In these mice, a high-fiber diet increased fecal SCFAs and mitigated inflammation and alveolar destruction, while antibiotics decreased fecal SCFAs and exacerbated disease features. However, in the elastase-induced model, fecal SCFA concentration remained unchanged, and high-fiber diet or antibiotic interventions had no significant effect. These findings suggest that smoking exposure alters gut microbiota and SCFA production through its systemic effects. The anti-inflammatory properties of SCFAs may play a role in COPD pathogenesis, highlighting their potential as therapeutic targets.
Collapse
Affiliation(s)
- Shiro Otake
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Shinjuku-ku, 160-8582 Tokyo, Japan
| | - Shotaro Chubachi
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Shinjuku-ku, 160-8582 Tokyo, Japan.
| | - Junki Miyamoto
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, 183-8509 Tokyo, Japan.
| | - Yuri Haneishi
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, 183-8509 Tokyo, Japan
| | - Tetsuya Arai
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Shinjuku-ku, 160-8582 Tokyo, Japan
| | - Hideto Iizuka
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Shinjuku-ku, 160-8582 Tokyo, Japan
| | - Takashi Shimada
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Shinjuku-ku, 160-8582 Tokyo, Japan
| | - Kaori Sakurai
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Shinjuku-ku, 160-8582 Tokyo, Japan
| | - Shinichi Okuzumi
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, 183-8509 Tokyo, Japan
| | - Hiroki Kabata
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Shinjuku-ku, 160-8582 Tokyo, Japan.
| | - Takanori Asakura
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Shinjuku-ku, 160-8582 Tokyo, Japan; Department of Clinical Medicine (Laboratory of Bioregulatory Medicine), Kitasato University School of Pharmacy, 108-8641 Tokyo, Japan; Department of Respiratory Medicine, Kitasato University Kitasato Institute Hospital, 108-8642 Tokyo, Japan
| | - Jun Miyata
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Shinjuku-ku, 160-8582 Tokyo, Japan
| | - Junichiro Irie
- Department of Endocrinology, Metabolism and Nephrology, Keio University School of Medicine, Shinjuku-ku, 160-8582 Tokyo, Japan; Division of Diabetes, Department of Medicine 2, Kansai Medical University, 573-1191 Osaka, Japan
| | - Koichiro Asano
- Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, 259-1193 Kanagawa, Japan
| | - Hidetoshi Nakamura
- Department of Respiratory Medicine, Saitama Medical University, 350-0495 Saitama, Japan
| | - Ikuo Kimura
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, 606-8507 Kyoto, Japan
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Shinjuku-ku, 160-8582 Tokyo, Japan
| |
Collapse
|
3
|
Tian Y, Cheng J, Yang Y, Wang H, Fu Y, Li X, Wang W, Ma S, Xu X, Lu F, Feng P, Han S, Chen H, Hou H, Hu Q, Wu C. A 90-Day Subchronic Exposure to Heated Tobacco Product Aerosol Caused Differences in Intestinal Inflammation and Microbiome Dysregulation in Rats. Nicotine Tob Res 2025; 27:438-446. [PMID: 39028556 DOI: 10.1093/ntr/ntae179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 06/17/2024] [Accepted: 07/14/2024] [Indexed: 07/20/2024]
Abstract
INTRODUCTION Smoking is one of the most important predisposing factors of intestinal inflammatory diseases. Heated tobacco product (HTP) is a novel tobacco category that is claimed to deliver reduced chemicals to humans those reported in combustible cigarette smoke (CS). However, the effect of HTP on the intestine is still unknown. AIMS AND METHODS Our study aims to explore the potential effects of HTP on intestine. In the framework of Organization for Economic Co-operation and Development guidelines 413 guidelines, Sprague-Dawley rats were exposed to HTP aerosol and CS for 13 weeks. The atmosphere was characterized and oxidative stress and inflammation of the intestine were investigated after exposure. Furthermore, the feces we performed with 16S sequencing and metabolomics analysis. RESULTS HTP aerosol and CS led to obvious intestinal damage evidenced by increased intestinal proinflammatory cytokines and oxidative stress in male and female rats After HTP and CS exposure, the abundance that obviously changed were Lactobacillus and Turiciacter in male rats and Lactobacillus and Prevotella in female rats. HTP mainly induces the metabolism of amino acids and fatty acyls such as short-chain fatty acids and tryptophan, while CS is involved in the main metabolism of bile acids, especially indole and derivatives. Although different metabolic pathways in the gut are mediated by HTP and CS, both inflammation and oxidative stress were ultimately induced. CONCLUSIONS HTP aerosol and CS-induced intestinal damage are mediated by different gut microbiota and metabolites, while both lead to inflammation and oxidative stress. IMPLICATIONS The concentration of various harmful components in heated tobacco product aerosol is reported lower than that of traditional cigarette smoke, however, its health risk impact on consumers remains to be studied. Our research findings indicate that heated tobacco products and cigarette smoke inhalation induced intestinal damage through different metabolic pathways mediated by the gut microbiome, indicating the health risk of heated tobacco products in the intestine.
Collapse
Affiliation(s)
- Yushan Tian
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Jiale Cheng
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanan Yang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hongjuan Wang
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Yaning Fu
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Xianmei Li
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Wenming Wang
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Shuhao Ma
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Xiaoxiao Xu
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Fengjun Lu
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Pengxia Feng
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Shulei Han
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Huan Chen
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Hongwei Hou
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Qingyuan Hu
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Chongming Wu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
4
|
Fan J, Zeng F, Zhong H, Cai J, Shen W, Cheng C, He C, Liu Y, Zhou Y, Chen S, Zhu Y, Liu T, Zheng JS, Wang L, Chen YM, Ma W, Zhou D. Potential roles of cigarette smoking on gut microbiota profile among Chinese men. BMC Med 2025; 23:25. [PMID: 39838369 PMCID: PMC11753143 DOI: 10.1186/s12916-025-03852-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Cigarette smoking is posited as a potential factor in disrupting the balance of the human gut microbiota. However, existing studies with limited sample size have yielded inconclusive results. METHODS Here, we assessed the association between cigarette smoking and gut microbial profile among Chinese males from four independent studies (N total = 3308). Both 16S rRNA and shotgun metagenomic sequencing methods were employed, covering 206 genera and 237 species. Microbial diversity and abundance were compared among non-smokers, current smokers, and former smokers. RESULTS Actinomyces[g], Atopobium[g], Haemophilus[g], Turicibacter[g], and Lachnospira[g] were found to be associated with smoking status (current smokers vs. non-smokers). Metagenomic data provided a higher resolution at the species level, particularly for the Actinomyces[g] branch. Additionally, serum γ-glutamylcysteine (γ-Glu-Cys) was found to have a potential role in connecting smoking and Actinomyces[g]. Furthermore, we revealed putative mediation roles of the gut microbiome in the associations between smoking and common diseases including cholecystitis and type 2 diabetes. CONCLUSIONS We characterized the gut microbiota profile in male smokers and further revealed their potential involvement in mediating the impact of smoking on health outcomes. These findings advance our understanding of the intricate association between cigarette smoking and the gut microbiome.
Collapse
Affiliation(s)
- Jiayao Fan
- The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, 388 Yuhangtang Road, Hangzhou, 310058, Zhejiang, China
| | - Fangfang Zeng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Haili Zhong
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jun Cai
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Wentao Shen
- Department of Gastroenterology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chunxiao Cheng
- The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, 388 Yuhangtang Road, Hangzhou, 310058, Zhejiang, China
| | - Chunfeng He
- The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, 388 Yuhangtang Road, Hangzhou, 310058, Zhejiang, China
| | - Yuanjiao Liu
- Department of Epidemiology & Biostatistics, School of Public Health, Zhejiang University, 388 Yuhangtang Road, Hangzhou, 310058, Zhejiang, China
| | - Yuan Zhou
- The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, 388 Yuhangtang Road, Hangzhou, 310058, Zhejiang, China
| | - Shujie Chen
- Department of Gastroenterology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yimin Zhu
- Department of Epidemiology & Biostatistics, School of Public Health, Zhejiang University, 388 Yuhangtang Road, Hangzhou, 310058, Zhejiang, China
| | - Tao Liu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Ju-Sheng Zheng
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Lan Wang
- Department of Gastroenterology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang, China.
- Institute of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Yu-Ming Chen
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| | - Wenjun Ma
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China.
| | - Dan Zhou
- The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, 388 Yuhangtang Road, Hangzhou, 310058, Zhejiang, China.
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
5
|
Huang J, Li J, Geng Z, Yin L, Niu M, Li Q, Liu X, Cheng X, Zhang X, Song X, Wang Y, Wang L, Zuo L, Hu J. Cynaroside ameliorates TNBS-induced colitis by inhibiting intestinal epithelial cell apoptosis via the PI3K/AKT signalling pathway. Front Pharmacol 2025; 15:1496068. [PMID: 39902073 PMCID: PMC11788346 DOI: 10.3389/fphar.2024.1496068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/30/2024] [Indexed: 02/05/2025] Open
Abstract
Background and aims Patients with Crohn's disease (CD) exhibit excessive apoptosis of intestinal epithelial cells (IECs), which contributes to damage to the intestinal barrier structure and function, thereby playing a role in the progression of colitis. Preventing IEC apoptosis and protecting the intestinal barrier are critical to alleviating colitis. Natural plant monomers have been reported to possess multiple pharmacological properties, particularly with the potential to treat CD. This study focuses on Cynaroside (Cyn) to explore its effect on IEC apoptosis and evaluate its pharmacological impact on the intestinal barrier and colitis. Methods The 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced CD-like colitis mice model was employed in this study. We assessed the therapeutic effect of Cyn on CD-like colitis by evaluating the disease activity index (DAI), body weight changes, intestinal tissue pathological damage, and inflammatory factor levels. Immunofluorescence and Western blotting were used to detect the expression and localization of tight junction (TJ) proteins, allowing us to analyze the intestinal barrier structure. The function of the intestinal barrier was examined using FITC-dextran (FD4), TEER values, and bacterial translocation. Network pharmacology enrichment analysis revealed that Cyn could inhibit cell apoptosis. We also explored the effect and underlying mechanism of Cyn in inhibiting IEC apoptosis on intestinal barrier function and colitis using both the TNF-α-induced colonic organoid model and the TNBS-induced mouse model. Results Our findings show that Cyn significantly alleviates TNBS-induced colitis symptoms in mice, as evidenced by reduced body weight loss, colon shortening, DAI score, colon histopathology score, and lower levels of inflammatory factors (IL-1β, TNF-α, and IL-6) compared to the model group. Additionally, the Cyn intervention group showed significant improvements in both the intestinal barrier structure (elevated tight junction protein levels and proper localization) and function (reduced serum FD4 levels, increased intestinal TEER, and decreased bacterial translocation rates in mesenteric lymph nodes [MLNs] and livers). Combining network pharmacology prediction analysis with our validation data from animal models and colonic organoids, we demonstrated that Cyn significantly inhibits IEC apoptosis, as indicated by a decrease in the proportion of TUNEL-positive cells and changes in apoptosis-related protein levels. KEGG enrichment analysis and signaling pathway intervention experiments confirmed that Cyn inhibits the activation of PI3K/AKT signaling. Conclusion Cyn inhibits IEC apoptosis by blocking the PI3K/AKT signaling pathway, which is the primary mechanism underlying its protective effects on the intestinal barrier and its ability to improve CD-like colitis. This study also supports the potential of the Chinese medicine monomer Cyn as a promising therapeutic agent for the treatment of CD.
Collapse
Affiliation(s)
- Ju Huang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Jing Li
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Zhijun Geng
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Lixia Yin
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Minzhu Niu
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Qingqing Li
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Department of Clinical Laboratory, The Third the People’s Hospital of Bengbu, Bengbu, Anhui, China
| | - Xinyue Liu
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Xinke Cheng
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Xiaofeng Zhang
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Xue Song
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Yueyue Wang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Lian Wang
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Lugen Zuo
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Jianguo Hu
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| |
Collapse
|
6
|
Barra RHD, Piovezan BR, Matheus HR, Vitória OAP, de Abreu Furquim EM, Fiorin LG, Santos EO, de Almeida JM. Effect of coenzyme Q10 on tibial fracture resistance in nicotine-exposed rats. PLoS One 2025; 20:e0315462. [PMID: 39752356 PMCID: PMC11698406 DOI: 10.1371/journal.pone.0315462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/26/2024] [Indexed: 01/06/2025] Open
Abstract
The study aimed to evaluate the potential protection against fractures of oral Q10 supplementation in the tibias of rats exposed to nicotine. Nicotine is known to negatively impact bone density and increase the risk of fractures, in addition to affecting other systems such as the gastrointestinal system, impairing its absorption capacity, negatively affecting bone health. To investigate this, eighty male rats were divided into four groups (n = 20) receiving either nicotine hemisulfate or saline solution (SS) for 28 days. Two daily subcutaneous applications were administered accordingly. Concurrently, vegetable glycerin and Q10 gavage began on day "0". SS: the animals in this group received two daily subcutaneous applications of sodium chloride solution during the entire trial period. 30 days after starting the SS applications subcutaneously, the animals received vegetable glycerin daily until the end of the experiment. SS-Q10: the animals received the SS protocol and daily supplementation with Q10 until the end of the experiment. NIC: The animals received the protocol for NIC and vegetable glycerin daily until the end of the experiment. NIC-Q10: The animals received the protocol for NIC and daily supplementation and Q10 until the end of the experiment. Euthanasia occurred at 7 and 28 days after the beginning the gavage. The tibiae collected were processed for morphometric, densitometric, mechanical, and microtomographic (micro-Ct) analysis. A complementary analysis of intestinal changes was performed. The groups that received Q10 showed slightly better results regarding the mechanical resistance and micro-Ct parameters and to intestinal histomorphometry, as compared with groups not supplemented with Q10. Thus, in rats, it can be concluded that coenzyme Q10 exhibited a protective property to the skeletal system and the gastrointestinal tract, even in the presence of nicotine.
