1
|
Villanacci V, Del Sordo R, Lanzarotto F, Ricci C, Sidoni A, Manenti S, Mino S, Bugatti M, Bassotti G. Claudin-2: A marker for a better evaluation of histological mucosal healing in inflammatory bowel diseases. Dig Liver Dis 2025; 57:827-832. [PMID: 39155205 DOI: 10.1016/j.dld.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Histological mucosal healing has become a paramount target goal to achieve in the treatment of inflammatory bowel diseases. However, there is still a lack of agreement on the best way to reach this goal, since numerous histological scores are available worldwide. AIMS We investigated whether claudin-2, a member of claudin family involved in the regulation of intestinal tight junctions, might be useful to assess the presence of active disease in patients with inflammatory bowel diseases. METHODS Biopsies from 123 patients with ulcerative colitis, Crohn's disease, infectious colitides and irritable bowel syndrome patients where tested with immunohistochemistry for claudin-2. RESULTS Claudin-2 appeared to be a very sensitive marker of disease activity in inflammatory bowel diseases, but was negative in the other kinds of patients. In addition, immunohistochemistry for claudin-2 showed good reproducibility by different pathologists. CONCLUSIONS Should these findings be confirmed in more numerous cohorts of patients, and especially in those with minimal or focal residual disease activity, this simple assessment could be useful in the routine daily practice to facilitate the task of pathologists and clinicians in the diagnosis and management of patients with inflammatory bowel diseases.
Collapse
Affiliation(s)
- Vincenzo Villanacci
- Institute of Pathology, ASST Spedali Civili and University of Brescia, Brescia, Italy
| | - Rachele Del Sordo
- Department of Medicine and Surgery, Section of Anatomic Pathology and Histology, University of Perugia, Perugia, Italy.
| | - Francesco Lanzarotto
- Gastroenterology Unit, Department of Clinical and Experimental Sciences, Spedali Civili Hospital, University of Brescia, Brescia, Italy
| | - Chiara Ricci
- Gastroenterology Unit, Department of Clinical and Experimental Sciences, Spedali Civili Hospital, University of Brescia, Brescia, Italy
| | - Angelo Sidoni
- Department of Medicine and Surgery, Section of Anatomic Pathology and Histology, University of Perugia, Perugia, Italy
| | - Stefania Manenti
- Institute of Pathology, ASST Spedali Civili and University of Brescia, Brescia, Italy
| | - Sara Mino
- Institute of Pathology, ASST Spedali Civili and University of Brescia, Brescia, Italy
| | - Mattia Bugatti
- Institute of Pathology, ASST Spedali Civili and University of Brescia, Brescia, Italy
| | - Gabrio Bassotti
- Gastroenterology and Hepatology Section, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
2
|
Alqudah A, Qnais E, Gammoh O, Bseiso Y, Wedyan M, Alqudah M, Aljabali AAA, Tambuwala M. Exploring Scopoletin's Therapeutic Efficacy in DSS-Induced Ulcerative Colitis: Insights into Inflammatory Pathways, Immune Modulation, and Microbial Dynamics. Inflammation 2025; 48:575-589. [PMID: 38918333 PMCID: PMC12053357 DOI: 10.1007/s10753-024-02048-9] [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: 04/22/2024] [Accepted: 05/07/2024] [Indexed: 06/27/2024]
Abstract
This study aimed to investigate the therapeutic potential of scopoletin in ulcerative colitis, with a primary focus on its impact on crucial inflammatory pathways and immune responses. A male mouse model of DSS-induced colitis was employed with six distinct groups: a control group, a group subjected to DSS only, three groups treated with varying scopoletin doses, and the final group treated with dexamethasone. The investigation included an assessment of the effects of scopoletin on colitis symptoms, including alterations in body weight, Disease Activity Index (DAI), and histopathological changes in colonic tissue. Furthermore, this study scrutinized the influence of scopoletin on cytokine production, PPARγ and NF-κB expression, NLRP3 inflammasome, and the composition of intestinal bacteria. Scopoletin treatment yielded noteworthy improvements in DSS-induced colitis in mice, as evidenced by reduced weight loss and colonic shortening (p < 0.05, < 0.01, respectively). It effectively diminished TNF-α, IL-1β, and IL-12 cytokine levels (p < 0.01, p < 0.05), attenuated NLRP3 inflammasome activation and the associated cytokine release (p < 0.05, p < 0.01), and modulated the immune response by elevating PPARγ expression while suppressing NF-κB pathway activation (p < 0.05, p < 0.01). Additionally, scopoletin induced alterations in the gut microbiota composition, augmenting beneficial Lactobacillus and Bifidobacteria while reducing E. coli (p < 0.05). It also enhanced tight junction proteins, signifying an improvement in the intestinal barrier integrity (p < 0.05, < 0.01). Scopoletin is a promising therapeutic agent for managing ulcerative colitis, showing benefits that extend beyond mere anti-inflammatory actions to encompass regulatory effects on gut microbiota and restoration of intestinal integrity.
