1
|
Wang S, Hong Y, Li Y, Zhang Z, Han J, Yang Z, Yang Y, Ma Z, Wang Q. Ferulic Acid Inhibits Arsenic-Induced Colon Injury by Improving Intestinal Barrier Function. ENVIRONMENTAL TOXICOLOGY 2024; 39:4821-4831. [PMID: 38881217 DOI: 10.1002/tox.24360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/13/2024] [Accepted: 05/25/2024] [Indexed: 06/18/2024]
Abstract
The prolonged exposure to arsenic results in intestinal barrier dysfunction, which is strongly concerned with detrimental processes such as oxidative stress and the inflammatory response. Ferulic acid (FA), as a phenolic acid, possesses the capability to mitigate arsenic-induced liver damage and cardiotoxic effects dependent on inhibition of oxidative stress and inflammatory responses. FA can mitigate testicular tissue damage and alveolar epithelial dysfunction, the mechanism of which may rely on nuclear factor erythroid 2-related factor 2/heme oxygenase 1 (Nrf2/HO-1) activation and nuclear factor-kappa B (NF-κB) pathway blocking. Based on the antioxidant and anti-inflammatory properties of FA, we speculated that FA might have the potential to inhibit arsenic-induced intestinal damage. To confirm this scientific hypothesis, mice exposed to sodium arsenite were treated with FA to observe colonic histopathology and TJ protein levels, and oxidative stress and TJ protein levels in Caco-2 cells exposed to sodium arsenite were assessed after FA intervention. In addition, molecular levels of NF-κB and Nrf2/HO-1 pathway in colon and Caco-2 cells were also detected. As shown in our data, FA inhibited arsenic-induced colon injury, which was reflected in the improvement of mucosal integrity, the decrease of down-regulated expression of tight junction (TJ) proteins (Claudin-1, Occludin, and ZO-1) and the inhibition of oxidative stress. Similarly, treatment with FA attenuated the inhibitory effect of arsenic on TJ protein expression in Caco-2 cells. In addition to suppressing the activation of NF-κB pathway, FA retrieved the activation of Nrf2/HO-1 pathway in colon and intestinal epithelial cells induced by arsenic. In summary, our findings propose that FA has the potential to mitigate arsenic-induced intestinal damage by preserving the integrity of intestinal epithelial TJs and suppressing oxidative stress. These results lay the groundwork for the potential use of FA in treating colon injuries caused by arsenic.
Collapse
Affiliation(s)
- Shumin Wang
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Yan Hong
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Yuxiu Li
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Zhenfen Zhang
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Jing Han
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Zhe Yang
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Yanping Yang
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Zhaolei Ma
- Department of Geriatrics, The Affiliated Hospital of Guizhou Medical University, Guiyang, People's Republic of China
| | - Qi Wang
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| |
Collapse
|
2
|
Zeng L, Wang Y, Shen J, Wei X, Wu Y, Chi X, Zheng X, Yu X, Shi Y, Liu W. TIPE2 aggravates experimental colitis and disrupts intestinal epithelial barrier integrity by activating JAK2/STAT3/SOCS3 signal pathway. Exp Cell Res 2024; 443:114287. [PMID: 39426612 DOI: 10.1016/j.yexcr.2024.114287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/07/2024] [Accepted: 10/16/2024] [Indexed: 10/21/2024]
Abstract
Ulcerative colitis (UC) is a chronic relapsing and progressive inflammatory disease of the colon. TIPE2 is a negative regulator of innate and adaptive immunity that maintains immune homeostasis. We found that TIPE2 was highly expressed in mucosa of mice with colitis. However, the role of TIPE2 in colitis remains unclear. We induced colitis in mice with dextran sulfate sodium (DSS) and treated them with TIPE2, and investigated the inflammatory activity of the colon in vivo by cytokines detection and histopathological analyses. We also measured inflammatory alteration and tight junctions induced by DSS in vitro. The results demonstrated that administration of TIPE2 promoted the severity of colitis in mice and human colon epithelial cells. Furthermore, TIPE2 aggravated intestinal epithelial barrier dysfunction by decreasing the expression of the tight junction proteins Occludin, Claudin-1 and ZO-1. In addition, TIPE2 exacerbated intestinal inflammatory response by inhibiting the expression of SOCS3, remarkably activating JAK2/STAT3 signaling pathway, and increasing the translocation of phosphorylated STAT3 into the nucleus. Silencing of TIPE2 attenuated the DSS-induced activation of JAK2/STAT3, thereby rescuing epithelial inflammatory injury and restoring barrier dysfunction. These results indicate that TIPE2 augments experimental colitis and disrupted the integrity of the intestinal epithelial barrier by activating the JAK2/STAT3/SOCS3 signaling pathway.
Collapse
Affiliation(s)
- Lingli Zeng
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Endoscopy Center, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Yuping Wang
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Endoscopy Center, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Jiaxin Shen
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Endoscopy Center, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xujin Wei
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Endoscopy Center, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Yilong Wu
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Endoscopy Center, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xintong Chi
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Endoscopy Center, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xueyan Zheng
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Department of Gastroenterology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Xing Yu
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Department of Gastroenterology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Ying Shi
- Department of Gastroenterology, The First Afiiliated Hospital of Jinan University, Guangzhou, Guangdong, China.
| | - Wenming Liu
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Endoscopy Center, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
3
|
Zhu H, Hou P, Chu F, Li X, Zhang W, Sun X, Liu Y, Zhao G, Gao Y, He DC, Wang H, He H. PBLD promotes IRF3 mediated the type I interferon (IFN-I) response and apoptosis to inhibit viral replication. Cell Death Dis 2024; 15:727. [PMID: 39362857 PMCID: PMC11450232 DOI: 10.1038/s41419-024-07083-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 09/07/2024] [Accepted: 09/16/2024] [Indexed: 10/05/2024]
Abstract
Recent studies have implicated the phenazine biosynthesis-like domain-containing protein (PBLD) in the negative regulation of the development and progression of various cancers. However, its function in viral infection remains unknown. In this study, we found that PBLD plays important roles in multiple virus infections including BPIV3, SeV, VSV, and HSV-1. Our study revealed that PBLD enhances the expression of type I interferon (IFN-I) and ISGs through interferon regulatory factor 3 (IRF3). Further study indicated that PBLD promotes transcriptional phosphorylation of IRF3 (S385/386), thereby facilitating virus-induced IFN-I production. Interestingly, PBLD mediates virus-triggered mitochondrial apoptosis through its dependence on IRF3 (K313/315). Mechanistically, PBLD facilitated virus-induced apoptosis by recruiting the Puma protein to the mitochondria via IRF3. Additionally, we performed mutational analyses of IRF3, showing that its loss of either transcriptional or apoptotic function markedly increased viral replication. Moreover, macrophages with PBLD deficiency during viral infection exhibited decreased the IFN-I and ISGs expression, exacerbating viral infection. Importantly, mice deficient in PBLD exhibited increased viral replication and susceptibility to SeV infection, leading to decreased survival. Notably, Cedrelone, a chemical activator of PBLD, has the ability to reduce SeV replication. Collectively, we first discovered the new function of PBLD in viral infection, broadening our understanding of potential therapeutic targets and offering new insights for antiviral drug development.
Collapse
Affiliation(s)
- Hongchao Zhu
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Peili Hou
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
| | - Fengyun Chu
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Xingyu Li
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Wenjia Zhang
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Xiaonan Sun
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Yu Liu
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Guimin Zhao
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Yuwei Gao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Daniel Chang He
- The College of Arts and Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hongmei Wang
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China.
| | - Hongbin He
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China.