Collapse
Affiliation(s)
- Ruan Henrique Delmonica Barra
- Division of Periodontics, Department of Diagnosis and Surgery, UNESP, São Paulo State University “Júlio de Mesquita Filho”, Araçatuba, Brazil
- Nucleus of Study and Research in Periodontics and Implantology (NEPPI), School of Dentistry, Sao Paulo, State University (Unesp), Aracatuba, SP, Brazil
| | - Bianca Rafaeli Piovezan
- Division of Periodontics, Department of Diagnosis and Surgery, UNESP, São Paulo State University “Júlio de Mesquita Filho”, Araçatuba, Brazil
- Nucleus of Study and Research in Periodontics and Implantology (NEPPI), School of Dentistry, Sao Paulo, State University (Unesp), Aracatuba, SP, Brazil
| | - Henrique Rinaldi Matheus
- Division of Periodontics, Department of Diagnosis and Surgery, UNESP, São Paulo State University “Júlio de Mesquita Filho”, Araçatuba, Brazil
- Nucleus of Study and Research in Periodontics and Implantology (NEPPI), School of Dentistry, Sao Paulo, State University (Unesp), Aracatuba, SP, Brazil
- Discipline of Periodontics, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Otávio Augusto Pacheco Vitória
- Division of Periodontics, Department of Diagnosis and Surgery, UNESP, São Paulo State University “Júlio de Mesquita Filho”, Araçatuba, Brazil
- Nucleus of Study and Research in Periodontics and Implantology (NEPPI), School of Dentistry, Sao Paulo, State University (Unesp), Aracatuba, SP, Brazil
| | - Elisa Mara de Abreu Furquim
- Division of Periodontics, Department of Diagnosis and Surgery, UNESP, São Paulo State University “Júlio de Mesquita Filho”, Araçatuba, Brazil
- Nucleus of Study and Research in Periodontics and Implantology (NEPPI), School of Dentistry, Sao Paulo, State University (Unesp), Aracatuba, SP, Brazil
| | - Luiz Guilherme Fiorin
- Division of Periodontics, Department of Diagnosis and Surgery, UNESP, São Paulo State University “Júlio de Mesquita Filho”, Araçatuba, Brazil
- Nucleus of Study and Research in Periodontics and Implantology (NEPPI), School of Dentistry, Sao Paulo, State University (Unesp), Aracatuba, SP, Brazil
| | - Ester Oliveira Santos
- Division of Periodontics, Department of Diagnosis and Surgery, UNESP, São Paulo State University “Júlio de Mesquita Filho”, Araçatuba, Brazil
- Nucleus of Study and Research in Periodontics and Implantology (NEPPI), School of Dentistry, Sao Paulo, State University (Unesp), Aracatuba, SP, Brazil
| | - Juliano Milanezi de Almeida
- Division of Periodontics, Department of Diagnosis and Surgery, UNESP, São Paulo State University “Júlio de Mesquita Filho”, Araçatuba, Brazil
- Nucleus of Study and Research in Periodontics and Implantology (NEPPI), School of Dentistry, Sao Paulo, State University (Unesp), Aracatuba, SP, Brazil
| |
Collapse
|
7
|
Zhang X, Zuo L, Song X, Zhang W, Yang Z, Wang Z, Guo Y, Ge S, Wang L, Wang Y, Geng Z, Li J, Hu J. The mesenteric adipokine SFRP5 alleviated intestinal epithelial apoptosis improving barrier dysfunction in Crohn's disease. iScience 2024; 27:111517. [PMID: 39759008 PMCID: PMC11699250 DOI: 10.1016/j.isci.2024.111517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 09/21/2024] [Accepted: 11/28/2024] [Indexed: 01/07/2025] Open
Abstract
The hypertrophic mesenteric adipose tissue (htMAT) of Crohn disease (CD) participates in inflammation through the expression of adipokines, but the exact mechanism of this action in the intestine is unknown. Here, we analyzed the expression of secreted frizzled-related protein 5 (SFRP5), an adipokine with cytoprotective effects, in htMAT and its role in CD. The results of this study revealed that the level of SFPR5 increased in the diseased MAT (htMAT) of CD patients and aggregated among intestinal epithelial cells in the diseased intestine and that it could ameliorate intestinal barrier dysfunction in tumor necrosis factor alpha (TNF-α)-stimulated colonic organoids and 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced mice at least in part through the inhibition of Wnt5a-mediated apoptosis in epithelial cells. This study elucidates possible mechanisms by which mesenteric adipokines influence the progression of enteritis and provides a new theoretical basis for the treatment of CD via the mesenteric pathway.
Collapse
Affiliation(s)
- Xiaofeng Zhang
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Lugen Zuo
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Xue Song
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Wenjing Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Zi Yang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Zhiyuan Wang
- Clinical Medical College, Bengbu Medical University, Bengbu, China
| | - Yibing Guo
- Clinical Medical College, Bengbu Medical University, Bengbu, China
| | - Sitang Ge
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Lian Wang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Yueyue Wang
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Zhijun Geng
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Jing Li
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Jianguo Hu
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| |
Collapse
|
8
|
Lin HM, Zhang JR, Li MX, Hou H, Wang H, Huang Y. Cigarette smoking and alcohol-related liver disease. LIVER RESEARCH 2024; 8:237-245. [PMID: 39958918 PMCID: PMC11771264 DOI: 10.1016/j.livres.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/27/2024] [Accepted: 12/04/2024] [Indexed: 02/18/2025]
Abstract
China is a major consumer of alcohol and tobacco. Tobacco and alcohol use are closely linked, with up to 90% of alcoholics having a history of tobacco use, and heavy smokers also tending to be alcoholics. Alcohol-related liver disease (ALD), one of the most common and serious complications of chronic alcohol intake, involving hepatic steatosis, hepatitis, hepatic fibrosis, cirrhosis and hepatocellular carcinoma (HCC), has become one of the globally prevalent chronic diseases. An increasing number of studies have focused on the association between smoking and ALD and explored the mechanisms involved. Clinical evidence suggests that smoking has a negative impact on the incidence and severity of fatty liver disease, progression of liver fibrosis, development of HCC, prognosis of patients with advanced liver disease, and alcohol-related liver transplant recipients. The underlying mechanisms are complex and involve different pathophysiological pathways, including free radical exposure, endoplasmic reticulum stress, insulin resistance, and oncogenic signaling. This review discusses the deleterious effects of smoking on ALD patients and the possible underlying mechanisms at several levels. It emphasizes the importance of discouraging smoking among ALD patients. Finally, the pathogenic role of electronic cigarettes, which have emerged in recent years, is discussed, calling for an emphasis on social missions for young people.
Collapse
Affiliation(s)
- Hui-Min Lin
- Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
- Department of Pharmacy, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jing-Rong Zhang
- Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Meng-Xue Li
- Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Hui Hou
- Department of Pharmacy, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Hua Wang
- Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Yan Huang
- Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
9
|
Larsen MGR, Overgaard SH, Petersen SR, Møllegaard KM, Munk HL, Nexøe AB, Glerup H, Guldmann T, Pedersen N, Saboori S, Dahlerup JF, Hvas CL, Andersen KW, Jawhara M, Haagen Nielsen O, Bergenheim FO, Brodersen JB, Bygum A, Ellingsen T, Kjeldsen J, Christensen R, Andersen V. Effects of smoking on clinical treatment outcomes amongst patients with chronic inflammatory diseases initiating biologics: secondary analyses of the prospective BELIEVE cohort study. Scand J Immunol 2024; 100:e13395. [PMID: 38973149 DOI: 10.1111/sji.13395] [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: 04/09/2024] [Revised: 06/05/2024] [Accepted: 06/22/2024] [Indexed: 07/09/2024]
Abstract
The prevalence and disease burden of chronic inflammatory diseases (CIDs) are predicted to rise. Patients are commonly treated with biological agents, but the individual treatment responses vary, warranting further research into optimizing treatment strategies. This study aimed to compare the clinical treatment responses in patients with CIDs initiating biologic therapy based on smoking status, a notorious risk factor in CIDs. In this multicentre cohort study including 233 patients with a diagnosis of Crohn's disease, ulcerative colitis, rheumatoid arthritis, axial spondyloarthritis, psoriatic arthritis or psoriasis initiating biologic therapy, we compared treatment response rates after 14 to 16 weeks and secondary outcomes between smokers and non-smokers. We evaluated the contrast between groups using logistic regression models: (i) a "crude" model, only adjusted for the CID type, and (ii) an adjusted model (including sex and age). Among the 205 patients eligible for this study, 53 (26%) were smokers. The treatment response rate among smokers (n = 23 [43%]) was lower compared to the non-smoking CID population (n = 92 [61%]), corresponding to a "crude" OR of 0.51 (95% CI: [0.26;1.01]) while adjusting for sex and age resulted in consistent findings: 0.51 [0.26;1.02]. The contrast was apparently most prominent among the 38 RA patients, with significantly lower treatment response rates for smokers in both the "crude" and adjusted models (adjusted OR 0.13, [0.02;0.81]). Despite a significant risk of residual confounding, patients with CIDs (rheumatoid arthritis in particular) should be informed that smoking probably lowers the odds of responding sufficiently to biological therapy. Registration: Clinical.Trials.gov NCT03173144.
Collapse
Affiliation(s)
- Maja Graves Rosenkilde Larsen
- Department of Internal Medicine, Molecular Diagnostics and Clinical Research Unit, University Hospital of Southern Denmark, Aabenraa, Denmark
- Section for Biostatistics and Evidence-Based Research, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
- The Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Silja Hvid Overgaard
- Department of Internal Medicine, Molecular Diagnostics and Clinical Research Unit, University Hospital of Southern Denmark, Aabenraa, Denmark
- Section for Biostatistics and Evidence-Based Research, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Sofie Ronja Petersen
- Department of Clinical Research, University Hospital of Southern Denmark, Odense, Denmark
| | - Karen Mai Møllegaard
- Department of Internal Medicine, Molecular Diagnostics and Clinical Research Unit, University Hospital of Southern Denmark, Aabenraa, Denmark
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Heidi Lausten Munk
- Research Unit of Rheumatology, Department of Clinical Research, University of Southern Denmark, Odense University Hospital, Odense, Denmark
- Center for Rheumatology and Spine Diseases, Copenhagen, Denmark
| | - Anders Bathum Nexøe
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Cancer and Inflammation Research, Odense University Hospital, Odense, Denmark
- Department of Medical Gastroenterology, Odense University Hospital, Odense, Denmark
| | - Henning Glerup
- University Research Clinic for Innovative Patient Pathways, Silkeborg Regional Hospital, Silkeborg, Denmark
| | - Tanja Guldmann
- University Research Clinic for Innovative Patient Pathways, Silkeborg Regional Hospital, Silkeborg, Denmark
| | - Natalia Pedersen
- Department of Gastroenterology, Slagelse Hospital, Slagelse, Denmark
| | - Sanaz Saboori
- Department of Gastroenterology, Slagelse Hospital, Slagelse, Denmark
| | - Jens Frederik Dahlerup
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Christian Lodberg Hvas
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Karina Winther Andersen
- Department of Internal Medicine, Molecular Diagnostics and Clinical Research Unit, University Hospital of Southern Denmark, Aabenraa, Denmark
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Mohamad Jawhara
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Ole Haagen Nielsen
- Department of Gastroenterology, Esbjerg and Grindsted Hospital, University of Southern Denmark, Esbjerg, Denmark
| | - Fredrik Olof Bergenheim
- Department of Gastroenterology, Esbjerg and Grindsted Hospital, University of Southern Denmark, Esbjerg, Denmark
| | - Jacob Broder Brodersen
- Department of Internal Medicine, Molecular Diagnostics and Clinical Research Unit, University Hospital of Southern Denmark, Aabenraa, Denmark
- Department of Surgery, University Hospital of Southern Denmark, Aabenraa, Denmark
| | - Anette Bygum
- Department of Dermatology and Allergy Centre, Odense University Hospital, Odense, Denmark
- Clinical Institute, University of Southern Denmark, Odense, Denmark
| | - Torkell Ellingsen
- Research Unit of Rheumatology, Department of Clinical Research, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | - Jens Kjeldsen
- Department of Medical Gastroenterology, Odense University Hospital, Odense, Denmark
- Research Unit of Medical Gastroenterology, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Robin Christensen
- Section for Biostatistics and Evidence-Based Research, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
- Research Unit of Rheumatology, Department of Clinical Research, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | - Vibeke Andersen
- Department of Internal Medicine, Molecular Diagnostics and Clinical Research Unit, University Hospital of Southern Denmark, Aabenraa, Denmark
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
10
|
Geng Z, Zuo L, Li J, Yin L, Yang J, Duan T, Wang L, Zhang X, Song X, Wang Y, Hu J. Ginkgetin improved experimental colitis by inhibiting intestinal epithelial cell apoptosis through EGFR/PI3K/AKT signaling. FASEB J 2024; 38:e23817. [PMID: 39003633 DOI: 10.1096/fj.202400211rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/15/2024]
Abstract
Excessive apoptosis of intestinal epithelial cells leads to intestinal barrier dysfunction, which is not only one of the pathological features of inflammatory bowel disease (IBD) but also a therapeutic target. A natural plant extract, Ginkgetin (GK), has been reported to have anti-apoptotic activity, but its role in IBD is unknown. This study aimed to explore whether GK has anti-colitis effects and related mechanisms. An experimental colitis model induced by dextran sulfate sodium (DSS) was established, and GK was found to relieve colitis in DSS-induced mice as evidenced by improvements in weight loss, colon shortening, Disease Activity Index (DAI), macroscopic and tissue scores, and proinflammatory mediators. In addition, in DSS mice and TNF-α-induced colonic organoids, GK protected the intestinal barrier and inhibited intestinal epithelial cell apoptosis, by improving permeability and inhibiting the number of apoptotic cells and the expression of key apoptotic regulators (cleaved caspase 3, Bax and Bcl-2). The underlying mechanism of GK's protective effect was explored by bioinformatics, rescue experiments and molecular docking, and it was found that GK might directly target and activate EGFR, thereby interfering with PI3K/AKT signaling to inhibit apoptosis of intestinal epithelial cells in vivo and in vitro. In conclusion, GK inhibited intestinal epithelial apoptosis in mice with experimental colitis, at least in part, by activating EGFR and interfering with PI3K/AKT activation, explaining the underlying mechanism for ameliorating colitis, which may provide new options for the treatment of IBD.
Collapse
Affiliation(s)
- Zhijun Geng
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Lugen Zuo
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Jing Li
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Lixia Yin
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- School of Clinical Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Jingjing Yang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- School of Clinical Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Ting Duan
- School of Clinical Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Lian Wang
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Xiaofeng Zhang
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Xue Song
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Yueyue Wang
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Jianguo Hu
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| |
Collapse
|
11
|
Qiu Q, Geng Z, Wang L, Zuo L, Deng M, Zhang H, Yang Y, Wang Y, Zhao Z, Wen H, Wang Q, Wang Y, He X, Li J, Wang Y, Zhang X, Liu M, Song X. Peiminine ameliorates Crohn's disease-like colitis by enhancing the function of the intestinal epithelial barrier through Nrf2/HO1 signal. Int Immunopharmacol 2024; 136:112380. [PMID: 38850790 DOI: 10.1016/j.intimp.2024.112380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND AND AIMS Impaired intestinal barrier function is key in maintaining intestinal inflammation in Crohn's disease (CD). However, no targeted treatment in clinical practice has been developed. Peiminine (Pm) strongly protects the epithelial barrier, the purpose of this study is to investigate whether Pm affects CD-like colitis and potential mechanisms for its action. METHODS Trinitro-benzene-sulfonic acid (TNBS)-induced mice and Il-10-/- mice were used as CD animal models. Colitis symptoms, histological analysis, and intestinal barrier permeability were used to assess the Pm's therapeutic effect on CD-like colitis. The colon organoids were induced by TNF-α to evaluate the direct role of Pm in inhibiting apoptosis of the intestinal epithelial cells. Western blotting and small molecule inhibitors were used to investigate further the potential mechanism of Pm in inhibiting apoptosis of intestinal epithelial cells. RESULTS Pm treatment reduced body weight loss, disease activity index (DAI) score, and inflammatory score, demonstrating that colonic inflammation in mice were alleviated. Pm decreased the intestinal epithelial apoptosis, improved the intestinal barrier function, and prevented the loss of tight junction proteins (ZO1 and claudin-1) in the colon of CD mice and TNF-α-induced colonic organoids. Pm activated Nrf2/HO1 signaling, which may protect intestinal barrier function. CONCLUSIONS Pm inhibits intestinal epithelial apoptosis in CD mice by activating Nrf2/HO1 pathway. This partially explains the potential mechanism of Pm in ameliorating intestinal barrier function in mice and provides a new approach to treating CD.