Collapse
Affiliation(s)
- Abdelrahim Alqudah
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa, Jordan
| | - Esam Qnais
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Omar Gammoh
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Yousra Bseiso
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Mohammed Wedyan
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Mohammad Alqudah
- Physiology Department, School of Medicine and Biomedical Sciences, Arabian Gulf University, Manama, Bahrain
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, 21163, Jordan
| | - Murtaza Tambuwala
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates.
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln, LN6 7TS, UK.
| |
Collapse
|
3
|
Liu J, Dai Y, Yang W, Chen ZY. Role of Mushroom Polysaccharides in Modulation of GI Homeostasis and Protection of GI Barrier. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6416-6441. [PMID: 40063730 PMCID: PMC11926878 DOI: 10.1021/acs.jafc.5c00745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Edible and medicinal mushroom polysaccharides (EMMPs) have been widely studied for their various biological activities. It has been shown that EMMPs could modulate microbiota in the large intestine and improve intestinal health. However, the role of EMMPs in protecting the gastric barrier, regulating gastric microbiota, and improving gastric health cannot be ignored. Hence, this review will elucidate the effect of EMMPs on gastric and intestinal barriers, with emphasis on the interaction of EMMPs with microbiota in maintaining overall gastrointestinal health. Additionally, this review highlights the gastroprotective effects and underlying mechanisms of EMMPs against gastric mucosa injury, gastritis, gastric ulcer, and gastric cancer. Furthermore, the effects of EMMPs on intestinal diseases, including inflammatory bowel disease, colorectal cancer, and intestinal infection, are also summarized. This review will also discuss the future perspective and challenges in the use of EMMPs as a dietary supplement or a nutraceutical in preventing and treating gastrointestinal diseases.