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China.
| |
Collapse
|
4
|
Xu Y, Ou J, Zhang C, Chen J, Chen J, Li A, Huang B, Zhao X. Rapamycin promotes the intestinal barrier repair in ulcerative colitis via the mTOR/PBLD/AMOT signaling pathway. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167287. [PMID: 38862095 DOI: 10.1016/j.bbadis.2024.167287] [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: 01/13/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/13/2024]
Abstract
Intestinal barrier dysfunction characterized by the functional loss of the intestinal epithelium's tight junction (TJ) barrier is a key factor in the pathogenesis of ulcerative colitis (UC). Although rapamycin, an mTOR (mechanistic target of rapamycin) inhibitor, has shown promise in inducing clinical remission and mucosal healing in inflammatory bowel disease, its underlying mechanism remains elusive. Thus, this study investigated the role of the mTOR pathway in regulating the intestinal barrier. To investigate the molecular mechanism regulating the intestinal barrier, specific intestinal epithelial phenazine biosynthesis-like domain-containing protein (PBLD)-deficient (PBLDIEC-/-) mice and control wild-type (WT) mice were intraperitoneally injected with rapamycin or MHY1485. To determine the relevance of the findings for UC, we analyzed transcriptome data and single-cell expression profiles from public databases and intestinal mucosal tissues obtained from patients with active UC or colon cancer. We observed that mTOR activation in the intestinal epithelium of patients with active UC. Moreover, in vivo, rapamycin markedly increased the expressions of PBLD and TJ proteins and reduced intestinal inflammation in mice with dextran sulfate sodium-induced enteritis. However, the therapeutic efficacy of rapamycin was notably reduced in PBLDIEC-/- mice. In vitro, rapamycin influenced PBLD expression by modulating the nuclear transcription of transcription factor EB (TFEB). Angiomotin (AMOT) could directly bind to PBLD, and rapamycin could not effectively increase the expression of TJ proteins after the knockdown of PBLD or AMOT. In summary, the administration of rapamycin is a potential treatment for UC, and targeting the mTOR/PBLD/AMOT axis is a potential novel approach for UC treatment.
Collapse
Affiliation(s)
- Yan Xu
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China
| | - Jinyuan Ou
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China
| | - Chuhong Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China
| | - Jiayue Chen
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China
| | - Junsheng Chen
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China
| | - Aimin Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China.
| | - Bing Huang
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China.
| | - Xinmei Zhao
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China.
| |
Collapse
|
5
|
Chen Y, Liang J, Chen S, Lin N, Xu S, Miao J, Zhang J, Chen C, Yuan X, Xie Z, Zhu E, Cai M, Wei X, Hou S, Tang H. Discovery of vitexin as a novel VDR agonist that mitigates the transition from chronic intestinal inflammation to colorectal cancer. Mol Cancer 2024; 23:196. [PMID: 39272040 PMCID: PMC11395219 DOI: 10.1186/s12943-024-02108-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Colitis-associated colorectal cancer (CAC) frequently develops in patients with inflammatory bowel disease (IBD) who have been exposed to a prolonged state of chronic inflammation. The investigation of pharmacological agents and their mechanisms to prevent precancerous lesions and inhibit their progression remains a significant focus and challenge in CAC research. Previous studies have demonstrated that vitexin effectively mitigates CAC, however, its precise mechanism of action warrants further exploration. This study reveals that the absence of the Vitamin D receptor (VDR) accelerates the progression from chronic colitis to colorectal cancer. Our findings indicate that vitexin can specifically target the VDR protein, facilitating its translocation into the cell nucleus to exert transcriptional activity. Additionally, through a co-culture model of macrophages and cancer cells, we observed that vitexin promotes the polarization of macrophages towards the M1 phenotype, a process that is dependent on VDR. Furthermore, ChIP-seq analysis revealed that vitexin regulates the transcriptional activation of phenazine biosynthesis-like domain protein (PBLD) via VDR. ChIP assays and dual luciferase reporter assays were employed to identify the functional PBLD regulatory region, confirming that the VDR/PBLD pathway is critical for vitexin-mediated regulation of macrophage polarization. Finally, in a mouse model with myeloid VDR gene knockout, we found that the protective effects of vitexin were abolished in mid-stage CAC. In summary, our study establishes that vitexin targets VDR and modulates macrophage polarization through the VDR/PBLD pathway, thereby alleviating the transition from chronic colitis to colorectal cancer.
Collapse
Affiliation(s)
- Yonger Chen
- School of Basic Medical Sciences, State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, Guangzhou Medical University; The Affiliated Panyu Central Hospital of Guangzhou Medical University; Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine; School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Jian Liang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine; School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Shuxian Chen
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
| | - Nan Lin
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
| | - Shuoxi Xu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine; School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Jindian Miao
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine; School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Jing Zhang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine; School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Chen Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine; School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Xin Yuan
- Department of Pharmacy, The Second Affiliated Hospital of Guangzhou, University of Chinese Medicine, Guangzhou, 510120, China
| | - Zhuoya Xie
- State Key Laboratory of Oncology in South China Guangdong Provincial Clinical Research, Center for Cancer Sun Yat-Sen University Cancer Center Guangzhou, Guangzhou, 510060, China
| | - Enlin Zhu
- Clinical Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Mingsheng Cai
- School of Basic Medical Sciences, State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, Guangzhou Medical University; The Affiliated Panyu Central Hospital of Guangzhou Medical University; Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 511436, China.
| | - Xiaoli Wei
- State Key Laboratory of Oncology in South China Guangdong Provincial Clinical Research, Center for Cancer Sun Yat-Sen University Cancer Center Guangzhou, Guangzhou, 510060, China.
| | - Shaozhen Hou
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine; School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Hailin Tang
- State Key Laboratory of Oncology in South China Guangdong Provincial Clinical Research, Center for Cancer Sun Yat-Sen University Cancer Center Guangzhou, Guangzhou, 510060, China.
| |
Collapse
|
6
|
Qian S, Li R, He Y, Wang H, Zhang D, Sun A, Yu L, Song X, Zhao T, Chen Z, Yang Z. Immunogenicity and protective efficacy of a recombinant lactococcus lactis vaccine against HSV-1 infection. Microb Cell Fact 2024; 23:244. [PMID: 39252072 PMCID: PMC11385484 DOI: 10.1186/s12934-024-02517-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 08/31/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Herpes simplex virus type 1 (HSV-1) is a major cause of viral encephalitis, genital mucosal infections, and neonatal infections. Lactococcus lactis (L. lactis) has been proven to be an effective vehicle for delivering protein antigens and stimulating both mucosal and systemic immune responses. In this study, we constructed a recombinant L. lactis system expressing the protective antigen glycoprotein D (gD) of HSV-1. RESULTS To improve the stability and persistence of antigen stimulation of the local mucosa, we inserted the immunologic adjuvant interleukin (IL)-2 and the Fc fragment of IgG into the expression system, and a recombinant L. lactis named NZ3900-gD-IL-2-Fc was constructed. By utilizing this recombinant L. lactis strain to elicit an immune response and evaluate the protective effect in mice, the recombinant L. lactis vaccine induced a significant increase in specific neutralizing antibodies, IgG, IgA, interferon-γ, and IL-4 levels in the serum of mice. Furthermore, in comparison to the mice in the control group, the vaccine also enhanced the proliferation levels of lymphocytes in response to gD. Moreover, recombinant L. lactis expressing gD significantly boosted nonspecific immune reactions in mice through the activation of immune-related genes. Furthermore, following the HSV-1 challenge of the murine lung mucosa, mice inoculated with the experimental vaccine exhibited less lung damage than control mice. CONCLUSION Our study presents a novel method for constructing a recombinant vaccine using the food-grade, non-pathogenic, and non-commercial bacterium L. lactis. The findings indicate that this recombinant vaccine shows promise in preventing HSV-1 infection in mice.
Collapse
Affiliation(s)
- Shaoju Qian
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, 453003, Henan, China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Ruixue Li
- Department of Otolaryngology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, China
| | - Yeqing He
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Hexi Wang
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Danqiong Zhang
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Aiping Sun
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, 453003, Henan, China
| | - Lili Yu
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, 453003, Henan, China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xiangfeng Song
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, 453003, Henan, China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Tiesuo Zhao
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, 453003, Henan, China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Zhiguo Chen
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, 453003, Henan, China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Zishan Yang
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, 453003, Henan, China.
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
7
|
Wang R, Shu RR, Seldin L. Noncanonical functions of adhesion proteins in inflammation. Am J Physiol Cell Physiol 2024; 327:C505-C515. [PMID: 38981610 PMCID: PMC11427013 DOI: 10.1152/ajpcell.00292.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/24/2024] [Accepted: 07/01/2024] [Indexed: 07/11/2024]
Abstract
Cell adhesion proteins localize to epithelial and endothelial cell membranes to form junctional complexes between neighboring cells or between cells and the underlying basement membrane. The structural and functional integrities of these junctions are critical to establish cell polarity and maintain tissue barrier function, while also facilitating leukocyte migration and adhesion to sites of inflammation. In addition to their adhesive properties, however, junctional proteins can also serve important noncanonical functions in inflammatory signaling and transcriptional regulation. Intriguingly, recent work has unveiled novel roles for cell adhesion proteins as both signaling initiators and downstream targets during inflammation. In this review, we discuss both the traditional functions of junction proteins in cell adhesion and tissue barrier function as well as their noncanonical signaling roles that have been implicated in facilitating diverse inflammatory pathologies.