Collapse
Affiliation(s)
- Quanwei Qiu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Zhijun Geng
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Lian Wang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Lugen Zuo
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Min Deng
- Department of Gastroenterology, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Hao Zhang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Yiqun Yang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Yijun Wang
- Bengbu Medical University, Bengbu, China
| | | | - Hexin Wen
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Qiusheng Wang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Yitong Wang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Xuxu He
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Jing Li
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Yueyue Wang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Xiaofeng Zhang
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Mulin Liu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China.
| | - Xue Song
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China.
| |
Collapse
|
12
|
Djouina M, Ollivier A, Waxin C, Kervoaze G, Pichavant M, Caboche S, Achour D, Grare C, Beury D, Hot D, Anthérieu S, Lo-Guidice JM, Dubuquoy L, Launay D, Vignal C, Gosset P, Body-Malapel M. Chronic Exposure to Both Electronic and Conventional Cigarettes Alters Ileum and Colon Turnover, Immune Function, and Barrier Integrity in Mice. J Xenobiot 2024; 14:950-969. [PMID: 39051349 PMCID: PMC11270428 DOI: 10.3390/jox14030053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/11/2024] [Accepted: 07/13/2024] [Indexed: 07/27/2024] Open
Abstract
Although the effects of cigarette smoke (CS) on the development of several intestinal diseases is well documented, the impact of e-cigarette aerosol (e-cig) on digestive health is largely unknown. To compare the effects of e-cig and CS on mouse ileum and colon, animals were chronically exposed for 6 months by nose-only inhalation to e-cig at 18 or 30 W power, or to 3R4F CS. Results showed that e-cig exposure decreased colon cell proliferation. Several other proliferative defects were observed in response to both e-cig and CS exposure, including up- and down-regulation of cyclin D1 protein levels in the ileum and colon, respectively. E-cig and CS exposure reduced myeloperoxidase activity in the ileum. In the colon, both exposures disrupted gene expression of cytokines and T cell transcription factors. For tight junction genes, ZO-1- and occludin-protein expression levels were reduced in the ileum and colon, respectively, by e-cig and CS exposure. The 16S sequencing of microbiota showed specific mild dysbiosis, according to the type of exposure. Overall, e-cig exposure led to altered proliferation, inflammation, and barrier function in both the ileum and colon, and therefore may be a gut hazard on par with conventional CS.
Collapse
Affiliation(s)
- Madjid Djouina
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE—Institute for Translational Research in Inflammation, F-59000 Lille, France; (M.D.); (C.W.); (L.D.); (D.L.); (C.V.)
| | - Anaïs Ollivier
- Univ. Lille, CNRS, INSERM, Institut Pasteur de Lille, CHU Lille, Center for Infection and Immunity of Lille (CIIL), UMR9017-U1019, F-59000 Lille, France; (A.O.); (G.K.); (M.P.); (P.G.)
| | - Christophe Waxin
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE—Institute for Translational Research in Inflammation, F-59000 Lille, France; (M.D.); (C.W.); (L.D.); (D.L.); (C.V.)
| | - Gwenola Kervoaze
- Univ. Lille, CNRS, INSERM, Institut Pasteur de Lille, CHU Lille, Center for Infection and Immunity of Lille (CIIL), UMR9017-U1019, F-59000 Lille, France; (A.O.); (G.K.); (M.P.); (P.G.)
| | - Muriel Pichavant
- Univ. Lille, CNRS, INSERM, Institut Pasteur de Lille, CHU Lille, Center for Infection and Immunity of Lille (CIIL), UMR9017-U1019, F-59000 Lille, France; (A.O.); (G.K.); (M.P.); (P.G.)
| | - Ségolène Caboche
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US41-UAR 2014-PLBS, F-59000 Lille, France; (S.C.); (D.B.); (D.H.)
| | - Djamal Achour
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483-IMPECS—IMPact de l’Environnement Chimique sur la Santé, F-59000 Lille, France; (D.A.); (C.G.); (S.A.); (J.-M.L.-G.)
| | - Céline Grare
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483-IMPECS—IMPact de l’Environnement Chimique sur la Santé, F-59000 Lille, France; (D.A.); (C.G.); (S.A.); (J.-M.L.-G.)
| | - Delphine Beury
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US41-UAR 2014-PLBS, F-59000 Lille, France; (S.C.); (D.B.); (D.H.)
| | - David Hot
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US41-UAR 2014-PLBS, F-59000 Lille, France; (S.C.); (D.B.); (D.H.)
| | - Sébastien Anthérieu
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483-IMPECS—IMPact de l’Environnement Chimique sur la Santé, F-59000 Lille, France; (D.A.); (C.G.); (S.A.); (J.-M.L.-G.)
| | - Jean-Marc Lo-Guidice
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483-IMPECS—IMPact de l’Environnement Chimique sur la Santé, F-59000 Lille, France; (D.A.); (C.G.); (S.A.); (J.-M.L.-G.)
| | - Laurent Dubuquoy
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE—Institute for Translational Research in Inflammation, F-59000 Lille, France; (M.D.); (C.W.); (L.D.); (D.L.); (C.V.)
| | - David Launay
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE—Institute for Translational Research in Inflammation, F-59000 Lille, France; (M.D.); (C.W.); (L.D.); (D.L.); (C.V.)
| | - Cécile Vignal
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE—Institute for Translational Research in Inflammation, F-59000 Lille, France; (M.D.); (C.W.); (L.D.); (D.L.); (C.V.)
| | - Philippe Gosset
- Univ. Lille, CNRS, INSERM, Institut Pasteur de Lille, CHU Lille, Center for Infection and Immunity of Lille (CIIL), UMR9017-U1019, F-59000 Lille, France; (A.O.); (G.K.); (M.P.); (P.G.)
| | - Mathilde Body-Malapel
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE—Institute for Translational Research in Inflammation, F-59000 Lille, France; (M.D.); (C.W.); (L.D.); (D.L.); (C.V.)
| |
Collapse
|
13
|
Wang L, Song X, Zhou Y, Xia Y, Yang Z, Chen X, Shi R, Geng Z, Zhang X, Wang Y, Li J, Hu J, Zuo L. Sclareol protected against intestinal barrier dysfunction ameliorating Crohn's disease-like colitis via Nrf2/NF-B/MLCK signalling. Int Immunopharmacol 2024; 133:112140. [PMID: 38669952 DOI: 10.1016/j.intimp.2024.112140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND Inflammation-induced intestinal barrier dysfunction is not only a pathological feature of Crohn's disease (CD) but also an important therapeutic target. Sclareol (SCL) is a nontoxic natural plant compound with anti-inflammatory effect, but its role in CD has not been established. METHODS In vivo studies of mice with TNBS-induced colitis were carried out to evaluate the effects of SCL on CD-like colitis and intestinal barrier function. In vitro, a TNF-α-induced colonic organoid model was established to test the direct effect of SCL on inflammation-induced intestinal barrier injure and inflammatory response. The Nrf2/NF-κB/MLCK signalling was analysed to explore the mechanism of SCL. RESULTS In vivo, SCL largely alleviated the colitis in TNBS mice, as evidenced by improvements in the weight loss, colitis symptoms, endoscopic score, macroscopic histological score, and histological inflammation score. Moreover, SCL significantly improved intestinal barrier dysfunction, manifested as reduced intestinal permeability and decreased intestinal bacterial translocation in TNBS mice. Importantly, SCL antagonised the intestinal mucosal inflammation while protecting tight junctions in TNBS mice. In vitro, SCL largely depressed pro-inflammatory cytokines levels and improved intestinal epithelial permeability in a TNF-α-induced colonic organoid model. In the context of CD, the protective effects of SCL against inflammation and intestinal barrier damage are at least partially results from the Nrf2 signalling activation and the NF-κB/MLCK signalling inhibition. CONCLUSIONS SCL improved intestinal barrier dysfunction and alleviated CD-like colitis, possibly through modulation of Nrf2/NF-κB/MLCK signalling. In view of SCL's safety profile, there is hope that it will be useful in the clinic.
Collapse
Affiliation(s)
- Lian Wang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Xue Song
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Yueqing Zhou
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Yongsheng Xia
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Zi Yang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Xiaohua Chen
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Ruohan Shi
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Zhijun Geng
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Xiaofeng Zhang
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Yueyue Wang
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Jing Li
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Jianguo Hu
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China.
| | - Lugen Zuo
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China.
| |
Collapse
|
14
|
She D, Jiang S, Yuan S. Association between serum cotinine and hepatic steatosis and liver fibrosis in adolescent: a population-based study in the United States. Sci Rep 2024; 14:11424. [PMID: 38763979 PMCID: PMC11102917 DOI: 10.1038/s41598-024-61771-3] [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: 01/24/2024] [Accepted: 05/09/2024] [Indexed: 05/21/2024] Open
Abstract
Tobacco exposure is known to be associated with a higher prevalence and incidence of liver diseases. Cotinine, a metabolite of nicotine, is a typical indicator of tobacco exposure. However, the relationship of serum cotinine levels with hepatic steatosis and liver fibrosis remains controversial and these relationships need more research to explored in American teenagers. Cross-sectional data included 1433 participants aged 12-19 from the National Health and Nutrition Examination Survey (NHANES) from 2017 to 2020 were thoroughly used for this study. The linear relationships between serum cotinine levels and the Liver Stiffness Measurement (LSM) and Controlled Attenuation Parameter (CAP) were examined using multiple linear regression models. Subgroup analysis, interaction tests, and nonlinear interactions were also carried out. Serum cotinine levels > 2.99 ng/ml [β = 0.41 (0.07, 0.76), p = 0.018] and 0.05-2.99 ng/ml [β = 0.24 (0.00, 0.49), p = 0.048] showed a significant positive connection with LSM in multivariate linear regression analysis when compared to serum cotinine levels ≤ 0.05 ng/ml (p for trend = 0.006). Moreover, we discovered an inverted U-shaped association of log2-transformed cotinine with LSM with an inflection point of 4.53 using a two-stage linear regression model. However, according to multiple regression analysis, serum cotinine and CAP did not significantly correlate (p = 0.512). In conclusion, this study demonstrated that smoking cessation and keep away from secondhand smoking may beneficial for liver health in American teenagers.
Collapse
Affiliation(s)
- Dan She
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Shangming Jiang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| | - Siqi Yuan
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| |
Collapse
|
15
|
Li Q, Li J, Yin L, Huang J, Liu X, Shi J, Geng Z, Song X, Wang L, Wang Y, Zhang X, Zuo L, Hu J. Sophoricoside improved Crohn's disease-like colitis by inhibiting intestinal epithelial cell apoptosis through PI3K/AKT signaling. Int Immunopharmacol 2024; 131:111886. [PMID: 38493691 DOI: 10.1016/j.intimp.2024.111886] [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: 01/23/2024] [Revised: 03/05/2024] [Accepted: 03/13/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND AND AIMS Increased apoptosis of intestinal epithelial cells (IECs) is a significant cause of intestinal barrier dysfunction in Crohn's disease (CD). Sophoricoside (SOP) is an isoflavone glycoside known for its anti-apoptotic properties. The aim of this study was to investigate the effects of SOP on mice with CD-like colitis and to understand the underlying mechanisms. METHODS Mice treated with 2,4,6-trinitrobenzene sulfonic acid (TNBS) were used to examine the therapeutic effect of SOP on CD-like colitis and intestinal barrier damage. To further explore SOP's impact on IECs apoptosis and intestinal barrier protection, an in vitro colonic organoid apoptosis model induced by TNF-α was utilized. Network pharmacology was employed to predict the relevant pathways and molecular processes associated with SOP in the treatment of CD. RESULTS Treatment with SOP significantly improved colitis symptoms in TNBS mice, as demonstrated by reductions in the Disease Activity Index (DAI), weight loss, colon shortening, macroscopic scores, colonic tissue inflammatory scores, and the expression of pro-inflammatory factors. Our experiments confirmed that SOP protects the intestinal barrier by counteracting IECs apoptosis. Additionally, this study established that SOP reduced IECs apoptosis by inhibiting the PI3K/AKT signaling pathway. CONCLUSIONS SOP can reduce IECs apoptosis through the inhibition of the PI3K/AKT signaling pathway, thereby protecting the intestinal barrier. This study is the first to illustrate how SOP ameliorates colitis and protects the intestinal barrier, suggesting SOP has potential clinical application in treating CD.
Collapse
Affiliation(s)
- Qingqing Li
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; School of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Jing Li
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China; Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Lixia Yin
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; School of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Ju Huang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; School of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Xinyue Liu
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Jinran Shi
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Zhijun Geng
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China; Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Xue Song
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China; Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Lian Wang
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China; Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Yueyue Wang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China; Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Xiaofeng Zhang
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China; Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Lugen Zuo
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China; Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Jianguo Hu
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China; Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China.
| |
Collapse
|
16
|
Shraideh Z, Badran D, Alzbeede A. Effect of Honey and Aqueous Garlic Extracts against Short-Term Exposure of Cigarette Tobacco Smoking in Mice: Histopathological and Biochemical Investigations. J Toxicol 2024; 2024:5539447. [PMID: 38410096 PMCID: PMC10896654 DOI: 10.1155/2024/5539447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 12/15/2023] [Accepted: 01/09/2024] [Indexed: 02/28/2024] Open
Abstract
It is well known that cigarette smoking adversely affects human health and induces oxidative stress in most vital organs. This study aims to assess the biochemical, histological, and ultrastructural values of honey and garlic extracts in ameliorating the effects of short-term exposure to cigarette smoke in mice. Forty-eight mice were randomly divided into six equal groups: group I was exposed to fresh air only, group II was exposed to cigarette smoke, group III was given 0.2 ml of honey extract, group IV was exposed to cigarette smoke and was given 0.2 ml of honey extract, group V was given 0.2 ml of garlic extract, and group VI was exposed to cigarette smoke and was given 0.2 ml of aqueous garlic extract. These exposures were repeated daily for 21 consecutive days among the treated groups. By the end of the third week, the animals were euthanized by physical cervical dislocation. Blood was taken for biochemical study, and the selected organs of the liver, kidney, and jejunum were processed for histological and ultrastructural studies. The biochemical results showed that short-term exposure of experimental mice to cigarette smoking did not alter the liver function tests except for decreasing the albumin level. Moreover, cigarette smoking elevates the concentration of carbonyl protein content and cystatin C. Histologically, the use of honey and garlic showed good protection to the liver, kidney, and jejunum, which was proved by transmission electron microscopy, in addition to lowering the oxidative stress biomarkers. In conclusion, using honey and/or garlic helps protect the liver, kidney, and jejunum against the hazardous effects of cigarette smoke.