Collapse
Affiliation(s)
- Jianhui Liu
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong 999077, China
| | - Yi Dai
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Wenjian Yang
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Zhen-Yu Chen
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong 999077, China
| |
Collapse
|
4
|
Gavazza A, Mangiaterra S, Galosi L, Dottori A, Biagini L, Pengo G, Suchodolski J, Cerquetella M, Rossi G. Evaluation of serum and fecal parameters in cats with low-grade intestinal T-cell lymphoma (LGITCL). Res Vet Sci 2025; 184:105521. [PMID: 39753058 DOI: 10.1016/j.rvsc.2024.105521] [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: 01/08/2024] [Revised: 11/29/2024] [Accepted: 12/20/2024] [Indexed: 02/01/2025]
Abstract
Lymphoma is the most common neoplasia in the intestine of cats. According to ACVIM consensus statement, low-grade intestinal T-cell lymphoma (LGITCL) represents a monomorphic infiltration of the lamina propria or epithelium or both of cats with small, mature, neoplastic (clonal) T lymphocytes. Despite the importance as contributing factors of inheritance and environment in the pathogenesis of LGITCL, the chronic inflammatory status plays a fundamental role. The aim of the present study was to investigate possible diagnostic and prognostic parameters in LGITCL. Selected fecal bacteria and serum biomarkers (serum amyloid A [SAA]; citrulline; total protein; amylase; lipase; DGGR lipase; cholesterol; lipopolysaccharides [LPS], and zonulin) were evaluated in 12 cats (lymphoma group - LG) with histologically diagnosed low-grade intestinal T- cell lymphoma (LGITCL) and then compared with a control group consisting of 12 clinically healthy cats (CG). The evaluation of fecal bacterial population showed the significant decrease of Faecalibacterium spp. (P = 0,045) and Clostridium hiranonis (P = 0,0433) and a significant increase in E. coli (P = 0,045), Streptococcus spp. (P = 0,0003) and Turicibacter spp. (P = 0,0056) in the lymphoma group. Serology showed a significant decrease of total proteins (P = 0,092), amylase (P = 0,092) and cholesterol (P = 0,0112) in LG group compared to controls. The present results allowed the authors to state that in cats LGITCL is possibly associated to a change in the gastrointestinal environment and to a condition of protein losing enteropathy. Further studies with a larger cohort of patients are needed to confirm the present results and to point out other possible changes.
Collapse
Affiliation(s)
- Alessandra Gavazza
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024 Matelica, MC, Italy
| | - Sara Mangiaterra
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024 Matelica, MC, Italy.
| | - Livio Galosi
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024 Matelica, MC, Italy
| | - Alessia Dottori
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024 Matelica, MC, Italy
| | - Lucia Biagini
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024 Matelica, MC, Italy
| | | | - Jan Suchodolski
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843-4474, United States of America
| | - Matteo Cerquetella
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024 Matelica, MC, Italy
| | - Giacomo Rossi
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024 Matelica, MC, Italy
| |
Collapse
|
5
|
Soranno DE, Coopersmith CM, Brinkworth JF, Factora FNF, Muntean JH, Mythen MG, Raphael J, Shaw AD, Vachharajani V, Messer JS. A review of gut failure as a cause and consequence of critical illness. Crit Care 2025; 29:91. [PMID: 40011975 DOI: 10.1186/s13054-025-05309-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 02/05/2025] [Indexed: 02/28/2025] Open
Abstract
In critical illness, all elements of gut function are perturbed. Dysbiosis develops as the gut microbial community loses taxonomic diversity and new virulence factors appear. Intestinal permeability increases, allowing for translocation of bacteria and/or bacterial products. Epithelial function is altered at a cellular level and homeostasis of the epithelial monolayer is compromised by increased intestinal epithelial cell death and decreased proliferation. Gut immunity is impaired with simultaneous activation of maladaptive pro- and anti-inflammatory signals leading to both tissue damage and susceptibility to infections. Additionally, splanchnic vasoconstriction leads to decreased blood flow with local ischemic changes. Together, these interrelated elements of gastrointestinal dysfunction drive and then perpetuate multi-organ dysfunction syndrome. Despite the clear importance of maintaining gut homeostasis, there are very few reliable measures of gut function in critical illness. Further, while multiple therapeutic strategies have been proposed, most have not been shown to conclusively demonstrate benefit, and care is still largely supportive. The key role of the gut in critical illness was the subject of the tenth Perioperative Quality Initiative meeting, a conference to summarize the current state of the literature and identify key knowledge gaps for future study. This review is the product of that conference.