Collapse
Affiliation(s)
- Ruochong Wang
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Raphael R Shu
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Lindsey Seldin
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, United States
- Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia, United States
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States
| |
Collapse
|
8
|
Zhu J, Wu Y, Zhang L, Bai B, Han W, Wang H, Mei Q. Epithelial Piezo1 deletion ameliorates intestinal barrier damage by regulating ferroptosis in ulcerative colitis. Free Radic Biol Med 2024; 224:S0891-5849(24)00633-6. [PMID: 39216559 DOI: 10.1016/j.freeradbiomed.2024.08.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/16/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Ferroptosis, a recently discovered form of regulated cell death, has been implicated in the development of ulcerative colitis (UC). While Piezo1's role in inducing ferroptosis in chondrocytes and pulmonary endothelial cells is documented, its regulatory function in ferroptosis and intestinal epithelial cells in UC remains unclear. To address this, colonic tissue samples from patients with UC were examined, and specific intestinal epithelial Piezo1-deficient (Piezo1ΔIEC) mice were created to investigate Piezo1's role in UC pathogenesis. Elevated epithelial Piezo1 levels were observed in patients with UC, correlating with increased ferroptosis and tight junction (TJ) disruption. In dextran sulfate sodium (DSS)-induced colitis, Piezo1ΔIEC mice exhibited significantly reduced intestinal inflammation and improved gut barrier function compared to wild-type (WT) mice. Moreover, Piezo1 deficiency in colitis mice and lipopolysaccharide (LPS)-stimulated Caco-2 cells led to higher TJ protein levels, reduced lipid peroxidation, enhanced mitochondrial function, and altered expression of ferroptosis-associated proteins. Additionally, erastin, a ferroptosis activator, reversed the protective effect of Piezo1 silencing against LPS-induced ferroptosis in Caco-2 cells. Mechanistically, Piezo1 was found to regulate ferroptosis via the AMPK/mTOR signaling pathway. These findings highlight a novel role for Piezo1 deletion in mitigating ferroptosis in intestinal epithelial cells, suggesting Piezo1 as a potential therapeutic target for UC treatment.
Collapse
Affiliation(s)
- Jiejie Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China
| | - Yumei Wu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China
| | - Luyao Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China
| | - Bingqing Bai
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China
| | - Wei Han
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China.
| | - Qiao Mei
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China.
| |
Collapse
|
9
|
Liu S, Li J, Wu X. [Swertiamarin ameliorates 2, 4, 6-trinitrobenzenesulfonic acid-induced colitis in mice by inhibiting intestinal epithelial cell apoptosis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:1545-1552. [PMID: 39276050 PMCID: PMC11378047 DOI: 10.12122/j.issn.1673-4254.2024.08.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 09/16/2024]
Abstract
OBJECTIVE To investigate the mechanism by which swertiamarin (STM) ameliorates CD-like colitis in mice. METHODS A Caco-2 cell model of TNF-α-stimulated apoptosis was established and divided into three groups: Con, TNF-α and STM, and the effects of STM on apoptosis and barrier function were assessed by Tunel staining, western blotting, immunofluorescence, and transepithelial electric resistance (TEER). A mouse model of 2, 4, 6-trinitrobenzenesulfonic acid (TNBS) -induced CD-like colitis was established to assess the effects of STM on colitis, intestinal barrier function and epithelial cell apoptosis. The regulatory role of the PI3K/AKT pathway in STM-induced resistance to intestinal epithelial cell apoptosis was investigated in both the cell model and mouse models. RESULTS TUNEL staining showed that in Caco-2 cells with TNF-α stimulation, STM treatment significantly reduced the percentage of TUNEL-stained cells (P<0.05). STM obviously reduced TNF-α-induced enhancement of cleaved-caspase 3 and Bax expressions (P<0.05), increased Bcl-2 expression (P<0.05), protected intestinal barrier integrity and function by restoring transepithelial electrical resistance (TEER) of the cells, promoted normal localization and expressions of the tight junction proteins (ZO1 and claudin 1) (P<0.05), and inhibited the expression of pro-inflammatory factors (IL-6 and CCL3) (P<0.05) in TNF-α-stimulated Caco-2 cells. In the mouse models, STM significantly alleviated TNBS-induced CD-like colitis and intestinal barrier dysfunction (P<0.05) as shown by improved weight loss, lowered Disease Activity Index (DAI) score and inflammation score, reduction of IL-6 and CCL3 release, and restoration of intestinal barrier permeability, colonic TEER, bacterial translocation, and localization and expressions of the tight junction proteins. Mechanistically, STM inhibited the expressions of p-PI3K and p-AKT in both the cell model and mouse model(P<0.05), and treatment with 740Y-P (a PI3K/AKT pathway activator) significantly attenuated the inhibitory effect of STM on TNF-α-induced apoptosis in Caco-2 cells (P<0.05). CONCLUSION STM inhibits intestinal epithelial cell apoptosis at least in part by suppressing activation of the PI3K/AKT pathway to ameliorate intestinal barrier dysfunction and colitis in mice.
Collapse
Affiliation(s)
- S Liu
- First Clinical Medical College, Anhui Medical University, Hefei 230000, China
| | - J Li
- Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233003, China
- Anhui Provincial Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu 233003, China
| | - X Wu
- Department of Radiology, First Affiliated Hospital of Anhui Medical University, Hefei 230000, China
| |
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [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
|
Kang M, Kang M, Lee J, Yoo J, Lee S, Oh S. Allium tuberosum-derived nanovesicles with anti-inflammatory properties prevent DSS-induced colitis and modify the gut microbiome. Food Funct 2024; 15:7641-7657. [PMID: 38953279 DOI: 10.1039/d4fo01366b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Edible plant-derived nanovesicles (ePDNs) have shown potential as a non-pharmacological option for inflammatory bowel disease (IBD) by maintaining gut health and showing anti-inflammatory effects. However, the effects of Allium tuberosum-derived nanovesicles (ADNs) on colitis have not been studied to date. Here, we extracted exosome-like nanovesicles from Allium tuberosum and investigated whether they have an anti-inflammatory effect in RAW 264.7 cells and colitis mice. The results showed that ADNs reduced the elevated levels of inflammatory factors such as IL-1β, IL-6, TNF-α, and NF-κB pathway-related proteins as a consequence of lipopolysaccharide (LPS) stimulation in RAW 264.7 cells. Furthermore, our mouse experiments demonstrated that ADNs could ameliorate dextran sulfate sodium (DSS)-induced colitis symptoms (e.g., increased disease activity index score, intestinal permeability, and histological appearance). Additionally, ADNs counteracted DSS-induced colitis by downregulating the expression of serum amyloid A (SAA), IL-1β, IL-6, and TNF-α and increasing the expression of tight junction proteins (ZO-1 and occludin) and the anti-inflammatory cytokine IL-10. 16S rRNA gene sequencing showed that ADN intervention restored the gut microbial composition, which was similar to that of the DSS non-treated group, by decreasing the ratio of Firmicutes to Bacteroidetes and the relative abundance of Proteobacteria. Furthermore, ADNs induced acetic acid production along with an increase in the abundance of Lactobacillus. Overall, our findings suggest that ADN supplementation has a crucial role in maintaining gut health and is a novel preventive therapy for IBD.
Collapse
Affiliation(s)
- Minkyoung Kang
- Department of Environmental Science and Biotechnology, Jeonju University, Jeonju 55069, Republic of Korea.
| | - Minji Kang
- Department of Environmental Science and Biotechnology, Jeonju University, Jeonju 55069, Republic of Korea.
| | - Juyeon Lee
- Department of Environmental Science and Biotechnology, Jeonju University, Jeonju 55069, Republic of Korea.
| | - Jiseon Yoo
- Department of Environmental Science and Biotechnology, Jeonju University, Jeonju 55069, Republic of Korea.
| | - Sujeong Lee
- Department of Environmental Science and Biotechnology, Jeonju University, Jeonju 55069, Republic of Korea.
| | - Sangnam Oh
- Department of Environmental Science and Biotechnology, Jeonju University, Jeonju 55069, Republic of Korea.