Collapse
Affiliation(s)
- Ziad Shraideh
- Department of Biological Sciences, School of Science, The University of Jordan, Amman 11942, Jordan
| | - Darwish Badran
- Department of Anatomy and Histology, Faculty of Medicine, The University of Jordan, and Ibn Sina University for Medical Sciences, Amman, Jordan
| | - Ahmed Alzbeede
- Department of Medical Laboratory Science, College of Science, Komar University of Science and Technology, Sulaymaniyah 46001, Kurdistan Region, Iraq
| |
Collapse
|
17
|
Szemes K, Farkas N, Sipos Z, Bor R, Fabian A, Szepes Z, Farkas K, Molnar T, Schafer E, Szamosi T, Salamon A, Vincze A, Sarlos P. Co-Administration of Proton Pump Inhibitors May Negatively Affect the Outcome in Inflammatory Bowel Disease Treated with Vedolizumab. Biomedicines 2024; 12:158. [PMID: 38255263 PMCID: PMC10813460 DOI: 10.3390/biomedicines12010158] [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: 12/06/2023] [Revised: 12/31/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Concomitant medications may alter the effect of biological therapy in inflammatory bowel disease. The aim was to investigate the effect of proton pump inhibitors on remission rates in patients with inflammatory bowel disease treated with the gut-selective vedolizumab. Patients from the Hungarian nationwide, multicenter vedolizumab cohort were selected for post hoc analysis. Primary outcomes were the assessment of clinical response and endoscopic and clinical remission at weeks 14 and 54. Secondary outcomes were the evaluation of the combined effect of concomitant steroid therapy and other factors, such as smoking, on remission. A total of 108 patients were identified with proton pump inhibitor data from 240 patients in the original cohort. Patients on steroids without proton pump inhibitors were more likely to have a clinical response at week 14 than patients on concomitant PPI (95% vs. 67%, p = 0.005). Non-smokers with IBD treated with VDZ were more likely to develop a clinical response at week 14 than smokers, particularly those not receiving PPI compared with patients on co-administered PPI therapy (81% vs. 53%, p = 0.041, and 92% vs. 74%, p = 0.029, respectively). We found that the use of PPIs in patients treated with VDZ may impair the achievement of response in certain subgroups. Unnecessary PPI prescriptions should be avoided.
Collapse
Affiliation(s)
- Kata Szemes
- Division of Gastroenterology, First Department of Medicine, Medical School, University of Pécs, 13 Ifjúság Street, 7624 Pecs, Hungary
| | - Nelli Farkas
- Institute of Bioanalysis, Medical School, University of Pécs, 7624 Pecs, Hungary
| | - Zoltan Sipos
- Institute of Bioanalysis, Medical School, University of Pécs, 7624 Pecs, Hungary
| | - Renata Bor
- First Department of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Anna Fabian
- First Department of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Zoltan Szepes
- First Department of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Klaudia Farkas
- First Department of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Tamas Molnar
- First Department of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Eszter Schafer
- Department of Gastroenterology, Hungarian Defence Forces Military Hospital, 1134 Budapest, Hungary
| | - Tamas Szamosi
- Department of Gastroenterology, Hungarian Defence Forces Military Hospital, 1134 Budapest, Hungary
| | | | - Aron Vincze
- Division of Gastroenterology, First Department of Medicine, Medical School, University of Pécs, 13 Ifjúság Street, 7624 Pecs, Hungary
| | - Patricia Sarlos
- Division of Gastroenterology, First Department of Medicine, Medical School, University of Pécs, 13 Ifjúság Street, 7624 Pecs, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pecs, Hungary
| |
Collapse
|
18
|
Song X, Dou X, Chang J, Zeng X, Xu Q, Xu C. The role and mechanism of gut-lung axis mediated bidirectional communication in the occurrence and development of chronic obstructive pulmonary disease. Gut Microbes 2024; 16:2414805. [PMID: 39446051 PMCID: PMC11509012 DOI: 10.1080/19490976.2024.2414805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/21/2024] [Accepted: 09/30/2024] [Indexed: 10/25/2024] Open
Abstract
The current studies have shown that the occurrence and development of chronic obstructive pulmonary disease (COPD) are closely related to the changes in gut health and its microenvironment, and even some gut diseases have significant clinical correlation with COPD. The dysbiosis of gut microbiota observed in COPD patients also suggests a potential bidirectional interaction between the gut and lung. Communication between the gut and lung may occur through circulating inflammatory cells, gut microbial metabolites, and circulating inflammatory mediators, but the mechanism of bidirectional communication between the gut and lung in COPD is still under study. Therefore, more research is still needed in this area. In this review, we summarize recent clinical studies and animal models on the role of the gut-lung axis in the occurrence and development of COPD and its mechanisms, so as to provide ideas for further research in this field. In addition, we also summarized the negative effects of COPD medication on gut microbiota and the gut microbiota risk factors for COPD and proposed the potential prevention and treatment strategies.
Collapse
Affiliation(s)
- Xiaofan Song
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, Shaanxi, China
| | - Xina Dou
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, Shaanxi, China
| | - Jiajing Chang
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, Shaanxi, China
| | - Xiaonan Zeng
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, Shaanxi, China
| | - Qinhong Xu
- Department of Geriatric Surgery, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Chunlan Xu
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, Shaanxi, China
| |
Collapse
|
19
|
Simão DO, Vieira VS, Tosatti JAG, Gomes KB. Lipids, Gut Microbiota, and the Complex Relationship with Alzheimer's Disease: A Narrative Review. Nutrients 2023; 15:4661. [PMID: 37960314 PMCID: PMC10649859 DOI: 10.3390/nu15214661] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/28/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Alzheimer's Disease (AD) is a multifactorial, progressive, and chronic neurodegenerative disorder associated with the aging process. Memory deficits, cognitive impairment, and motor dysfunction are characteristics of AD. It is estimated that, by 2050, 131.5 million people will have AD. There is evidence that the gastrointestinal microbiome and diet may contribute to the development of AD or act preventively. Communication between the brain and the intestine occurs through immune cells in the mucosa and endocrine cells, or via the vagus nerve. Aging promotes intestinal dysbiosis, characterized by an increase in pro-inflammatory pathogenic bacteria and a reduction in anti-inflammatory response-mediating bacteria, thus contributing to neuroinflammation and neuronal damage, ultimately leading to cognitive decline. Therefore, the microbiota-gut-brain axis has a significant impact on neurodegenerative disorders. Lipids may play a preventive or contributory role in the development of AD. High consumption of saturated and trans fats can increase cortisol release and lead to other chronic diseases associated with AD. Conversely, low levels of omega-3 polyunsaturated fatty acids may be linked to neurodegenerative diseases. Unlike other studies, this review aims to describe, in an integrative way, the interaction between the gastrointestinal microbiome, lipids, and AD, providing valuable insights into how the relationship between these factors affects disease progression, contributing to prevention and treatment strategies.
Collapse
Affiliation(s)
- Daiane Oliveira Simão
- Faculty of Medicine, Federal University of Minas Gerais, Professor Alfredo Balena Avenue, 190, Santa Efigênia, Belo Horizonte 30130-100, MG, Brazil;
| | - Vitoria Silva Vieira
- Department of Nutrition, School of Nursing, Federal University of Minas Gerais, Professor Alfredo Balena Avenue, 190, Santa Efigênia, Belo Horizonte 30130-100, MG, Brazil;
| | - Jéssica Abdo Gonçalves Tosatti
- Department of Clinical and Toxicological Analyzes, Faculty of Pharmacy, Federal University of Minas Gerais, Presidente Antônio Carlos Avenue, 6627, Pampulha, Belo Horizonte 31270-901, MG, Brazil;
| | - Karina Braga Gomes
- Faculty of Medicine, Federal University of Minas Gerais, Professor Alfredo Balena Avenue, 190, Santa Efigênia, Belo Horizonte 30130-100, MG, Brazil;
- Department of Clinical and Toxicological Analyzes, Faculty of Pharmacy, Federal University of Minas Gerais, Presidente Antônio Carlos Avenue, 6627, Pampulha, Belo Horizonte 31270-901, MG, Brazil;
| |
Collapse
|
20
|
Lim EY, Song EJ, Shin HS. Gut Microbiome as a Possible Cause of Occurrence and Therapeutic Target in Chronic Obstructive Pulmonary Disease. J Microbiol Biotechnol 2023; 33:1111-1118. [PMID: 37164760 PMCID: PMC10580882 DOI: 10.4014/jmb.2301.01033] [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: 01/26/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 05/12/2023]
Abstract
As a long-term condition that affects the airways and lungs, chronic obstructive pulmonary disease (COPD) is characterized by inflammation, emphysema, breathlessness, chronic cough, and sputum production. Currently, the bronchodilators and anti-inflammatory drugs prescribed for COPD are mostly off-target, warranting new disease management strategies. Accumulating research has revealed the gut-lung axis to be a bidirectional communication system. Cigarette smoke, a major exacerbating factor in COPD and lung inflammation, affects gut microbiota composition and diversity, causing gut microbiota dysbiosis, a condition that has recently been described in COPD patients and animal models. For this review, we focused on the gut-lung axis, which is influenced by gut microbial metabolites, bacterial translocation, and immune cell modulation. Further, we have summarized the findings of preclinical and clinical studies on the association between gut microbiota and COPD to provide a basis for using gut microbiota in therapeutic strategies against COPD. Our review also proposes that further research on probiotics, prebiotics, short-chain fatty acids, and fecal microbiota transplantation could assist therapeutic approaches targeting the gut microbiota to alleviate COPD.
Collapse
Affiliation(s)
- Eun Yeong Lim
- Food Functionality Research Division, Wanju-gun, Jeollabuk-do 55365, Republic of Korea
| | - Eun-Ji Song
- Food Functionality Research Division, Wanju-gun, Jeollabuk-do 55365, Republic of Korea
| | - Hee Soon Shin
- Food Functionality Research Division, Wanju-gun, Jeollabuk-do 55365, Republic of Korea
- Department of Food Biotechnology, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| |
Collapse
|
21
|
Chelslín F, Lodefalk M, Kruse R. Smoking during pregnancy is associated with the placental proteome. Reprod Toxicol 2023; 119:108409. [PMID: 37209868 DOI: 10.1016/j.reprotox.2023.108409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/10/2023] [Accepted: 05/17/2023] [Indexed: 05/22/2023]
Abstract
Maternal smoking during pregnancy (MSDP) is a significant risk factor for the development of foetal, neonatal, and childhood morbidities. We hypothesized that infants exposed to MSDP have a distinct proteomic expression in their term placentas compared to infants without such an exposure. A total of 39 infants exposed (cord blood cotinine levels of >1 ng/mL) and 44 infants not exposed to MSDP were included in the study. Women with chronic disease, body mass index of > 30, or a history of uterine surgery were excluded. Total proteome abundance was analysed with quantitative mass spectrometry. For univariate analysis of differences in placental protein levels between groups, ANOVA with multiple testing corrections by the Benjamini-Hochberg method was used. For multivariate analysis, we used principal component analysis, partial least squares, lasso, random forest, and neural networks. The univariate analyses showed four differentially abundant proteins (PXDN, CYP1A1, GPR183, and KRT81) when heavy and moderate smoking groups were compared to non-smokers. With the help of machine learning, we found that an additional six proteins (SEPTIN3, CRAT, NAAA, CD248, CADM3, and ZNF648) were discriminants of MSDP. The placental abundance of these ten proteins together explained 74.1% of the variation in cord blood cotinine levels (p = 0.002). Infants exposed to MSDP showed differential abundance of proteins in term placentas. We report differential placental abundance of several proteins for the first time in the setting of MSDP. We believe that these findings supplement the current understanding of how MSDP affects the placental proteome.
Collapse
Affiliation(s)
- Felix Chelslín
- University Health Care Research Centre, Faculty of Medicine and Health, Örebro University, SE-701 82 Örebro, Sweden.
| | - Maria Lodefalk
- University Health Care Research Centre, Faculty of Medicine and Health, Örebro University, SE-701 82 Örebro, Sweden; Department of Paediatrics, Faculty of Medicine and Health, Örebro University, SE-701 82 Örebro, Sweden
| | - Robert Kruse
- Department of Clinical Research Laboratory, Faculty of Medicine and Health, Örebro University, SE-701 82 Örebro, Sweden; Inflammatory Response and Infection Susceptibility Centre (iRiSC) and X-HiDE Consortium, Faculty of Medicine and Health, Örebro University, SE-701 82 Örebro, Sweden
| |
Collapse
|
22
|
Zhang X, Zuo L, Geng Z, Song X, Li J, Ge S, Jiang Y, Yang Z, Liu G, Zhao Y, Zhao H, Yu L, Hu J. Vindoline ameliorates intestinal barrier damage in Crohn's disease mice through MAPK signaling pathway. FASEB J 2022; 36:e22589. [PMID: 36197455 DOI: 10.1096/fj.202200234rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 08/09/2022] [Accepted: 09/23/2022] [Indexed: 11/11/2022]
Abstract
Intestinal inflammation and intestinal barrier damage are important pathological changes in Crohn's disease (CD). Vindoline is a natural monomer with anti-inflammatory effects. We employed CD model mice to explore the effect of Vindoline on CD-like colitis and the possible mechanism. Il-10-deficient (Il-10-/- ) mice and wild-type (WT) mice (both aged 15 weeks, male) were used to explore the effect of Vindoline on colitis and intestinal barrier damage, as well as macrophage-mediated inflammation. Bone-marrow-derived macrophages (BMDMs) and colonic organoids from mice were used to explore the inhibitory effect of Vindoline on macrophage-mediated inflammation and the protective effect on inflammation-induced intestinal barrier damage as well as the possible mechanism. We found that Vindoline significantly ameliorated colitis in CD mice, as evidenced by increased weight change and colon length and decreased the colon macroscopic injury score, histological inflammatory score, and the expression of pro-inflammatory mediators. Vindoline also protected against intestinal barrier damage in CD mice. Furthermore, Vindoline inhibited macrophage-mediated inflammation and protected against inflammation-induced intestinal barrier damage in the coculture system. In addition, Vindoline ameliorated colitis in CD mice by protecting against inflammation-induced intestinal barrier damage, which may be caused by inhibition of MAPK signaling pathway. This protective effect suggests that Vindoline has potential value for clinical application in the treatment of CD.
Collapse
Affiliation(s)
- Xiaofeng Zhang
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Lugen Zuo
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhijun Geng
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Xue Song
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Jing Li
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Sitang Ge
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yifan Jiang
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Zi Yang
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Guangyong Liu
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Yajing Zhao
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Hao Zhao
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Liang Yu
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Jianguo Hu
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China.,Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|
23
|
Smoking-induced microbial dysbiosis in health and disease. Clin Sci (Lond) 2022; 136:1371-1387. [PMID: 36156126 PMCID: PMC9527826 DOI: 10.1042/cs20220175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/09/2022] [Accepted: 09/07/2022] [Indexed: 11/25/2022]
Abstract
Smoking is associated with an increased risk of cancer, pulmonary and cardiovascular diseases, but the precise mechanisms by which such risk is mediated remain poorly understood. Additionally, smoking can impact the oral, nasal, oropharyngeal, lung and gut microbiome composition, function, and secreted molecule repertoire. Microbiome changes induced by smoking can bear direct consequences on smoking-related illnesses. Moreover, smoking-associated dysbiosis may modulate weight gain development following smoking cessation. Here, we review the implications of cigarette smoking on microbiome community structure and function. In addition, we highlight the potential impacts of microbial dysbiosis on smoking-related diseases. We discuss challenges in studying host–microbiome interactions in the context of smoking, such as the correlations with smoking-related disease severity versus causation and mechanism. In all, understanding the microbiome’s role in the pathophysiology of smoking-related diseases may promote the development of rational therapies for smoking- and smoking cessation-related disorders, as well as assist in smoking abstinence.
Collapse
|
24
|
Marti-Aguado D, Clemente-Sanchez A, Bataller R. Cigarette smoking and liver diseases. J Hepatol 2022; 77:191-205. [PMID: 35131406 DOI: 10.1016/j.jhep.2022.01.016] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/14/2022] [Accepted: 01/25/2022] [Indexed: 01/27/2023]
Abstract
Cigarette smoking is a preventable risk factor for premature morbidity and mortality. A history of smoking is observed in approximately 40% of patients with liver disease, while a growing number of studies are investigating the potential impact of smoking in chronic liver diseases. This review discusses the effects of smoking on liver diseases, at multiple levels, with a focus on its potential causal role. Clinical evidence indicates that cigarette smoking negatively impacts the incidence and severity of fatty liver disease, fibrosis progression, hepatocellular carcinoma development, and the outcomes of patients with advanced liver disease. The underlying mechanisms are complex and involve different pathophysiological pathways including oxidative stress and oncogenic signals. Importantly, smoking promotes cardiovascular disease and extrahepatic cancers in patients with steatohepatitis and in transplant recipients. We discuss how promoting smoking cessation could improve the rates of treatment response (in clinical trials) and fibrosis regression, while reducing the risk of hepatocellular carcinoma and improving liver transplant outcomes. Finally, we discuss current challenges such as the referral of smokers to specialised units for smoking cessation.
Collapse
Affiliation(s)
- David Marti-Aguado
- Digestive Disease Department, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain; Center for Liver Diseases, Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Pittsburgh Liver Research Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Ana Clemente-Sanchez
- Center for Liver Diseases, Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Pittsburgh Liver Research Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA; Liver Unit and Digestive Department, Hospital General Universitario Gregorio Marañon, CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Ramon Bataller
- Center for Liver Diseases, Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Pittsburgh Liver Research Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| |
Collapse
|
25
|
Martel J, Chang SH, Ko YF, Hwang TL, Young JD, Ojcius DM. Gut barrier disruption and chronic disease. Trends Endocrinol Metab 2022; 33:247-265. [PMID: 35151560 DOI: 10.1016/j.tem.2022.01.002] [Citation(s) in RCA: 250] [Impact Index Per Article: 83.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 01/03/2022] [Accepted: 01/10/2022] [Indexed: 02/06/2023]
Abstract
The intestinal barrier protects the host against gut microbes, food antigens, and toxins present in the gastrointestinal tract. However, gut barrier integrity can be affected by intrinsic and extrinsic factors, including genetic predisposition, the Western diet, antibiotics, alcohol, circadian rhythm disruption, psychological stress, and aging. Chronic disruption of the gut barrier can lead to translocation of microbial components into the body, producing systemic, low-grade inflammation. While the association between gut barrier integrity and inflammation in intestinal diseases is well established, we review here recent studies indicating that the gut barrier and microbiota dysbiosis may contribute to the development of metabolic, autoimmune, and aging-related disorders. Emerging interventions to improve gut barrier integrity and microbiota composition are also described.
Collapse
Affiliation(s)
- Jan Martel
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan; Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Shih-Hsin Chang
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan
| | - Yun-Fei Ko
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Chang Gung Biotechnology Corporation, Taipei, Taiwan; Biochemical Engineering Research Center, Ming Chi University of Technology, New Taipei City, Taiwan
| | - Tsong-Long Hwang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan; Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - John D Young
- Chang Gung Biotechnology Corporation, Taipei, Taiwan.
| | - David M Ojcius
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan; Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Department of Biomedical Sciences, Arthur Dugoni School of Dentistry, University of the Pacific, San Francisco, CA, USA.
| |
Collapse
|
26
|
Traini C, Nistri S, Calosi L, Vannucchi MG. Chronic Exposure to Cigarette Smoke Affects the Ileum and Colon of Guinea Pigs Differently. Relaxin (RLX-2, Serelaxin) Prevents Most Local Damage. Front Pharmacol 2022; 12:804623. [PMID: 35095510 PMCID: PMC8793690 DOI: 10.3389/fphar.2021.804623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/09/2021] [Indexed: 11/13/2022] Open
Abstract
Cigarette smoking (CS) is the cause of several organ and apparatus diseases. The effects of smoke in the gut are partially known. Accumulating evidence has shown a relationship between smoking and inflammatory bowel disease, prompting us to investigate the mechanisms of action of smoking in animal models. Despite the role played by neuropeptides in gut inflammation, there are no reports on their role in animal models of smoking exposure. The hormone relaxin has shown anti-inflammatory properties in the intestine, and it might represent a putative therapy to prevent gut damage caused by smoking. Presently, we investigate the effects of chronic smoke exposure on inflammation, mucosal secretion, and vasoactive intestinal peptide (VIP) and substance P (SP) expressions in the ileum and colon of guinea pigs. We also verify the ability of relaxin to counter the smoke-induced effects. Smoke impacted plasma carbon monoxide (CO). In the ileum, it induced inflammatory infiltrates, fibrosis, and acidic mucin production; reduced the blood vessel area; decreased c-kit-positive mast cells and VIP-positive neurons; and increased the SP-positive nerve fibers. In the colon, it reduced the blood vessel area and the goblet cell area and decreased c-kit-positive mast cells, VIP-positive neurons, and SP-positive nerve fibers. Relaxin prevented most of the smoking-induced changes in the ileum, while it was less effective in the colon. This study shows the diverse sensitivity to CS between the ileum and the colon and demonstrates that both VIP and SP are affected by smoking. The efficacy of relaxin proposes this hormone as a potential anti-inflammatory therapeutic to counteract gut damage in humans affected by inflammatory bowel diseases.
Collapse
Affiliation(s)
- Chiara Traini
- Department of Experimental and Clinical Medicine, Research Unit of Histology and Embryology, University of Florence, Florence, Italy
| | - Silvia Nistri
- Department of Experimental and Clinical Medicine, Research Unit of Histology and Embryology, University of Florence, Florence, Italy
| | - Laura Calosi
- Department of Experimental and Clinical Medicine, Research Unit of Histology and Embryology, University of Florence, Florence, Italy
| | - Maria Giuliana Vannucchi
- Department of Experimental and Clinical Medicine, Research Unit of Histology and Embryology, University of Florence, Florence, Italy
| |
Collapse
|
27
|
Di Tommaso N, Gasbarrini A, Ponziani FR. Intestinal Barrier in Human Health and Disease. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182312836. [PMID: 34886561 PMCID: PMC8657205 DOI: 10.3390/ijerph182312836] [Citation(s) in RCA: 267] [Impact Index Per Article: 66.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 02/07/2023]
Abstract
The intestinal mucosa provides a selective permeable barrier for nutrient absorption and protection from external factors. It consists of epithelial cells, immune cells and their secretions. The gut microbiota participates in regulating the integrity and function of the intestinal barrier in a homeostatic balance. Pathogens, xenobiotics and food can disrupt the intestinal barrier, promoting systemic inflammation and tissue damage. Genetic and immune factors predispose individuals to gut barrier dysfunction, and changes in the composition and function of the gut microbiota are central to this process. The progressive identification of these changes has led to the development of the concept of ‘leaky gut syndrome’ and ‘gut dysbiosis’, which underlie the relationship between intestinal barrier impairment, metabolic diseases and autoimmunity. Understanding the mechanisms underlying this process is an intriguing subject of research for the diagnosis and treatment of various intestinal and extraintestinal diseases.
Collapse
Affiliation(s)
- Natalia Di Tommaso
- Division of Internal Medicine, Gastroenterology—Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (N.D.T.); (A.G.)
| | - Antonio Gasbarrini
- Division of Internal Medicine, Gastroenterology—Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (N.D.T.); (A.G.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Division of Internal Medicine, Gastroenterology—Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (N.D.T.); (A.G.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy
- Correspondence: ; Tel.: +39-3471227242
| |
Collapse
|
28
|
Bamias G, Cominelli F. Exploring the Early Phase of Crohn's Disease. Clin Gastroenterol Hepatol 2021; 19:2469-2480. [PMID: 32949730 PMCID: PMC9217179 DOI: 10.1016/j.cgh.2020.09.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/24/2020] [Accepted: 09/12/2020] [Indexed: 02/06/2023]
Abstract
The development of Crohn's disease (CD) is characterized by a breakdown of homeostatic immune-bacterial communication, which takes place at the intestinal mucosa when environmental triggers impact genetically predisposed individuals. Converging lines of evidence support the hypothesis that this pathogenetic model develops through sequential, although inter-related, steps that indicate failure of mucosal defense mechanisms at various stages. In this context, immunologic phenomena that mediate the initial appearance of inflammatory lesions across the intestinal tissue may differ substantially from those that mediate and perpetuate chronic inflammatory responses. A compromise in the integrity of the epithelial barrier is among the earliest events and leads to accelerated influx of intraluminal antigens and intact microorganisms within the immunologically rich lamina propria. Inadequate clearance of invading microorganisms also may occur as a result of defects in innate immunity, preventing the timely and complete resolution of acute inflammatory responses. The final step is the development of persistent adaptive responses, which also differ between early and late Crohn's disease. Current progress in our ability to delineate single-cell transcriptomics and proteomics has allowed the discovery of cellular and molecular mechanisms that participate in each sequential step of CD development. This not only will advance our understanding of CD pathogenesis, but also facilitate the design of targeted therapeutic approaches.
Collapse
Affiliation(s)
- Giorgos Bamias
- Gastrointestinal Unit, Third Academic Department of Internal Medicine, National and Kapodistrian University of Athens, Sotiria Hospital, Athens, Greece
| | - Fabio Cominelli
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio.
| |
Collapse
|
29
|
Gallucci G, Santucci N, Díaz A, Bongiovanni B, Bértola D, Gardeñez W, Rassetto M, Bay ML, Bottasso O, D’Attilio L. Increased levels of circulating LPS during Tuberculosis prevails in patients with advanced pulmonary involvement. PLoS One 2021; 16:e0257214. [PMID: 34506568 PMCID: PMC8432878 DOI: 10.1371/journal.pone.0257214] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/25/2021] [Indexed: 11/18/2022] Open
Abstract
Our earlier studies in tuberculosis (TB) patients indicate that in those where the process evolves to a larger pulmonary involvement, the immune endocrine response may promote an unfavorable environment. Chronic infectious diseases, and their persistent proinflammatory response, may affect mucosal barriers integrity favoring the translocation of gastrointestinal bacteria, leading to an increase of circulating lipopolysaccharides (LPS). Consequently, we quantified LPS levels in TB patients, with different degrees of pulmonary involvement, and controls (Co) and analyzed the possible relationship between LPS and inflammatory mediators i.e., C reactive protein (CRP), interleukin 6 (IL-6) and Interferon-gamma (IFN-γ), Erythrocyte Sedimentation Rate (ESR), steroid hormones (Cortisol and Dehydroepiandrosterone, DHEA), and inflammatory transcripts from peripheral blood mononuclear cells (IL-1β, IL-6, IFN-γ). LPS was assessed by the Limulus amoebocyte lysate assay and the ELISA technique was used to quantify hormones and cytokines in the plasma samples. Cytokine transcripts from PBMC were evaluated by qRT-PCR. Non-parametric tests were used. LPS levels were increased in TB patients, as did levels of CRP, IL-6, IFN-γ, cortisol and ESR. Severe patients had the highest amounts of circulating LPS; with moderate and severe cases showing much higher levels of CRP, ESR, IL-6, IFN-γ and cortisol/DHEA ratio, as an endocrine imbalance. Only in PBMC from severe cases was mRNA for IL-1β increased. Correlation analysis showed that levels of LPS from severe patients were positively associated with IL-6 and IFN-γ plasma concentrations and with IL-1β transcripts, while IL-6 had a positive correlation with the cortisol/DHEA ratio. The higher levels of circulating LPS during progressive TB may emerge as a contributing factor for the persistence of the greater immune endocrine imbalance distinctive of advanced disease, which might suggest a vicious cycle among LPS, inflammation and endocrine imbalance.
Collapse
Affiliation(s)
- Georgina Gallucci
- Instituto de Inmunología Clínica y Experimental Rosario (IDICER), CONICET-UNR, Rosario, Santa Fe, Argentina
| | - Natalia Santucci
- Instituto de Inmunología Clínica y Experimental Rosario (IDICER), CONICET-UNR, Rosario, Santa Fe, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Santa Fe, Argentina
| | - Ariana Díaz
- Instituto de Inmunología Clínica y Experimental Rosario (IDICER), CONICET-UNR, Rosario, Santa Fe, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Santa Fe, Argentina
| | - Bettina Bongiovanni
- Instituto de Inmunología Clínica y Experimental Rosario (IDICER), CONICET-UNR, Rosario, Santa Fe, Argentina
| | - Diego Bértola
- Servicio de Clínica Médica, Hospital Provincial del Centenario, Rosario, Santa Fe, Argentina
| | - Walter Gardeñez
- Servicio de Neumología, Hospital Provincial del Centenario, Rosario, Santa Fe, Argentina
| | - Mauricio Rassetto
- Centro de investigación y biotecnología (CIBIO) Wiener Lab, Rosario, Argentina
| | - María Luisa Bay
- Instituto de Inmunología Clínica y Experimental Rosario (IDICER), CONICET-UNR, Rosario, Santa Fe, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Santa Fe, Argentina
| | - Oscar Bottasso
- Instituto de Inmunología Clínica y Experimental Rosario (IDICER), CONICET-UNR, Rosario, Santa Fe, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Santa Fe, Argentina
| | - Luciano D’Attilio
- Instituto de Inmunología Clínica y Experimental Rosario (IDICER), CONICET-UNR, Rosario, Santa Fe, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Santa Fe, Argentina
| |
Collapse
|
30
|
Janbazacyabar H, van Daal M, Leusink-Muis T, van Ark I, Garssen J, Folkerts G, van Bergenhenegouwen J, Braber S. The Effects of Maternal Smoking on Pregnancy and Offspring: Possible Role for EGF? Front Cell Dev Biol 2021; 9:680902. [PMID: 34485278 PMCID: PMC8415274 DOI: 10.3389/fcell.2021.680902] [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: 03/15/2021] [Accepted: 07/29/2021] [Indexed: 11/16/2022] Open
Abstract
Cigarette smoke exposure during pregnancy and lactation is associated with adverse pregnancy outcomes. Here, we investigated the effects of maternal smoke exposure on pregnancy and offspring immunity and explored whether, epidermal growth factor (EGF), an important growth-promoting factor in human colostrum and milk, might be a possible missing link in maternal smoke exposure and changes in infants’ immune responses. Pregnant BALB/c mice were exposed to either cigarette smoke or air during gestation and lactation, and effects on pulmonary inflammation in dams and immune responses in offspring were examined. Maternal smoke exposure increased airway hyperresponsiveness and accumulation of inflammatory cells in the lungs of pregnant dams compared to non-pregnant dams. The E-cadherin protein expression was reduced in mammary glands of cigarette smoke-exposed pregnant dams. EGF levels were higher in mammary glands and serum of smoke-exposed pregnant dams compared to air-exposed pregnant dams. Offspring from cigarette smoke-exposed dams exhibited elevated levels of IL-17A, MCP-1, IL-22, and IL-13 in anti-CD3 stimulated spleen cell culture supernatants. EGF levels were also increased in serum of offspring from smoke-exposed dams. A positive correlation was observed between serum EGF levels and neutrophil numbers in bronchoalveolar lavage fluid of the dams. Interestingly, IL-17A, MCP-1, IL-22, IL13, and IFN-γ levels in anti-CD3 stimulated spleen cell culture supernatants of male pups also showed a positive correlation with EGF serum levels. In summary, our results reveal that maternal smoke exposure predisposes dams to exacerbated airway inflammation and offspring to exacerbated immune responses and both phenomena are associated with elevated EGF concentrations.