- Department of Food and Nutrition, Jeonju University, Jeonju 55069, Republic of Korea
| |
Collapse
|
12
|
Chen M, Wei S, Wu X, Xiang Z, Li X, He H, Liao F, Wang X, Zhang J, Yu B, Dong W. 2'-Hydroxycinnamaldehyde Alleviates Intestinal Inflammation by Attenuating Intestinal Mucosal Barrier Damage Via Directly Inhibiting STAT3. Inflamm Bowel Dis 2024; 30:992-1008. [PMID: 38422244 PMCID: PMC11144992 DOI: 10.1093/ibd/izad283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Indexed: 03/02/2024]
Abstract
BACKGROUND The currently available clinical therapeutic drugs for ulcerative colitis (UC) are considered inadequate owing to certain limitations. There have been reports on the anti-inflammatory effects of 2'-hydroxycinnamaldehyde (HCA). However, whether HCA can improve UC is still unclear. Here, we aimed to investigate the pharmacological effects of HCA on UC and its underlying molecular mechanisms. METHODS The pharmacological effects of HCA were comprehensively investigated in 2 experimental setups: mice with dextran sulfate sodium (DSS)-induced colitis and lipopolysaccharide (LPS)-treated fetal human colon (FHC) cells. Furthermore, the interaction between HCA and signal transducer and activator of transcription 3 (STAT3) was investigated using molecular docking. The FHC cells with STAT3 knockdown or overexpression and mice with intestinal epithelium-specific STAT3 deletion (STAT3ΔIEC) were used to evaluate whether STAT3 mediated the pharmacological effects of HCA. RESULTS 2'-Hydroxycinnamaldehyde attenuated dysregulated expression of inflammatory cytokines in a dose-dependent manner while increasing the expression of tight junction proteins, reducing the apoptosis of intestinal epithelial cells, and effectively alleviating inflammation both in vivo and in vitro. 2'-Hydroxycinnamaldehyde bound directly to STAT3 and inhibited its activation. The modulation of STAT3 activation levels due to STAT3 knockdown or overexpression influenced the mitigating effects of HCA on colitis. Further analysis indicated that the remission effect of HCA was not observed in STAT3ΔIEC mice, indicating that STAT3 mediated the anti-inflammatory effects of HCA. CONCLUSIONS We present a novel finding that HCA reduces colitis severity by attenuating intestinal mucosal barrier damage via STAT3. This discovery holds promise as a potential new strategy to alleviate UC.
Collapse
Affiliation(s)
- Meilin Chen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shuchun Wei
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaohan Wu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zixuan Xiang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiangyun Li
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Haodong He
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fei Liao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoli Wang
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jixiang Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Baoping Yu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
13
|
Kulhari U, Rajanan A, Ambujakshan A, Verma S, Mugale MN, Sahu BD. Biochanin A mitigates ulcerative colitis and intestinal inflammation in mice by inhibiting MAPK/NF-kB (p65) axis. J Biochem Mol Toxicol 2024; 38:e23738. [PMID: 38764152 DOI: 10.1002/jbt.23738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/21/2024] [Accepted: 05/09/2024] [Indexed: 05/21/2024]
Abstract
Ulcerative colitis (UC) is a chronic problem of the intestine and relapsing in nature. Biochanin A is a nature-derived isoflavonoid and has numerous bioactivities. However, its role against UC and intestinal inflammation remains obscure. We aimed to comprehensively explore the pharmacological effect of biochanin A in alleviating colitis and to evaluate the potential mechanisms. Initially, we explored the anti-inflammatory action of biochanin A (15, 30, and 60 μM) by employing lipopolysaccharide (LPS)-activated RAW 264.7 cells. In RAW 264.7 cells under LPS stimulation, biochanin A inhibited the elevation of reactive oxygen species (ROS) (p < 0.0001), interleukin (IL)-1β (p < 0.0001), IL-18 (p < 0.01), and tumor necrosis factor (TNF)-α (p < 0.01) release, nitrite production (p < 0.0001), and the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) proteins. Next, we studied the effectiveness of biochanin A (20 and 40 mg/kg) in mouse colitis induced with dextran sulfate sodium (DSS) by assessing colon length, disease activity index (DAI) scoring, and performing colonoscopy and histological analysis. The pro-inflammatory cytokines were estimated using ELISA. Western blot studies were performed to assess underlying mechanisms. In mice, biochanin A treatment alleviated DAI score (p < 0.0001), restored colon length (p < 0.05) and morphology, and re-established colon histopathology. Biochanin A affects the phosphorylation of proteins associated with NF-κB (p65) and mitogen-activated protein kinase (MAPK) axis and regulates colonic inflammation by reducing the expression of inflammatory cytokines and myeloperoxidase (MPO) activity. Altogether, our findings support the idea that the anticolitis potential of biochanin A is allied with anti-inflammatory activity by inhibiting the MAPK/NF-κB (p65) axis. Hence, biochanin A may be an alternative option to alleviate the risk of colitis.
Collapse
Affiliation(s)
- Uttam Kulhari
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari, Assam, India
| | - Ashitha Rajanan
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari, Assam, India
| | - Anju Ambujakshan
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari, Assam, India
| | - Smriti Verma
- Toxicology & Experimental Medicine, CSIR-Central Drug Research Institute (CDRI), Lucknow, India
| | - Madhav Nilakanth Mugale
- Toxicology & Experimental Medicine, CSIR-Central Drug Research Institute (CDRI), Lucknow, India
| | - Bidya Dhar Sahu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari, Assam, India
| |
Collapse
|
14
|
Xiao C, Comer L, Pan X, Everaert N, Schroyen M, Song Z. Zinc glycinate alleviates LPS-induced inflammation and intestinal barrier disruption in chicken embryos by regulating zinc homeostasis and TLR4/NF-κB pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 272:116111. [PMID: 38350216 DOI: 10.1016/j.ecoenv.2024.116111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 01/26/2024] [Accepted: 02/11/2024] [Indexed: 02/15/2024]
Abstract
The effect of an immune challenge induced by a lipopolysaccharide (LPS) exposure on systemic zinc homeostasis and the modulation of zinc glycinate (Zn-Gly) was investigated using a chicken embryo model. 160 Arbor Acres broiler fertilized eggs were randomly divided into 4 groups: CON (control group, injected with saline), LPS (LPS group, injected with 32 µg of LPS saline solution), Zn-Gly (zinc glycinate group, injected with 80 µg of zinc glycinate saline solution) and Zn-Gly+LPS (zinc glycinate and LPS group, injected with the same content of zinc glycinate and LPS saline solution). Each treatment consisted of eight replicates of five eggs each. An in ovo feeding procedure was performed at 17.5 embryonic day and samples were collected after 12 hours. The results showed that Zn-Gly attenuated the effects of LPS challenge-induced upregulation of pro-inflammatory factor interleukin 1β (IL-1β) level (P =0.003). The LPS challenge mediated zinc transporter proteins and metallothionein (MT) to regulate systemic zinc homeostasis, with increased expression of the jejunum zinc export gene zinc transporter protein 1 (ZnT-1) and elevated expression of the import genes divalent metal transporter 1 (DMT1), Zrt- and Irt-like protein 3 (Zip3), Zip8 and Zip14 (P < 0.05). A similar trend could be observed for the zinc transporter genes in the liver, which for ZnT-1 mitigated by Zn-Gly supplementation (P =0.01). Liver MT gene expression was downregulated in response to the LPS challenge (P =0.004). These alterations caused by LPS resulted in decreased serum and liver zinc levels and increased small intestinal, muscle and tibial zinc levels. Zn-Gly reversed the elevated expression of the liver zinc finger protein A20 induced by the LPS challenge (P =0.025), while Zn-Gly reduced the gene expression of the pro-inflammatory factors IL-1β and IL-6, decreased toll-like receptor 4 (TLR4) and nuclear factor kappa-B p65 (NF-κB p65) (P < 0.05). Zn-Gly also alleviated the LPS-induced downregulation of the intestinal barrier gene Claudin-1. Thus, LPS exposure prompted the mobilization of zinc transporter proteins and MT to perform the remodeling of systemic zinc homeostasis, Zn-Gly participated in the regulation of zinc homeostasis and inhibited the production of pro-inflammatory factors through the TLR4/NF-κB pathway, attenuating the inflammatory response and intestinal barrier damage caused by an immune challenge.