Collapse
Affiliation(s)
- Hamed Janbazacyabar
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Marthe van Daal
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Thea Leusink-Muis
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Ingrid van Ark
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands.,Danone Nutricia Research, Utrecht, Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Jeroen van Bergenhenegouwen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands.,Danone Nutricia Research, Utrecht, Netherlands
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
31
|
Ratajczak AE, Szymczak-Tomczak A, Rychter AM, Zawada A, Dobrowolska A, Krela-Kaźmierczak I. Impact of Cigarette Smoking on the Risk of Osteoporosis in Inflammatory Bowel Diseases. J Clin Med 2021; 10:1515. [PMID: 33916465 PMCID: PMC8038608 DOI: 10.3390/jcm10071515] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 03/20/2021] [Accepted: 03/23/2021] [Indexed: 12/14/2022] Open
Abstract
Cigarette smoking constitutes one of the most important modifiable factors of osteoporosis, as well as contributes to an early death, tumors, and numerous chronic diseases. The group with an increased risk of a lower bone mineral density are patients suffering from inflammatory bowel diseases. In fact, tobacco smoke, which contains more than 7000 chemical compounds, affects bone mineral density (BMD) both directly and indirectly, as it has an impact on the RANK-RANKL-OPG pathway, intestinal microbiota composition, and calcium-phosphate balance. Constant cigarette use interferes with the production of protective mucus and inhibits the repair processes in the intestinal mucus. Nicotine as well as the other compounds of the cigarette smoke are important risk factors of the inflammatory bowel disease and osteoporosis. Additionally, cigarette smoking may decrease BMD in the IBD patients. Interestingly, it affects patients with Crohn's disease and ulcerative colitis in different ways-on the one hand it protects against ulcerative colitis, whereas on the other it increases the risk of Crohn's disease development. Nevertheless, all patients should be encouraged to cease smoking in order to decrease the risk of developing other disorders.
Collapse
Affiliation(s)
- Alicja Ewa Ratajczak
- Correspondence: (A.E.R.); (I.K.-K.); Tel.: +48-667-385-996 (A.E.R.); +48-8691-343 (I.K.-K.); Fax: +48-8691-686 (A.E.R.)
| | | | | | | | | | - Iwona Krela-Kaźmierczak
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznań University of Medical Sciences, 61-701 Poznań, Poland; (A.S.-T.); (A.M.R.); (A.Z.); (A.D.)
| |
Collapse
|
32
|
Wang Y, Lim YY, He Z, Wong WT, Lai WF. Dietary phytochemicals that influence gut microbiota: Roles and actions as anti-Alzheimer agents. Crit Rev Food Sci Nutr 2021; 62:5140-5166. [PMID: 33559482 DOI: 10.1080/10408398.2021.1882381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The last decide has witnessed a growing research interest in the role of dietary phytochemicals in influencing the gut microbiota. On the other hand, recent evidence reveals that dietary phytochemicals exhibit properties of preventing and tackling symptoms of Alzheimer's disease, which is a neurodegenerative disease that has also been linked with the status of the gut microbiota over the last decade. Till now, little serious discussions, however, have been made to link recent understanding of Alzheimer's disease, dietary phytochemicals and the gut microbiota together and to review the roles played by phytochemicals in gut dysbiosis induced pathologies of Alzheimer's disease. Deciphering these connections can provide insights into the development and future use of dietary phytochemicals as anti-Alzheimer drug candidates. This review aims at presenting latest evidence in the modulating role of phytochemicals in the gut microbiota and its relevance to Alzheimer's disease and summarizing the mechanisms behind the modulative activities. Limitations of current research in this field and potential directions will also be discussed for future research on dietary phytochemicals as anti-Alzheimer agents.
Collapse
Affiliation(s)
- Yi Wang
- School of Agriculture and Food Sciences, University of Queensland, St Lucia, Queensland, Australia.,School of Dentistry, University of Queensland, Herston, Queensland, Australia
| | - Yau-Yan Lim
- School of Science, Monash University, Bandar Sunway, Selangor, Malaysia
| | - Zhendan He
- College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| | - Wing-Tak Wong
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Wing-Fu Lai
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China.,School of Life and Health Sciences, The Chinese University of Hong Kong (Shenzhen), Shenzhen, China
| |
Collapse
|
33
|
Fusco R, Cordaro M, Siracusa R, Peritore AF, Gugliandolo E, Genovese T, D’Amico R, Crupi R, Smeriglio A, Mandalari G, Impellizzeri D, Cuzzocrea S, Di Paola R. Consumption of Anacardium Occidentale L. (Cashew Nuts) Inhibits Oxidative Stress through Modulation of the Nrf2/HO-1 and NF-kB Pathways. Molecules 2020; 25:molecules25194426. [PMID: 32993187 PMCID: PMC7582295 DOI: 10.3390/molecules25194426] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/22/2020] [Accepted: 09/25/2020] [Indexed: 12/31/2022] Open
Abstract
Ischemia/reperfusion injury is a severe disorder associated with a high mortality. Several antioxidant and pharmacological properties of cashew nuts (Anacardium occidentale L.) and its metabolites from different countries have recently been described. It is a medicinal plant with important therapeutic effects. This study aimed to verify the effect of an oral administration of cashew nuts in a rat model of ischemia/reperfusion (I/R). Adult male rats were subjected to intestinal I/R injury by clamping the superior mesenteric artery for 30 min and then allowing animals to 1 h of reperfusion. Rats subjected to I/R of the gut showed a significant increase in different biochemical markers. In particular, we evaluated lipid peroxidation, tissue myeloperoxidase activity, protein carbonyl content, reactive oxygen species generation and decreased antioxidant enzyme activities. Western blot analysis showed the activation of the NRF2 and NF-kB pathways. Increased immunoreactivity to nitrotyrosine, PARP, P-selectin, and ICAM-1 was observed in the ileum of rats subjected to I/R. Administration of cashew nuts (100 mg/kg) significantly reduced the mortality rate, the fall in arterial blood pressure, and oxidative stress and restored the antioxidant enzyme activities by a mechanism involving both NRF2 and NF-kB pathways. Cashew nuts treatments reduced cytokines plasma levels, nitrotyrosine, and PARP expression as well as adhesion molecules expressions. Additionally, cashew nuts decreased the intestinal barrier dysfunction and mucosal damage, the translocation of toxins and bacteria, which leads to systemic inflammation and associated organs injuries in particular of liver and kidney. Our study demonstrates that cashew nuts administration exerts antioxidant and pharmacological protective effects in superior mesenteric artery occlusion–reperfusion shock.
Collapse
Affiliation(s)
- Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
| | - Marika Cordaro
- Department of Biomedical, Dental and Morphological and Functional Imaging University of Messina, Via Consolare Valeria, 98125 Messina, Italy;
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
| | - Alessio Filippo Peritore
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
| | - Enrico Gugliandolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
| | - Tiziana Genovese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
| | - Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
| | - Rosalia Crupi
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy;
| | - Antonella Smeriglio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
| | - Giuseppina Mandalari
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
- Correspondence: (D.I.); (S.C.); Tel.: +39-090-676-5208 (D.I. & S.C.)
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, Saint Louis, MO 63104, USA
- Correspondence: (D.I.); (S.C.); Tel.: +39-090-676-5208 (D.I. & S.C.)
| | - Rosanna Di Paola
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
| |
Collapse
|
34
|
Raftery AL, Tsantikos E, Harris NL, Hibbs ML. Links Between Inflammatory Bowel Disease and Chronic Obstructive Pulmonary Disease. Front Immunol 2020; 11:2144. [PMID: 33042125 PMCID: PMC7517908 DOI: 10.3389/fimmu.2020.02144] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/07/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammatory bowel disease (IBD) and chronic obstructive pulmonary disease (COPD) are chronic inflammatory diseases of the gastrointestinal and respiratory tracts, respectively. These mucosal tissues bear commonalities in embryology, structure and physiology. Inherent similarities in immune responses at the two sites, as well as overlapping environmental risk factors, help to explain the increase in prevalence of IBD amongst COPD patients. Over the past decade, a tremendous amount of research has been conducted to define the microbiological makeup of the intestine, known as the intestinal microbiota, and determine its contribution to health and disease. Intestinal microbial dysbiosis is now known to be associated with IBD where it impacts upon intestinal epithelial barrier integrity and leads to augmented immune responses and the perpetuation of chronic inflammation. While much less is known about the lung microbiota, like the intestine, it has its own distinct, diverse microflora, with dysbiosis being reported in respiratory disease settings such as COPD. Recent research has begun to delineate the interaction or crosstalk between the lung and the intestine and how this may influence, or be influenced by, the microbiota. It is now known that microbial products and metabolites can be transferred from the intestine to the lung via the bloodstream, providing a mechanism for communication. While recent studies indicate that intestinal microbiota can influence respiratory health, intestinal dysbiosis in COPD has not yet been described although it is anticipated since factors that lead to dysbiosis are similarly associated with COPD. This review will focus on the gut-lung axis in the context of IBD and COPD, highlighting the role of environmental and genetic factors and the impact of microbial dysbiosis on chronic inflammation in the intestinal tract and lung.
Collapse
Affiliation(s)
- April L Raftery
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Evelyn Tsantikos
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Nicola L Harris
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Margaret L Hibbs
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
35
|
Papoutsopoulou S, Satsangi J, Campbell BJ, Probert CS. Review article: impact of cigarette smoking on intestinal inflammation-direct and indirect mechanisms. Aliment Pharmacol Ther 2020; 51:1268-1285. [PMID: 32372449 DOI: 10.1111/apt.15774] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/13/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND The inflammatory bowel diseases, Crohn's disease and ulcerative colitis are related multifactorial diseases. Their pathogenesis is influenced by each individual's immune system, the environmental factors within exposome and genetic predisposition. Smoking habit is the single best-established environmental factor that influences disease phenotype, behaviour and response to therapy. AIM To assess current epidemiological, experimental and clinical evidence that may explain how smoking impacts on the pathogenesis of inflammatory bowel disease. METHODS A Medline search for 'cigarette smoking', in combination with terms including 'passive', 'second-hand', 'intestinal inflammation', 'Crohn's disease', 'ulcerative colitis', 'colitis'; 'intestinal epithelium', 'immune system', 'intestinal microbiota', 'tight junctions', 'mucus', 'goblet cells', 'Paneth cells', 'autophagy'; 'epigenetics', 'genes', 'DNA methylation', 'histones', 'short noncoding/long noncoding RNAs'; 'carbon monoxide/CO' and 'nitric oxide/NO' was performed. RESULTS Studies found evidence of direct and indirect effects of smoking on various parameters, including oxidative damage, impairment of intestinal barrier and immune cell function, epigenetic and microbiota composition changes, that contribute to the pathogenesis of inflammatory bowel disease. CONCLUSIONS Cigarette smoking promotes intestinal inflammation by affecting the function and interactions among intestinal epithelium, immune system and microbiota/microbiome.
Collapse
Affiliation(s)
- Stamatia Papoutsopoulou
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Jack Satsangi
- Translational Gastroenterology Unit, Nuffield Department of Medicine, Experimental Medicine Division, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Barry J Campbell
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Chris S Probert
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
36
|
MitoQ Modulates Lipopolysaccharide-Induced Intestinal Barrier Dysfunction via Regulating Nrf2 Signaling. Mediators Inflamm 2020; 2020:3276148. [PMID: 32351320 PMCID: PMC7171662 DOI: 10.1155/2020/3276148] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/18/2020] [Accepted: 02/26/2020] [Indexed: 01/12/2023] Open
Abstract
Background Gut barrier dysfunction with alterant mucosal permeability during sepsis is a challenge problem in clinical practice. Intestinal epithelial cells (IECs) are strongly involved in mucosal oxidative stress and inflammatory response. The current study aimed at investigating the effect of MitoQ, a mitochondrial targeted antioxidant, in the treatment of intestinal injury and its potential mechanism during sepsis. Methods 30 minutes before sepsis induction by lipopolysaccharide (LPS) treatment, mice were treated with MitoQ. Intestinal histopathology, mucosal permeability, inflammatory cytokines, and mucosal barrier proteins were evaluated in the present study. Results MitoQ pretreatment significantly decreased the levels of plasma diamine oxidase, D-lactate, and intestinal histological damage and markedly restored the levels of tight junction proteins (ZO-1 and occludin) following LPS challenge. Furthermore, MitoQ inhibited the LPS-induced intestinal oxidative stress and inflammatory response, evidenced by increased levels of intestinal superoxide dismutase and glutathione, and decreased levels of intestinal IL-1, IL-6, TNF-α, and nitric oxide levels. Mechanically, we found that MitoQ inhibited the oxidative stress via activating nuclear factor E2-related factor 2 (Nrf2) signaling pathway and its downstream antioxidant genes, including HO-1, NQO-1, and GCLM. Conclusions MitoQ exerts antioxidative and anti-inflammatory effects against sepsis-associated gut barrier injury by promoting Nrf2 signaling pathway.
Collapse
|
37
|
Vrablicova Z, Soltys K, Krajcovicova A, Stuchlikova K, Sturdik I, Koller T, Huorka M, Payer J, Killinger Z, Jackuliak P, Tkacik M, Stuchlik S, Sekac J, Hlavaty T. Impact of smoking cigarette on the mRNA expression of cytokines in mucosa of inflammatory bowel disease. Physiol Res 2019; 68:S183-S192. [PMID: 31842582 DOI: 10.33549/physiolres.934301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
It is well known that smoking is the risk factor in the development and clinical course of Crohn s disease (CD), but on the other hand, smoking is a protective factor against ulcerative colitis (UC). The pathways that are influenced by smoking in CD and UC are poorly understood. The aim of our study was to analyse the influence of smoking on the mRNA expression of cytokines in mucosa in patients with CD and UC. We performed a cross-sectional study. The cohort consisted of 86 IBD patients (48 CD patients and 38 UC patients) and took place at the IBD Centre at the University Hospital Bratislava-Ružinov. We took the demographic and clinical data of each patient, including information about their smoking habits. We performed a colonoscopy on each patient and took biopsies from both inflamed and non-inflamed sigma (CD, UC) and terminal ileum (CD). mRNA was extracted from mucosal biopsy samples for each cytokine and was normalized to a housekeeping gene (GAPDH). Finally, we compared the mRNA expression of target cytokines in the mucosa of smokers and non-smokers in IBD patients. Smokers with Crohn s disease have a significantly higher mRNA expression of pro-inflammatory cytokine TNF ? (p=0.003) in inflamed mucosa in sigma compared with non-smokers. In smokers with ulcerative colitis, we observed significantly higher mRNA expression of anti-inflammatory cytokine IL 10 (p=0.022) in non-inflamed mucosa of sigma. Similarly, smokers with UC have a significantly decreased mRNA expression of cytokine TLR 2 (p=0.024) and CCR1 (p=0.049) in non-inflamed mucosa of sigma. Based on our results, smoking has a positive influence on cessation and the clinical course of UC due to the stimulation of anti-inflammatory cytokine IL 10 in mucosa. On the other hand, smokers with CD have a higher expression of pro-inflammatory cytokine TNF ?, which could be associated with a worsening of the disease and response to therapy.
Collapse
Affiliation(s)
- Z Vrablicova
- 5th Department of Internal Medicine, Sub-department of Gastroenterology and Hepatology, Faculty of Medicine, Comenius University Bratislava, University Hospital Bratislava, Slovak Republic
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Song X, Li J, Wang Y, Zhou C, Zhang Z, Shen M, Xiang P, Zhang X, Zhao H, Yu L, Zuo L, Hu J. Clematichinenoside AR ameliorated spontaneous colitis in Il-10 -/- mice associated with improving the intestinal barrier function and abnormal immune responses. Life Sci 2019; 239:117021. [PMID: 31678552 DOI: 10.1016/j.lfs.2019.117021] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 10/21/2019] [Accepted: 10/26/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Clematichinenoside AR (AR) is a saponin extracted for traditional Chinese medicine with the effects of improving the expression of tight junction (TJ) proteins and mediating anti-inflammatory activities. However, its effect on Crohn's disease (CD) is still unknown. We aimed to investigate the impact of AR on CD-like colitis and determine the mechanism underlying its effects. METHODS Interleukin-10 gene knockout (Il-10-/-) mice (male, fifteen weeks old) with spontaneous colitis were allocated to the positive control and AR-treated (32 mg/kg AR administered every other day by gavage for 4 weeks) groups. Wild-type (WT) mice (male, fifteen weeks old) composed the negative control group. The effects of AR on intestinal barrier function and structure and T cell responses as well as the potential mechanisms underlying these effects were investigated. RESULTS AR treatment significantly improved spontaneous colitis in Il-10-/- mice as demonstrated by reductions in the inflammatory score, disease activity index (DAI) and levels of inflammatory factors. The effects of AR on colitis in Il-10-/- mice were related to protecting intestinal barrier function and maintaining immune system homeostasis (regulatory T cell (Treg)/T helper 17 (Th17) cell balance). The anticolitis effect of AR may partly act by downregulating PI3K/Akt signaling. CONCLUSIONS AR may have therapeutic potential for treating CD in humans.
Collapse
Affiliation(s)
- Xue Song
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China; Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Jing Li
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China; Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yan Wang
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China; Department of Clinical Medicine, Bengbu Medical College, Bengbu, China
| | - Changmin Zhou
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China; Department of Clinical Medicine, Bengbu Medical College, Bengbu, China
| | - Zhichao Zhang
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China; Department of Clinical Medicine, Bengbu Medical College, Bengbu, China
| | - Mengdi Shen
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China; Department of Clinical Medicine, Bengbu Medical College, Bengbu, China
| | - Ping Xiang
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Xiaofeng Zhang
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China; Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Hao Zhao
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Liang Yu
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Lugen Zuo
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China; Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Jianguo Hu
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China; Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China.
| |
Collapse
|
39
|
Berkowitz L, Pardo-Roa C, Salazar GA, Salazar-Echegarai F, Miranda JP, Ramírez G, Chávez JL, Kalergis AM, Bueno SM, Álvarez-Lobos M. Mucosal Exposure to Cigarette Components Induces Intestinal Inflammation and Alters Antimicrobial Response in Mice. Front Immunol 2019; 10:2289. [PMID: 31608070 PMCID: PMC6773925 DOI: 10.3389/fimmu.2019.02289] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 09/10/2019] [Indexed: 12/15/2022] Open
Abstract
The main environmental risk factor associated with the development of Crohn's disease (CD) is cigarette smoking. Although the mechanism is still unknown, some studies have shown that cigarette exposure affects the intestinal barrier of the small bowel. Among the factors that may be involved in this process are Paneth cells. These specialized epithelial cells are located into the small intestine, and they are able to secrete antimicrobial peptides, having an essential role in the control of the growth of microorganisms. Alterations in its function are associated with inflammatory processes, such as CD. To study how cigarette components impact ileum homeostasis and Paneth cells integrity, we used intragastric administration of cigarette smoke condensate (CSC) in mice. Our results showed that inflammation was triggered after mucosal exposure of CSC, which induced particular alterations in Paneth cells granules, antimicrobial peptide production, and a reduction of bactericidal capacity. In fact, exposure to CSC generated an imbalance in the fecal bacterial population and increased the susceptibility of mice to develop ileal damage in response to bacterial infection. Moreover, our results obtained in mice unable to produce interleukin 10 (IL-10−/− mice) suggest that CSC treatment can induce a symptomatic enterocolitis with a pathological inflammation in genetically susceptible individuals.
Collapse
Affiliation(s)
- Loni Berkowitz
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina Pardo-Roa
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Geraldyne A Salazar
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco Salazar-Echegarai
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José P Miranda
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gigliola Ramírez
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José L Chávez
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Manuel Álvarez-Lobos
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
40
|
姜 从, 朱 平, 时 依, 项 武, 葛 思, 张 宗, 左 芦. [Protective effect of procyanidin B2 on intestinal barrier and against enteritis in a mouse model of trinitrobenzene sulphonic acid-induced colitis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:778-783. [PMID: 31340909 PMCID: PMC6765556 DOI: 10.12122/j.issn.1673-4254.2019.07.05] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the protective effect of procyanidin B2 (PCB2) on the intestinal barrier and against enteritis in mice with trinitrobenzene sulphonic acid (TNBS)-induced colitis and explore the possible mechanism. METHODS A mouse model of TNBS-induced colitis was established in male Balb/c mice aged 6-8 weeks. The successfully established mouse models were randomly divided into PCB2 treatment group (n=10) and model group (n=10) and were treated with daily intragastric administration of PCB2 (100 mg/kg, 0.2 mL) and 0.2 mL normal saline, respectively. After 4 weeks, the disease symptoms, intestinal inflammation, intestinal mucosal cell barrier function and the changes in PI3K/AKT signaling were evaluated using HE staining, immunofluorescence assay and Western blotting. RESULTS The disease activity index of the mice was significantly lower and the mean body weight was significantly greater in PCB2 group than in the model group in the 3rd and 4th weeks of intervention (P < 0.05). The levels of colonic inflammation and intestinal mucosal inflammatory mediators IL-1β and TNF-α were significantly lower while IL-10 was significantly higher in PCB2 group than in the model group (P < 0.05). Compared with those in the model group, the mice in PCB2 treatment group showed a significantly lower positive rate of bacterial translocation in the mesenteric lymph nodes and a lower thiocyanate-dextran permeability of the intestinal mucosa (P < 0.05). Western blotting showed that PCB2 treatment significantly increased the expressions of claudin-1 and ZO-1 (P < 0.05) and significantly lowered the expression levels of p-PI3K and p-AKT in the intestinal mucosa as compared with those in the model group (P < 0.05). CONCLUSIONS PCB2 suppresses intestinal inflammation and protects intestinal mucosal functions and structural integrity by inhibiting intestinal PI3K/AKT signaling pathway, suggesting the potential of PCB2 as a new drug for Crohn's disease.
Collapse
Affiliation(s)
- 从桥 姜
- />蚌埠医学院第一附属医院胃肠外科,安徽 蚌埠 233004Department of Gastroenterology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 平胜 朱
- />蚌埠医学院第一附属医院胃肠外科,安徽 蚌埠 233004Department of Gastroenterology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 依 时
- />蚌埠医学院第一附属医院胃肠外科,安徽 蚌埠 233004Department of Gastroenterology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 武军 项
- />蚌埠医学院第一附属医院胃肠外科,安徽 蚌埠 233004Department of Gastroenterology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 思堂 葛
- />蚌埠医学院第一附属医院胃肠外科,安徽 蚌埠 233004Department of Gastroenterology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 宗兵 张
- />蚌埠医学院第一附属医院胃肠外科,安徽 蚌埠 233004Department of Gastroenterology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 芦根 左
- />蚌埠医学院第一附属医院胃肠外科,安徽 蚌埠 233004Department of Gastroenterology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| |
Collapse
|
41
|
Zuo L, Li J, Ge S, Ge Y, Shen M, Wang Y, Zhou C, Wu R, Hu J. Bryostatin-1 ameliorated experimental colitis in Il-10 -/- Mice by protecting the intestinal barrier and limiting immune dysfunction. J Cell Mol Med 2019; 23:5588-5599. [PMID: 31251471 PMCID: PMC6652299 DOI: 10.1111/jcmm.14457] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 12/13/2022] Open
Abstract
Bryostatin‐1 (Bry‐1) has been proven to be effective and safe in clinical trials of a variety of immune‐related diseases. However, little is known about its effect on Crohn's disease (CD). We aimed to investigate the impact of Bry‐1 on CD‐like colitis and determine the mechanism underlying this effect. In the present study, 15‐week‐old male Il‐10−/− mice with spontaneous colitis were divided into positive control and Bry‐1‐treated (Bry‐1, 30 μg/kg every other day, injected intraperitoneally for 4 weeks) groups. Age‐matched, male wild‐type (WT) mice were used as a negative control. The effects of Bry‐1 on colitis, intestinal barrier function and T cell responses as well as the potential regulatory mechanisms were evaluated. We found that the systemic delivery of Bry‐1 significantly ameliorated colitis in Il‐10−/− mice, as demonstrated by decreases in the disease activity index (DAI), inflammatory score and proinflammatory mediator levels. The protective effects of Bry‐1 on CD‐like colitis included the maintenance of intestinal barrier integrity and the helper T cell (Th)/regulatory T cell (Treg) balance. These effects of Bry‐1 may act in part through nuclear factor erythroid 2‐related factor 2 (Nrf2) signalling activation and STAT3/4 signalling inhibition. The protective effect of Bry‐1 on CD‐like colitis suggests Bry‐1 has therapeutic potential in human CD, particularly given the established clinical safety of Bry‐1.
Collapse
Affiliation(s)
- Lugen Zuo
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Jing Li
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China.,Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Sitang Ge
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Yuanyuan Ge
- Department of Colorectal Surgery, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Mengdi Shen
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China.,Department of Clinical medicine, Bengbu medical college, Bengbu, China
| | - Yan Wang
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China.,Department of Clinical medicine, Bengbu medical college, Bengbu, China
| | - Changmin Zhou
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China.,Department of Clinical medicine, Bengbu medical college, Bengbu, China
| | - Rong Wu
- Department of General Surgery, Zhongda Hospital, Southeast University, Nanjing, China
| | - Jianguo Hu
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China.,Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|
42
|
Li J, Zuo L, Tian Y, He Y, Zhang Z, Guo P, Ge Y, Hu J. Spontaneous colitis in IL-10-deficient mice was ameliorated via inhibiting glutaminase1. J Cell Mol Med 2019; 23:5632-5641. [PMID: 31211512 PMCID: PMC6653008 DOI: 10.1111/jcmm.14471] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 04/09/2019] [Accepted: 04/26/2019] [Indexed: 12/30/2022] Open
Abstract
Immunity imbalance and barrier damage in the intestinal mucosa are the main pathogenic factors of Crohn's disease (CD). Bis‐2‐(5‐phenylacetamido‐1,2,4‐thiadiazol‐2‐yl) ethyl sulfide (BPTES) is a glutaminase 1 (Gls1) inhibitor with the dual functions of increasing glutamine levels and immune regulation. In this study, we focused on the role of BPTES in CD‐like enteritis and the possible mechanisms. We found that Gls1 expression was significantly increased in CD intestinal tissue compared with control tissue. Bis‐2‐(5‐phenylacetamido‐1,2,4‐thiadiazol‐2‐yl) ethyl sulfide treatment significantly ameliorated chronic colitis in the IL‐10−/−, as manifested by decreased disease activity index, body weight change, histological inflammatory degree and inflammatory cytokine expression. Bis‐2‐(5‐phenylacetamido‐1,2,4‐thiadiazol‐2‐yl) ethyl sulfide treatment exerted protective effects on CD that were associated with the maintenance of intestinal barrier integrity and the Th/Treg balance. Bis‐2‐(5‐phenylacetamido‐1,2,4‐thiadiazol‐2‐yl) ethyl sulfide treatment may act in part through TCR‐mediated mammalian target of rapamycin complex 1 (mTORC1) signalling activation. In conclusion, inhibition of Gls1 expression attenuated chronic colitis by maintaining intestinal barrier integrity and the Th/Treg balance, thereby ameliorating CD‐like colitis.
Collapse
Affiliation(s)
- Jing Li
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Lugen Zuo
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China.,Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Yun Tian
- Department of Oncology, Shanghai Dermatology Hospital, Tongji University, Shanghai, China.,Tongji University Cancer Center, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Yifan He
- Clinical Medicine of Bengbu Medical College, Bengbu, Anhui, China
| | - Zhichao Zhang
- Clinical Medicine of Bengbu Medical College, Bengbu, Anhui, China
| | - Pu Guo
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Yuanyuan Ge
- Department of Colorectal Surgery, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianguo Hu
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China.,Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| |
Collapse
|
43
|
Collagen peptides administration in early enteral nutrition intervention attenuates burn-induced intestinal barrier disruption: Effects on tight junction structure. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.02.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
44
|
Hu Q, Ren H, Li G, Wang D, Zhou Q, Wu J, Zheng J, Huang J, Slade DA, Wu X, Ren J. STING-mediated intestinal barrier dysfunction contributes to lethal sepsis. EBioMedicine 2019; 41:497-508. [PMID: 30878597 PMCID: PMC6443583 DOI: 10.1016/j.ebiom.2019.02.055] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/18/2019] [Accepted: 02/26/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Gut integrity is compromised in abdominal sepsis with increased cellular apoptosis and altered barrier permeability. Intestinal epithelial cells (IEC) form a physiochemical barrier that separates the intestinal lumen from the host's internal milieu and is strongly involved in the mucosal inflammatory response and immune response. Recent research indicates the involvement of the stimulator of interferons genes (STING) pathway in uncontrolled inflammation and gut mucosal immune response. METHODS We investigated the role of STING signaling in sepsis and intestinal barrier function using intestinal biopsies from human patients with abdominal sepsis and with an established model of abdominal sepsis in mice. FINDINGS In human abdominal sepsis, STING expression was elevated in peripheral blood mononuclear cells and intestinal biopsies compared with healthy controls, and the degree of STING expression in the human intestinal lamina propria correlated with the intestinal inflammation in septic patients. Moreover, elevated STING expression was associated with high levels of serum intestinal fatty acid binding protein that served as a marker of enterocyte damage. In mice, the intestinal STING signaling pathway was markedly activated following the induction of sepsis induced by cecal ligation perforation (CLP). STING knockout mice showed an alleviated inflammatory response, attenuated gut permeability, and decreased bacterial translocation. Whereas mice treated with a STING agonist (DMXAA) following CLP developed greater intestinal apoptosis and a more severe systemic inflammatory response. We demonstrated that mitochondrial DNA (mtDNA) was released during sepsis, inducing the intestinal inflammatory response through activating the STING pathway. We finally investigated DNase I administration at 5 hours post CLP surgery, showing that it reduced systemic mtDNA and inflammatory cytokines levels, organ damage, and bacterial translocation, suggesting that inhibition of mtDNA-STING signaling pathway protects against CLP-induced intestinal barrier dysfunction. INTERPRETATION Our results indicate that the STING signaling pathway can contribute to lethal sepsis by promoting IEC apoptosis and through disrupting the intestinal barrier. Our findings suggest that regulation of the mtDNA-STING pathway may be a promising therapeutic strategy to promote mucosal healing and protect the intestinal barrier in septic patients. FUND: National Natural Science Foundation of China.