Collapse
Affiliation(s)
- Chuanpi Xiao
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources, Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China; Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Luke Comer
- Nutrition and Animal Microbiota Ecosystems lab, Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Xue Pan
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources, Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China
| | - Nadia Everaert
- Nutrition and Animal Microbiota Ecosystems lab, Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Martine Schroyen
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Zhigang Song
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources, Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China.
| |
Collapse
|
15
|
Li J, Sun Y, Yang N, Zhang H, Hu Y, Wang H, Zhang R, Ge M. Protective effects of maternal administration of total saponins of Codonopsis pilosula in the mice offspring following diarrhea: role of immune function, antioxidant function, and intestinal inflammatory injury. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:113903-113916. [PMID: 37858017 DOI: 10.1007/s11356-023-30281-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 10/02/2023] [Indexed: 10/21/2023]
Abstract
Total saponins of Codonopsis (TSC) are a kind of critical bioactive substances in Codonopsis, which have anti-inflammatory, antioxidant, anti-ulcer, immunomodulatory effects, and protective effects on ulcerative enteritis. In this study, TSC (3.75 mL/kg, gavage) was administered once a day to 13-day gestation Kunming mice for 5 days. On day 13 of birth, the offspring were given Escherichia coli solution (0.15 mL/mouse, intraperitoneal injection) and senna leaf decoction (0.15 mL/mouse, gavage) once a day for 6 days. The results showed that gestation maternal administration of TSC effectively reduced the diarrhea index, increased the content of sIgA, IgG, SOD, and GSH, inhibited the TLR4/MyD88/NF-κB pathway in the intestine, reduced the expression of inflammatory factors, and alleviated intestinal injury in the littermates. The results provided a critical reference for the clinical application of TSC to control diarrhea in animal offspring.
Collapse
Affiliation(s)
- Jiali Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin, 150030, PR China
| | - Yiming Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin, 150030, PR China
| | - Naixi Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin, 150030, PR China
| | - Haolin Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin, 150030, PR China
| | - Yihan Hu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin, 150030, PR China
| | - Haibin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin, 150030, PR China
| | - Ruili Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin, 150030, PR China
| | - Ming Ge
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin, 150030, PR China.
| |
Collapse
|
16
|
Tan Y, Wang Y, Liu Q, Wang Z, Shi S, Su J. Comparative transcriptomes reveal geographic differences in the ability of the liver of plateau zokors (Eospalax baileyi) to respond and adapt to toxic plants. BMC Genomics 2023; 24:529. [PMID: 37674113 PMCID: PMC10483729 DOI: 10.1186/s12864-023-09642-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 08/31/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND Environmental changes are expected to intensify in the future. The invasion of toxic plants under environmental changes may change herbivore feeding environments. Herbivores living long-term in toxic plant-feeding environments will inevitably ingest plant secondary metabolites (PSMs), and under different feeding environments are likely to have unique protection mechanisms that support improved adaptation to PSMs in their habitat. We aimed to compare different subterranean herbivore population responses and adaptations to toxic plants to unveil their feeding challenges. RESULTS Here, we investigated the adaptive capacity of the liver in two geographically separated populations of plateau zokors (Eospalax baileyi) before and after exposure to the toxic plant Stellera chamaejasme (SC), at the organ, biochemical, and transcriptomic levels. The results showed no significant liver granules or inflammatory reactions in the Tianzhu (TZ) population after the SC treatment. The transaminase level in the TZ population was significantly lower than that in the Luqu population. Transcriptome analysis revealed that the TZ population exhibited interactions with other detoxification metabolic pathways by oxytocin pathway-associated genes, including diacylglycerol lipase alpha (Dagla), calcium/calmodulin dependent protein kinase II Alpha (Camk2a), and CD38 molecule (Cd38). The phase II process of liver drug metabolism increased to promote the rate of metabolism. We found that alternative splicing (AS) and the expression of the cyclin D (Ccnd1) gene interact-a TZ population hallmark-reduced liver inflammatory responses. CONCLUSION Our study supports the detoxification limitation hypothesis that differences in liver detoxification metabolism gene expression and AS are potential factors in herbivore adaptation to PSMs and may be a strategy of different herbivore populations to improve toxic plant adaptability.
Collapse
Affiliation(s)
- Yuchen Tan
- College of Grassland Science, Key Laboratory of Grassland Ecosystem (Ministry of Education), Gansu Agricultural University, Lanzhou, 730070, China
- Gansu Agricultural University-Massey University Research Centre for Grassland Biodiversity, Gansu Agricultural University, Lanzhou, 730070, China
| | - Yanli Wang
- College of Grassland Science, Key Laboratory of Grassland Ecosystem (Ministry of Education), Gansu Agricultural University, Lanzhou, 730070, China
- Gansu Agricultural University-Massey University Research Centre for Grassland Biodiversity, Gansu Agricultural University, Lanzhou, 730070, China
| | - Qianqian Liu
- College of Grassland Science, Key Laboratory of Grassland Ecosystem (Ministry of Education), Gansu Agricultural University, Lanzhou, 730070, China
- Gansu Agricultural University-Massey University Research Centre for Grassland Biodiversity, Gansu Agricultural University, Lanzhou, 730070, China
| | - Zhicheng Wang
- College of Grassland Science, Key Laboratory of Grassland Ecosystem (Ministry of Education), Gansu Agricultural University, Lanzhou, 730070, China
- Gansu Agricultural University-Massey University Research Centre for Grassland Biodiversity, Gansu Agricultural University, Lanzhou, 730070, China
| | - Shangli Shi
- College of Grassland Science, Key Laboratory of Grassland Ecosystem (Ministry of Education), Gansu Agricultural University, Lanzhou, 730070, China
- Gansu Agricultural University-Massey University Research Centre for Grassland Biodiversity, Gansu Agricultural University, Lanzhou, 730070, China
| | - Junhu Su
- College of Grassland Science, Key Laboratory of Grassland Ecosystem (Ministry of Education), Gansu Agricultural University, Lanzhou, 730070, China.
- Gansu Agricultural University-Massey University Research Centre for Grassland Biodiversity, Gansu Agricultural University, Lanzhou, 730070, China.
| |
Collapse
|
17
|
Liang J, Zhou Y, Cheng X, Chen J, Cao H, Guo X, Zhang C, Zhuang Y, Hu G. Baicalin Attenuates H 2O 2-Induced Oxidative Stress by Regulating the AMPK/Nrf2 Signaling Pathway in IPEC-J2 Cells. Int J Mol Sci 2023; 24:ijms24119435. [PMID: 37298392 DOI: 10.3390/ijms24119435] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/10/2023] [Accepted: 04/14/2023] [Indexed: 06/12/2023] Open
Abstract
Oxidative stress can adversely affect the health status of the body, more specifically by causing intestinal damage by disrupting the permeability of the intestinal barrier. This is closely related to intestinal epithelial cell apoptosis caused by the mass production of reactive oxygen species (ROS). Baicalin (Bai) is a major active ingredient in Chinese traditional herbal medicine that has antioxidant, anti-inflammatory, and anti-cancer properties. The purpose of this study was to explore the underlying mechanisms by which Bai protects against hydrogen peroxide (H2O2)-induced intestinal injury in vitro. Our results indicated that H2O2 treatment caused injury to IPEC-J2 cells, resulting in their apoptosis. However, Bai treatment attenuated H2O2-induced IPEC-J2 cell damage by up-regulating the mRNA and protein expression of ZO-1, Occludin, and Claudin1. Besides, Bai treatment prevented H2O2-induced ROS and MDA production and increased the activities of antioxidant enzymes (SOD, CAT, and GSH-PX). Moreover, Bai treatment also attenuated H2O2-induced apoptosis in IPEC-J2 cells by down-regulating the mRNA expression of Caspase-3 and Caspase-9 and up-regulating the mRNA expression of FAS and Bax, which are involved in the inhibition of mitochondrial pathways. The expression of Nrf2 increased after treatment with H2O2, and Bai can alleviate this phenomenon. Meanwhile, Bai down-regulated the ratio of phosphorylated AMPK to unphosphorylated AMPK, which is indicative of the mRNA abundance of antioxidant-related genes. In addition, knockdown of AMPK by short-hairpin RNA (shRNA) significantly reduced the protein levels of AMPK and Nrf2, increased the percentage of apoptotic cells, and abrogated Bai-mediated protection against oxidative stress. Collectively, our results indicated that Bai attenuated H2O2-induced cell injury and apoptosis in IPEC-J2 cells through improving the antioxidant capacity through the inhibition of the oxidative stress-mediated AMPK/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Jiahua Liang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, China
| | - Ying Zhou
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, China
| | - Xinyi Cheng
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, China
| | - Jiaqi Chen
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, China
| | - Huabin Cao
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, China
| | - Xiaoquan Guo
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, China
| | - Caiying Zhang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, China
| | - Yu Zhuang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, China
| | - Guoliang Hu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, China
| |
Collapse
|
18
|
Yuan SN, Wang MX, Han JL, Feng CY, Wang M, Wang M, Sun JY, Li NY, Simal-Gandara J, Liu C. Improved colonic inflammation by nervonic acid via inhibition of NF-κB signaling pathway of DSS-induced colitis mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 112:154702. [PMID: 36764096 DOI: 10.1016/j.phymed.2023.154702] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 01/14/2023] [Accepted: 02/03/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Nervonic acid (C24:1∆15, 24:1 ω-9, cis-tetracos-15-enoic acid; NA), a long-chain monounsaturated fatty acid, plays an essential role in prevention of metabolic diseases, and immune regulation, and has anti-inflammatory properties. As a chronic, immune-mediated inflammatory disease, ulcerative colitis (UC) can affect the large intestine. The influences of NA on UC are largely unknown. PURPOSE The present study aimed to decipher the anti-UC effect of NA in the mouse colitis model. Specifically, we wanted to explore whether NA can regulate the levels of inflammatory factors in RAW264.7 cells and mouse colitis model. METHODS To address the above issues, the RAW264.7 cell inflammation model was established by lipopolysaccharide (LPS), then the inflammatory factors tumor necrosis factor-α (TNF-α), Interleukin-6 (IL-6), Interleukin-1β (IL-1β), and Interleukin-10 (IL-10) were detected by Enzyme-linked immunosorbent assay (ELISA). The therapeutic effects of NA for UC were evaluated using C57BL/6 mice gavaged dextran sodium sulfate (DSS). Hematoxylin and eosin (H&E) staining, Myeloperoxidase (MPO) kit assay, ELISA, immunofluorescence assay, and LC-MS/MS were used to assess histological changes, MPO levels, inflammatory factors release, expression and distribution of intestinal tight junction (TJ) protein ZO-1, and metabolic pathways, respectively. The levels of proteins involved in the nuclear factor kappa-B (NF-κB) pathway in the UC were investigated by western blotting and RT-qPCR. RESULTS In vitro experiments verified that NA could reduce inflammatory response and inhibit the activation of key signal pathways associated with inflammation in LPS-induced RAW264.7 cells. Further, results from the mouse colitis model suggested that NA could restore intestinal barrier function and suppress NF-κB signal pathways to ameliorate DSS-induced colitis. In addition, untargeted metabolomics analysis of NA protection against UC found that NA protected mice from colitis by regulating citrate cycle, amino acid metabolism, pyrimidine and purine metabolism. CONCLUSION These results suggested that NA could ameliorate the secretion of inflammatory factors, suppress the NF-κB signaling pathway, and protect the integrity of colon tissue, thereby having a novel role in prevention or treatment therapy for UC. This work for the first time indicated that NA might be a potential functional food ingredient for preventing and treating inflammatory bowel disease (IBD).
Collapse
Affiliation(s)
- Sheng-Nan Yuan
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 202 Gongye North Road, Jinan 250100, China
| | - Mu-Xuan Wang
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 202 Gongye North Road, Jinan 250100, China
| | - Jin-Long Han
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 202 Gongye North Road, Jinan 250100, China
| | - Cai-Yun Feng
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 202 Gongye North Road, Jinan 250100, China
| | - Meng Wang
- Shanxi Functional Food Engineering Center Co. Ltd, Xian 710000, China
| | - Min Wang
- College of Food Science and Engineering, Northwest A&F University, Xianyang 712100, China
| | - Jin-Yue Sun
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 202 Gongye North Road, Jinan 250100, China; Shandong Huatai Nutrition and Health Industry Technology Research Institute Co. Ltd, Jinan 250100, China.
| | - Ning-Yang Li
- Key Laboratory of Food Processing Technology and Quality Control in Shandong Province, College of Food Science and Engineering, Shandong Agricultural University, Taian, China.
| | - Jesus Simal-Gandara
- Universidade de Vigo, Nutrition and Bromatology Group, Analytical Chemistry and Food Science Department, Faculty of Science, E-32004 Ourense, Spain.
| | - Chao Liu
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 202 Gongye North Road, Jinan 250100, China; Shandong Huatai Nutrition and Health Industry Technology Research Institute Co. Ltd, Jinan 250100, China.
| |
Collapse
|
19
|
Smith AD, Chen C, Cheung L, Dawson HD. Raw potato starch alters the microbiome, colon and cecal gene expression, and resistance to Citrobacter rodentium infection in mice fed a Western diet. Front Nutr 2023; 9:1057318. [PMID: 36704785 PMCID: PMC9871501 DOI: 10.3389/fnut.2022.1057318] [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: 09/29/2022] [Accepted: 12/05/2022] [Indexed: 01/11/2023] Open
Abstract
Resistant starches (RS) are fermented in the cecum and colon to produce short-chain fatty acids and other microbial metabolites that can alter host physiology and the composition of the microbiome. We previously showed that mice fed a Total Western Diet (TWD) based on NHANES data that mimics the composition of a typical American diet, containing resistant potato starch (RPS), produced concentration dependent changes to the cecal short-chain fatty acids, the microbiome composition as well as gene expression changes in the cecum and colon that were most prevalent in mice fed the 10% RPS diet. We were then interested in whether feeding TWD/RPS would alter the resistance to bacterial-induced colitis caused by Citrobacter rodentium (Cr), a mouse pathogen that shares 66.7% of encoded genes with Enteropathogenic Escherichia coli. Mice were fed the TWD for 6 weeks followed by a 3-weeks on the RPS diets before infecting with Cr. Fecal Cr excretion was monitored over time and fecal samples were collected for 16S sequencing. Mice were euthanized on day 12 post-infection and cecal contents collected for 16S sequencing. Cecum and colon tissues were obtained for gene expression analysis, histology and to determine the level of mucosa-associated Cr. Feeding RPS increased the percentage of mice productively infected by Cr and fecal Cr excretion on day 4 post-infection. Mice fed the TWD/10% RPS diet also had greater colonization of colonic tissue at day 12 post-infection and colonic pathology. Both diet and infection altered the fecal and cecal microbiome composition with increased levels of RPS resulting in decreased α-diversity that was partially reversed by Cr infection. RNASeq analysis identified several mechanistic pathways that could be associated with the increased colonization of Cr-infected mice fed 10% RPS. In the distal colon we found a decrease in enrichment for genes associated with T cells, B cells, genes associated with the synthesis of DHA-derived SPMs and VA metabolism/retinoic acid signaling. We also found an increase in the expression of the potentially immunosuppressive gene, Ido1. These results suggest that high-level consumption of RPS in the context of a typical American diet, may alter susceptibility to gastrointestinal bacterial infections.
Collapse
|
20
|
Prasad R, Floyd JL, Dupont M, Harbour A, Adu-Agyeiwaah Y, Asare-Bediako B, Chakraborty D, Kichler K, Rohella A, Calzi SL, Lammendella R, Wright J, Boulton ME, Oudit GY, Raizada MK, Stevens BR, Li Q, Grant MB. Maintenance of Enteral ACE2 Prevents Diabetic Retinopathy in Type 1 Diabetes. Circ Res 2023; 132:e1-e21. [PMID: 36448480 PMCID: PMC9822874 DOI: 10.1161/circresaha.122.322003] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/16/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND We examined components of systemic and intestinal renin-angiotensin system on gut barrier permeability, glucose homeostasis, systemic inflammation, and progression of diabetic retinopathy (DR) in human subjects and mice with type 1 diabetes (T1D). METHODS T1D individual with (n=18) and without (n=20) DR and controls (n=34) were examined for changes in gut-regulated components of the immune system, gut leakage markers (FABP2 [fatty acid binding protein 2] and peptidoglycan), and Ang II (angiotensin II); Akita mice were orally administered a Lactobacillus paracasei (LP) probiotic expressing humanized ACE2 (angiotensin-converting enzyme 2) protein (LP-ACE2) as either a prevention or an intervention. Akita mice with genetic overexpression of humanAce2 by small intestine epithelial cells (Vil-Cre.hAce2KI-Akita) were similarly examined. After 9 months of T1D, circulatory, enteral, and ocular end points were assessed. RESULTS T1D subjects exhibit elevations in gut-derived circulating immune cells (ILC1 cells) and higher gut leakage markers, which were positively correlated with plasma Ang II and DR severity. The LP-ACE2 prevention cohort and genetic overexpression of intestinal ACE2 preserved barrier integrity, reduced inflammatory response, improved hyperglycemia, and delayed development of DR. Improvements in glucose homeostasis were due to intestinal MasR activation, resulting in a GSK-3β (glycogen synthase kinase-3 beta)/c-Myc (cellular myelocytomatosis oncogene)-mediated decrease in intestinal glucose transporter expression. In the LP-ACE2 intervention cohort, gut barrier integrity was improved and DR reversed, but no improvement in hyperglycemia was observed. These data support that the beneficial effects of LP-ACE2 on DR are due to the action of ACE2, not improved glucose homeostasis. CONCLUSIONS Dysregulated systemic and intestinal renin-angiotensin system was associated with worsening gut barrier permeability, gut-derived immune cell activation, systemic inflammation, and progression of DR in human subjects. In Akita mice, maintaining intestinal ACE2 expression prevented and reversed DR, emphasizing the multifaceted role of the intestinal renin-angiotensin system in diabetes and DR.