Collapse
Affiliation(s)
- Qiongyuan Hu
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China; Medical School of Nanjing University, Nanjing, China
| | - Huajian Ren
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Guanwei Li
- Department of colorectal and anal surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Dingyu Wang
- Medical School of Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Collaborative Innovation Center of Genetics and Development, Model Animal Research Center, Nanjing, China
| | - Quan Zhou
- Medical School of Nanjing University, Nanjing, China
| | - Jie Wu
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China; Medical School of Nanjing University, Nanjing, China
| | - Jiashuo Zheng
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China; Medical School of Nanjing University, Nanjing, China
| | - Jinjian Huang
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Dominic A Slade
- Medical School of Nanjing University, Nanjing, China; Department of Surgery, Salford Royal NHS Foundation Trust, Stott Lane, Salford, United Kingdom.
| | - Xiuwen Wu
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Jianan Ren
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
45
|
Shen W, Li Y, Zou Y, Cao L, Cai X, Gong J, Xu Y, Zhu W. Mesenteric Adipose Tissue Alterations in Crohn's Disease Are Associated With the Lymphatic System. Inflamm Bowel Dis 2019; 25:283-293. [PMID: 30295909 DOI: 10.1093/ibd/izy306] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Mesenteric fat wrapping and thickening are typical characteristics of Crohn's disease (CD). The purpose of this study was to explore the cause of mesenteric adipose hypertrophy and analyze the role of lymphatic vessels in mesenteric adipose tissue in CD. METHODS Twenty-three CD patients who underwent ileocolonic resection were included. In CD patients, specimens were obtained from hypertrophic mesenteric adipose tissue (htMAT) next to the diseased ileum. The mesenteric lymphatic vessels in mesenteric adipose tissue were separated under stereoscope microscope. Transmission electron microscopy and immunofluorescence were used to observe the structure of mesenteric lymphatic vessels. The NF-κB signaling pathway in mesenteric adipose tissue was detected in CD specimens using Western blotting. RESULTS Electron microscopy showed that the structure of mesenteric lymphatic vessel was discontinuous, and the microstructure of lymphatic endothelial cells appeared ruptured and incomplete. Through an immunofluorescence technique, we found that the surface of lymphatic endothelial cells lacked tight junction protein staining in CD. Also, the expression of claudin-1, occludin, and ZO-1 in the mesenteric lymphatic vessel of htMAT was significantly lower than that of control. These results indicated that the structure of the mesenteric lymphatic vessel in htMAT was mispatterned and ruptured, which could lead to lymph leakage. Leaky lymph factors could stimulate adipose tissue to proliferate. Antigens that leaked into the mesenteric adipose tissue could effectively elicit an immune response. The levels of cytokines (TNF-a, IL-1β, IL-6) was increased in the htMAT of CD patients by activated NF-κB signaling pathway. CONCLUSIONS Our findings demonstrated that the hypertrophy of mesenteric adipose tissue may result from mispatterned and ruptured lymphatic vessels. Alteration of mesenteric adipose tissue was associated with activated NF-κB signaling pathway. This study enhances support for elucidating the importance of mesenteric lymphatic vessels and adipose tissue in CD.
Collapse
Affiliation(s)
- Weisong Shen
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yi Li
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yujie Zou
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, China
| | - Lei Cao
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xingchen Cai
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jianfeng Gong
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yihan Xu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Weiming Zhu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
46
|
Shen W, Li Y, Cao L, Cai X, Ge Y, Zhu W. Decreased Expression of Prox1 Is Associated With Postoperative Recurrence in Crohn's Disease. J Crohns Colitis 2018; 12:1210-1218. [PMID: 29947772 DOI: 10.1093/ecco-jcc/jjy091] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Prox1 is a transcription factor necessary for lymphangiogenesis and lymphatic function. The aim of the study was to assess the correlation between the expression of Prox1 and postoperative recurrence in Crohn's disease [CD]. METHODS Forty CD patients who underwent ileo-colonic resection were included. Expression levels of Prox1 and D2-40 were detected using immunohistochemistry. Expression levels of Prox1, VEGFR3, and VEGFC protein were also detected in fresh CD specimens using western blotting and quantitative polymerase chain reaction [Q-PCR]. Endoscopic recurrence was used as the endpoint. Patients comprised two groups: endoscopic recurrence [Group R+] and no endoscopic recurrence [Group R-]. RESULTS Prox1 protein expression was significantly higher in CD than in normal tissues [p <0.05], as detected using both immunohistochemistry and western blotting. Analysis of inter-relationships revealed significant correlation between Prox1 expression and lymphatic vessel density [p <0.001, r = 0.823]. There was also significant correlation between Prox1 expression and the visceral fat area [VFA] [p = 0.002, r = -0.469]. The Group R- patients had significantly higher Prox1 expression than the Group R+ patients [21.08 ± 1.61 versus 15.64 ± 1.17, p = 0.011]. Also, the lymphatic vessel density value was lower in Group R+ than in Group R- patients [6.02 ± 0.39 versus 8.13 ± 0.59, p = 0.004]. Moreover, there was a significant difference in the VFA between Group R- and Group R+ patients [64.43 ± 7.76 versus 90.44 ± 6.11, p = 0.016]. In addition to Prox1, VEGFC/VEGFR3 was found to increase, which was further confirmed using Q-PCR. CONCLUSIONS Prox1 expression could be useful as a protective factor against recurrence in CD patients. The therapeutic role of Prox1 may lead to improved treatments.
Collapse
Affiliation(s)
- Weisong Shen
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yi Li
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lei Cao
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xingchen Cai
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yuanyuan Ge
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Weiming Zhu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
47
|
Smoking Status at Diagnosis and Subsequent Smoking Cessation: Associations With Corticosteroid Use and Intestinal Resection in Crohn's Disease. Am J Gastroenterol 2018; 113:1689-1700. [PMID: 30323269 DOI: 10.1038/s41395-018-0273-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/31/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND The impact of smoking at diagnosis and subsequent smoking cessation on clinical outcomes in Crohn's disease (CD) has not been evaluated in a population-based cohort. METHODS Using a nationally representative clinical research database, we identified incident cases of CD between 2005 and 2014. We compared the following outcomes: overall corticosteroid (CS) use; flares requiring CS; CS dependency and intestinal surgery between smokers and non-smokers at time of CD diagnosis. Differences in these outcomes were also compared between persistent smokers and smokers who quit within 2 years of diagnosis. RESULTS We identified 3553 patients with a new CD diagnosis over the study period of whom 1121 (32%) were smokers. Smokers at CD diagnosis had significantly higher CS-use (56 versus 47%, p < 0.0001), proportionally more CS flares (>1 CS flare/year: 9 versus 6%, p < 0.0001), and higher CS dependency (27 versus 21%, p < 0.0001) than non-smokers. Regression analysis identified smoking at diagnosis to be associated with a higher risk of intestinal surgery (HR 1.64, 95% CI 1.16-2.52). There was a significantly higher proportion of 'quitters' who remained steroid-free through follow-up in comparison to 'persistent smokers' (45.4 versus 37.5%, respectively, p = 0.02). 'Quitters' also had lower rates of CS dependency compared to 'persistent smokers' (24 versus 33%, p = 0.008). CONCLUSIONS Smokers at CD diagnosis have higher CS-use, CS dependency and higher risk of intestinal surgery. Quitting smoking appears to have beneficial effects on disease related outcomes, including reducing CS dependency highlighting the importance of offering early smoking cessation support.
Collapse
|
48
|
Hu Q, Ren H, Ren J, Liu Q, Wu J, Wu X, Li G, Wang G, Gu G, Guo K, Hong Z, Liu S, Li J. Released Mitochondrial DNA Following Intestinal Ischemia Reperfusion Induces the Inflammatory Response and Gut Barrier Dysfunction. Sci Rep 2018; 8:7350. [PMID: 29743484 PMCID: PMC5943336 DOI: 10.1038/s41598-018-25387-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 03/06/2018] [Indexed: 01/10/2023] Open
Abstract
Ischemia-reperfusion (I/R) injury is a challenging clinical problem, especially injuries involving the gastrointestinal tract. Mitochondrial DNA (mtDNA) is released upon cell death and stress, and can induce the inflammatory response. We aimed to investigate the role of mtDNA in the pathogenesis of intestinal I/R. Intestinal I/R model was established with clamping of the superior mesenteric artery, and IEC-6 cells were incubated under hypoxia/reoxygenation (H/R) conditions to simulate I/R injury. Using in vitro models, H/R up-regulated oxidative stress, disrupted mitochondrial activity and the mitochondrial membrane potential, induced apoptosis and elevated the mtDNA levels in the supernatant of intestinal epithelial cells, and the co-culture of mtDNA with human primary dendritic cells significantly elevated TLR9-MyD88 expression and enhanced the production of inflammatory cytokines and chemokines. MtDNA was also released in a mouse model of intestinal I/R and was associated with the increased secretion of inflammatory cytokines and increased gut barrier injury compared with that of the sham group. We concluded that mtDNA contributes to I/R injury and may serve as a biomarker of intestinal I/R. We further suggest that oxidized mtDNA originated from IECs during intestinal I/R exacerbates the acute proinflammatory process by eliciting the production of proinflammatory cytokines and chemokines.
Collapse
Affiliation(s)
- Qiongyuan Hu
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Huajian Ren
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
- Jinling college, Nanjing Medical University, Nanjing, China
| | - Jianan Ren
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Qinjie Liu
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
- Jinling college, Nanjing Medical University, Nanjing, China
| | - Jie Wu
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiuwen Wu
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Guanwei Li
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Gefei Wang
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Guosheng Gu
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Kun Guo
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zhiwu Hong
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Song Liu
- Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jieshou Li
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
49
|
Hu Q, Ren J, Li G, Wu J, Wu X, Wang G, Gu G, Ren H, Hong Z, Li J. The mitochondrially targeted antioxidant MitoQ protects the intestinal barrier by ameliorating mitochondrial DNA damage via the Nrf2/ARE signaling pathway. Cell Death Dis 2018; 9:403. [PMID: 29540694 PMCID: PMC5851994 DOI: 10.1038/s41419-018-0436-x] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 02/06/2018] [Accepted: 02/13/2018] [Indexed: 12/11/2022]
Abstract
Disruption of the mucosal barrier following intestinal ischemia reperfusion (I/R) is life threatening in clinical practice. Mitochondrial dysfunction and oxidative stress significantly contribute to the early phase of I/R injury and amplify the inflammatory response. MitoQ is a mitochondrially targeted antioxidant that exerts protective effects following I/R injury. In the present study, we aimed to determine whether and how MitoQ protects intestinal epithelial cells (IECs) from I/R injury. In both in vivo and in vitro studies, we found that MitoQ pretreatment downregulated I/R-induced oxidative stress and stabilized the intestinal barrier, as evidenced by MitoQ-treated I/R mice exhibiting attenuated intestinal hyperpermeability, inflammatory response, epithelial apoptosis, and tight junction damage compared to controls. Mechanistically, I/R elevated mitochondrial 8-hydroxyguanine content, reduced mitochondrial DNA (mtDNA) copy number and mRNA transcription levels, and induced mitochondrial disruption in IECs. However, MitoQ pretreatment dramatically inhibited these deleterious effects. mtDNA depletion alone was sufficient to induce apoptosis and mitochondrial dysfunction of IECs. Mitochondrial transcription factor A (TFAM), a key activator of mitochondrial transcription, was significantly reduced during I/R injury, a phenomenon that was prevented by MitoQ treatment. Furthermore, we observed that thee protective properties of MitoQ were affected by upregulation of cellular antioxidant genes, including HO-1, NQO-1, and γ-GCLC. Transfection with Nrf2 siRNA in IECs exposed to hypoxia/reperfusion conditions partially blocked the effects of MitoQ on mtDNA damage and mitochondrial oxidative stress. In conclusion, our data suggest that MitoQ exerts protective effect on I/R-induced intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Qiongyuan Hu
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
- Lab for Trauma and Surgical Infection, Nanjing, China
| | - Jianan Ren
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
- Lab for Trauma and Surgical Infection, Nanjing, China.
| | - Guanwei Li
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jie Wu
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
- Lab for Trauma and Surgical Infection, Nanjing, China
| | - Xiuwen Wu
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
- Lab for Trauma and Surgical Infection, Nanjing, China.
| | - Gefei Wang
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Guosheng Gu
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Huajian Ren
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
- Medical school of Nanjing Medical University, Nanjing, China
| | - Zhiwu Hong
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jieshou Li
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
50
|
Berkowitz L, Schultz BM, Salazar GA, Pardo-Roa C, Sebastián VP, Álvarez-Lobos MM, Bueno SM. Impact of Cigarette Smoking on the Gastrointestinal Tract Inflammation: Opposing Effects in Crohn's Disease and Ulcerative Colitis. Front Immunol 2018; 9:74. [PMID: 29441064 PMCID: PMC5797634 DOI: 10.3389/fimmu.2018.00074] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 01/11/2018] [Indexed: 01/06/2023] Open
Abstract
Cigarette smoking is a major risk factor for gastrointestinal disorders, such as peptic ulcer, Crohn’s disease (CD), and several cancers. The mechanisms proposed to explain the role of smoking in these disorders include mucosal damage, changes in gut irrigation, and impaired mucosal immune response. Paradoxically, cigarette smoking is a protective factor for the development and progression of ulcerative colitis (UC). UC and CD represent the two most important conditions of inflammatory bowel diseases, and share several clinical features. The opposite effects of smoking on these two conditions have been a topic of great interest in the last 30 years, and has not yet been clarified. In this review, we summarize the most important and well-understood effects of smoking in the gastrointestinal tract; and particularly, in intestinal inflammation, discussing available studies that have addressed the causes that would explain the opposite effects of smoking in CD and UC.
Collapse
Affiliation(s)
- Loni Berkowitz
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bárbara M Schultz
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Geraldyne A Salazar
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina Pardo-Roa
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Valentina P Sebastián
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Manuel M Álvarez-Lobos
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|