Collapse
Affiliation(s)
- Ram Prasad
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Jason L. Floyd
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Mariana Dupont
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Angela Harbour
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Yvonne Adu-Agyeiwaah
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Bright Asare-Bediako
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Dibyendu Chakraborty
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Kara Kichler
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Aayush Rohella
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Sergio Li Calzi
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | | | | | - Michael E. Boulton
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Gavin Y. Oudit
- Division of Cardiology, Department of Medicine, University of Alberta, Mazankowski Alberta Heart Institute, Edmonton, AB, T6G 2B7, Canada
| | - Mohan K. Raizada
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Bruce R. Stevens
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Qiuhong Li
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Maria B. Grant
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| |
Collapse
|
21
|
Zhao X, Cui DJ, Yang LC, Yuan WQ, Yan F. Long Noncoding RNA FBXL19-AS1-Mediated Ulcerative Colitis-Associated Intestinal Epithelial Barrier Defect. Tissue Eng Regen Med 2022; 19:1077-1088. [PMID: 36048401 PMCID: PMC9478015 DOI: 10.1007/s13770-022-00479-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/18/2022] [Accepted: 07/04/2022] [Indexed: 10/14/2022] Open
Abstract
BACKGROUND This study commenced to uncover the role of long non-coding RNA FBXL19 antisense RNA 1 (FBXL19-AS1) in the development of ulcerative colitis (UC) and its possible mechanism. METHODS FBXL19-AS1 expression in the colonic sigmoid mucosa of UC patients was detected. A colitis model was induced in mice using 5% dextran sodium sulfate. Hematoxylin-eosin staining was performed for histopathological examination. Apoptosis was detected by Tunel staining and tissue fibrosis was detected by immunohistochemistry. Also, intestinal permeability was examined. The concentrations of inflammatory factors IL-1β and IL-18 were detected by enzyme-linked immunosorbent assay. The relationship between FBXL19-AS1, miR-339-3p and RHOB was verified by RNA immunoprecipitation assay and dual luciferase reporter assay. RESULTS The expression of FBXL19-AS1 was increased in dextran sodium sulfate (DSS)-induced colitis mouse model. FBXL19-AS1 interference or miR-339-3p overexpression inhibited DSS-induced colonic epithelial cell apoptosis and inflammatory response, and improved intestinal epithelial barrier defects, thereby ameliorating DSS-induced colitis injury in mice. FBXL19-AS1 sponged miR-339-3p while miR-339-3p targeted RHOB. Overexpression of RHOB reversed the protective effect of inhibition of FBXL19-AS1 on DSS-induced colitis in mice. CONCLUSION FBXL19-AS1 reduces miR-339-3p-mediated targeting of RHOB and aggravates intestinal epithelial barrier defect in DSS-induced colitis in mice.
Collapse
Affiliation(s)
- Xun Zhao
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, No. 83, East Zhongshan Road, Guiyang City, 550002, Guizhou Province, China.
| | - De-Jun Cui
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, No. 83, East Zhongshan Road, Guiyang City, 550002, Guizhou Province, China
| | - Liu-Chan Yang
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, No. 83, East Zhongshan Road, Guiyang City, 550002, Guizhou Province, China
| | - Wen-Qiang Yuan
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, No. 83, East Zhongshan Road, Guiyang City, 550002, Guizhou Province, China
| | - Fang Yan
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, No. 83, East Zhongshan Road, Guiyang City, 550002, Guizhou Province, China
| |
Collapse
|
22
|
Wang K, Chen D, Yu B, He J, Mao X, Huang Z, Yan H, Wu A, Luo Y, Zheng P, Yu J, Luo J. Eugenol alleviates transmissible gastroenteritis virus-induced intestinal epithelial injury by regulating NF-κB signaling pathway. Front Immunol 2022; 13:921613. [PMID: 36052062 PMCID: PMC9427193 DOI: 10.3389/fimmu.2022.921613] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 07/29/2022] [Indexed: 01/13/2023] Open
Abstract
Increasing evidence supports the ability of eugenol to maintain intestinal barrier integrity and anti-inflammatory in vitro and in vivo; however, whether eugenol alleviates virus-mediated intestinal barrier damage and inflammation remains a mystery. Transmissible gastroenteritis virus (TGEV), a coronavirus, is one of the main causative agents of diarrhea in piglets and significantly impacts the global swine industry. Here, we found that eugenol could alleviate TGEV-induced intestinal functional impairment and inflammatory responses in piglets. Our results indicated that eugenol improved feed efficiency in TGEV-infected piglets. Eugenol not only increased serum immunoglobulin concentration (IgG) but also significantly decreased serum inflammatory cytokine concentration (TNF-α) in TGEV-infected piglets. In addition, eugenol also significantly decreased the expression of NF-κB mRNA and the phosphorylation level of NF-κB P65 protein in the jejunum mucosa of TGEV-infected piglets. Eugenol increased villus height and the ratio of villus height to crypt depth in the jejunum and ileum, and decreased serum D-lactic acid levels. Importantly, eugenol increased tight junction protein (ZO-1) and mRNA expression levels of nutrient transporter-related genes (GluT-2 and CaT-1) in the jejunum mucosa of TGEV-infected piglets. Meanwhile, compared with TGEV-infected IPEC-J2 cells, treatment with eugenol reduced the cell cytopathic effect, attenuated the inflammatory response. Interestingly, eugenol did not increase the expression of ZO-1 and Occludin in IPEC-J2 cells. However, western blot and immunofluorescence results showed that eugenol restored TGEV-induced down-regulation of ZO-1 and Occludin, while BAY11-7082 (The NF-κB specific inhibitor) enhanced the regulatory ability of eugenol. Our findings demonstrated that eugenol attenuated TGEV-induced intestinal injury by increasing the expression of ZO-1 and Occludin, which may be related to the inhibition of NF-κB signaling pathway. Eugenol may offer some therapeutic opportunities for coronavirus-related diseases.
Collapse
Affiliation(s)
- Kang Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-resistant Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-resistant Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-resistant Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-resistant Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-resistant Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Zhiqing Huang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-resistant Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-resistant Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Aimin Wu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-resistant Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Yuheng Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-resistant Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-resistant Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Jie Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-resistant Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Junqiu Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-resistant Nutrition, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
23
|
Liu X, Ren X, Zhou L, Liu K, Deng L, Qing Q, Li J, Zhi F, Li M. Tollip Orchestrates Macrophage Polarization to Alleviate Intestinal Mucosal Inflammation. J Crohns Colitis 2022; 16:1151-1167. [PMID: 35134154 DOI: 10.1093/ecco-jcc/jjac019] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/13/2021] [Accepted: 01/29/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Regulation of macrophage polarization is a promising strategy for treating inflammatory bowel disease [IBD]. Tollip is an important negative regulator of Toll-like receptor [TLR]-mediated innate immunity with downregulated expression in the colon tissues of patients with IBD. This study aimed to regulate the expression of Tollip to affect macrophage polarization. METHODS A molecular, targeted immunotherapy method was developed by linking mannose-modified trimethyl chitosan [MTC] with Tollip-expressing plasmids via ionic cross-linking, forming MTC-Tollip nanoparticles with a targeting function. MTC-Tollip selectively targeted mouse intestinal macrophages to regulate the polarization of macrophages for mucosal repair. RESULTS Orally administered MTC-Tollip significantly elevated Tollip expression in intestinal tissue. Compared with MTC-negative control [NC]-treated mice in which colitis was induced with dextran sodium sulphate [DSS], the MTC-Tollip nanoparticle-treated mice exhibited decreased body weight loss and colon shortening, lower proinflammatory cytokine expression in colon tissues, and greater mucosal barrier integrity. MTC-Tollip treatment decreased TNF-α and iNOS expression but increased CD206 and Arg-1 expression in colon tissue. Tollip overexpression in mouse peritoneal macrophages inhibited lipopolysaccharide [LPS]-induced proinflammatory cytokine production and promoted IL-4-induced M2 expression. The progression of peritoneal macrophages extracted from Tollip-/- mice confirmed the effect of Tollip on macrophage polarization. Western blots showed that Tollip overexpression attenuated the upregulation of TLR pathway-associated targets in M1 macrophages. CONCLUSIONS MTC nanoparticles can be 'intelligent' carriers in immunotherapy. The modulation of Tollip expression in macrophages may be a novel treatment approach for IBD.
Collapse
Affiliation(s)
- Xiaoming Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xingxing Ren
- Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lifeng Zhou
- Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Gastroenterology, Nanchong Central Hospital, the Second Clinical Medical College of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Ke Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liangjun Deng
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Qing Qing
- Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jin Li
- Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fachao Zhi
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mingsong Li
- Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
24
|
The Potential Role of m6A in the Regulation of TBI-Induced BGA Dysfunction. Antioxidants (Basel) 2022; 11:antiox11081521. [PMID: 36009239 PMCID: PMC9405408 DOI: 10.3390/antiox11081521] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/31/2022] [Accepted: 08/02/2022] [Indexed: 02/01/2023] Open
Abstract
The brain–gut axis (BGA) is an important bidirectional communication pathway for the development, progress and interaction of many diseases between the brain and gut, but the mechanisms remain unclear, especially the post-transcriptional regulation of BGA after traumatic brain injury (TBI). RNA methylation is one of the most important modifications in post-transcriptional regulation. N6-methyladenosine (m6A), as the most abundant post-transcriptional modification of mRNA in eukaryotes, has recently been identified and characterized in both the brain and gut. The purpose of this review is to describe the pathophysiological changes in BGA after TBI, and then investigate the post-transcriptional bidirectional regulation mechanisms of TBI-induced BGA dysfunction. Here, we mainly focus on the characteristics of m6A RNA methylation in the post-TBI BGA, highlight the possible regulatory mechanisms of m6A modification in TBI-induced BGA dysfunction, and finally discuss the outcome of considering m6A as a therapeutic target to improve the recovery of the brain and gut dysfunction caused by TBI.
Collapse
|
25
|
Du C, Zhao Y, Wang K, Nan X, Chen R, Xiong B. Effects of Milk-Derived Extracellular Vesicles on the Colonic Transcriptome and Proteome in Murine Model. Nutrients 2022; 14:nu14153057. [PMID: 35893911 PMCID: PMC9332160 DOI: 10.3390/nu14153057] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/20/2022] [Accepted: 07/20/2022] [Indexed: 12/29/2022] Open
Abstract
Evidence shows that effective nutritional intervention can prevent or mitigate the risk and morbidity of inflammatory bowel disease (IBD). Bovine milk extracellular vesicles (mEVs), a major bioactive constituent of milk, play an important role in maintaining intestinal health. The aims of this study were to assess the effects of mEV pre-supplementation on the colonic transcriptome and proteome in dextran sulphate sodium (DSS)-induced acute colitis, in order to understand the underlying molecular mechanisms of mEV protection against acute colitis. Our results revealed that dietary mEV supplementation alleviated the severity of acute colitis, as evidenced by the reduced disease activity index scores, histological damage, and infiltration of inflammatory cells. In addition, transcriptome profiling analysis found that oral mEVs significantly reduced the expression of pro-inflammatory cytokines (IL-1β, IL-6, IL-17A and IL-33), chemokine ligands (CXCL1, CXCL2, CXCL3, CXCL5, CCL3 and CCL11) and chemokine receptors (CXCR2 and CCR3). Moreover, oral mEVs up-regulated 109 proteins and down-regulated 150 proteins in the DSS-induced murine model, which were involved in modulating amino acid metabolism and lipid metabolism. Collectively, this study might provide new insights for identifying potential targets for the therapeutic effects of mEVs on colitis.
Collapse
|
26
|
Ouyang W, Wu M, Wu A, Xiao H. Circular RNA_0001187 participates in the regulation of ulcerative colitis development via upregulating myeloid differentiation factor 88. Bioengineered 2022; 13:12863-12875. [PMID: 35609334 PMCID: PMC9275921 DOI: 10.1080/21655979.2022.2077572] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Circular RNA (circRNA) had been confirmed to participate in ulcerative colitis (UC) development. Circular RNA_0001187 (Circ_0001187) had been found to be overexpressed in patients with Crohn disease. Therefore, circ_0001187 might be an important circRNA regulating intestinal inflammatory diseases. However, the role and mechanism of circ_0001187 in UC progression remains unclear. The colonic mucosal tissues were obtained from 23 UC patients and 23 healthy normal controls. Tumor necrosis factor-α (TNF-α) was used to mimic UC cell model in vitro. Cell function was assessed by cell counting kit 8 assay, EdU assay, flow cytometry, ELISA assay and oxidative stress detection. RNA interaction was confirmed by dual-luciferase reporter assay and RIP assay. Serum exosomes were isolated by ultracentrifugation and identified by transmission electron microscope. Circ_0001187 was overexpressed in UC patients. Circ_0001187 knockdown enhanced the proliferation, while suppressed apoptosis, inflammation and oxidative stress of TNF-α-induced FHC cells. Circ_0001187 acted as miR-1236-3p sponge, and the effects of circ_0001187 downregulation on TNF-α-induced FHC cell injury were overturned by miR-1236-3p inhibitor. MYD88 was targeted by miR-1236-3p, and circ_0001187 sponged miR-1236-3p to regulate MYD88. MYD88 knockdown alleviated TNF-α-induced FHC cell injury, and its upregulation revoked the inhibition effect of miR-1236-3p on TNF-α-induced FHC cell injury. High expression of circ_0001187 also was observed in the serum exosomes of UC patients. Our data confirmed that circ_0001187 facilitated UC progression through miR-1236-3p/MYD88 axis, which might be a potential treatment and diagnosis biomarker for UC.
Collapse
Affiliation(s)
- Wei Ouyang
- Department of Oncology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou City, China
| | - Min Wu
- Department of Emergency, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou City, China
| | - Anshan Wu
- Department of Oncology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou City, China
| | - Heng Xiao
- Department of Anorectal, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou City, China
| |
Collapse
|
27
|
De Fazio L, Beghetti I, Bertuccio SN, Marsico C, Martini S, Masetti R, Pession A, Corvaglia L, Aceti A. Necrotizing Enterocolitis: Overview on In Vitro Models. Int J Mol Sci 2021; 22:6761. [PMID: 34201786 PMCID: PMC8268427 DOI: 10.3390/ijms22136761] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a gut inflammatory disorder which constitutes one of the leading causes of morbidity and mortality for preterm infants. The pathophysiology of NEC is yet to be fully understood; several observational studies have led to the identification of multiple factors involved in the pathophysiology of the disease, including gut immaturity and dysbiosis of the intestinal microbiome. Given the complex interactions between microbiota, enterocytes, and immune cells, and the limited access to fetal human tissues for experimental studies, animal models have long been essential to describe NEC mechanisms. However, at present there is no animal model perfectly mimicking human NEC; furthermore, the disease mechanisms appear too complex to be studied in single-cell cultures. Thus, researchers have developed new approaches in which intestinal epithelial cells are exposed to a combination of environmental and microbial factors which can potentially trigger NEC. In addition, organoids have gained increasing attention as promising models for studying NEC development. Currently, several in vitro models have been proposed and have contributed to describe the disease in deeper detail. In this paper, we will provide an updated review of available in vitro models of NEC and an overview of current knowledge regarding its molecular underpinnings.
Collapse
Affiliation(s)
- Luigia De Fazio
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Pediatric Oncology and Hematology “Lalla Seragnoli”, Pediatric Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Isadora Beghetti
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Neonatal Intensive Care Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Salvatore Nicola Bertuccio
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Pediatric Oncology and Hematology “Lalla Seragnoli”, Pediatric Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Concetta Marsico
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Neonatal Intensive Care Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Silvia Martini
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Neonatal Intensive Care Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Riccardo Masetti
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Pediatric Oncology and Hematology “Lalla Seragnoli”, Pediatric Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Andrea Pession
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Pediatric Oncology and Hematology “Lalla Seragnoli”, Pediatric Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Luigi Corvaglia
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Neonatal Intensive Care Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Arianna Aceti
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Neonatal Intensive Care Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| |
Collapse
|