Collapse
Affiliation(s)
- Danielle E Soranno
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Craig M Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University, Atlanta, GA, USA
| | - Jessica F Brinkworth
- Department of Anthropology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Faith N F Factora
- Intensive Care and Resuscitation, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Julia H Muntean
- Intensive Care and Resuscitation, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Monty G Mythen
- Perioperative Medicine, University College London, London, England
| | - Jacob Raphael
- Anesthesiology and Perioperative Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Andrew D Shaw
- Intensive Care and Resuscitation, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Vidula Vachharajani
- Department of Pulmonary and Critical Care, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Jeannette S Messer
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
6
|
Wang D, Jiang Q, Li P, Yu C, Yuan R, Dong Z, Meng T, Hu F, Wang J, Yuan H. Orally Administrated Precision Nanomedicine for Restoring the Intestinal Barrier and Alleviating Inflammation in Treatment of Inflammatory Bowel Disease. ACS APPLIED MATERIALS & INTERFACES 2025; 17:10986-11001. [PMID: 39931937 DOI: 10.1021/acsami.4c19742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Inflammatory bowel disease (IBD) presents a significant challenge in healthcare, characterized by its chronicity and complex pathogenesis involving genetic, immune, and environmental factors. Current treatment modalities, including anti-inflammatory drugs, immunomodulators, and biologics, often lack sufficient efficacy and are accompanied by adverse effects, necessitating the urgent search for therapeutic approaches targeting mucosal barrier restoration and inflammation modulation. Precision nanomedicine emerges as a promising solution to directly address these challenges. This study introduces the development of a targeted sequential nanomedicine for precise IBD treatment. This innovative formulation combines a prodrug carrier containing quercetin to restore intestinal barrier integrity through the regulation of tight junctions and an anti-inflammatory agent dexamethasone acetate to alleviate inflammation. Surface modification with pectin enables colon-specific drug delivery, facilitated by degradation by colon-specific microbiota. Responsive drug release, controlled by reactive oxygen species-sensitive chemical bonds within the carrier, ensures both spatial and temporal accuracy. In vitro and in vivo investigations confirm the nanomedicine's favorable physicochemical properties, release kinetics, and therapeutic efficacy, elucidating potential underlying mechanisms. Oral administration of the nanomedicine shows promising results in restoring intestinal barrier function, reducing inflammation, and modulating the gut microbiota. Consequently, this study presents a promising nanomedicine candidate for advancing IBD treatment paradigms.
Collapse
Affiliation(s)
- Ding Wang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, P. R. China
| | - Qi Jiang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, P. R. China
| | - Peirong Li
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, P. R. China
| | - Caini Yu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, P. R. China
| | - Renxiang Yuan
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, P. R. China
| | - Zhefan Dong
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, P. R. China
| | - Tingting Meng
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, P. R. China
| | - Fuqiang Hu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, P. R. China
- Jinhua Institute of Zhejiang University, Jinhua 321299, P. R. China
| | - Jianwei Wang
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, P. R. China
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, P. R. China
- Jinhua Institute of Zhejiang University, Jinhua 321299, P. R. China
| |
Collapse
|
7
|
Lialios P, Alimperti S. Role of E-cadherin in epithelial barrier dysfunction: implications for bacterial infection, inflammation, and disease pathogenesis. Front Cell Infect Microbiol 2025; 15:1506636. [PMID: 40007608 PMCID: PMC11850337 DOI: 10.3389/fcimb.2025.1506636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/15/2025] [Indexed: 02/27/2025] Open
Abstract
Epithelial barriers serve as critical defense lines against microbial infiltration and maintain tissue homeostasis. E-cadherin, an essential component of adherens junctions, has emerged as a pivotal molecule that secures epithelial homeostasis. Lately, its pleiotropic role beyond barrier function, including its involvement in immune responses, has become more evident. Herein, we delve into the intricate relationship between (dys)regulation of epithelial homeostasis and the versatile functionality of E-cadherin, describing complex mechanisms that underlie barrier integrity and disruption in disease pathogenesis such as bacterial infection and inflammation, among others. Clinical implications of E-cadherin perturbations in host pathophysiology are emphasized; downregulation, proteolytic phenomena, abnormal localization/signaling and aberrant immune reactions are linked with a broad spectrum of pathology beyond infectious diseases. Finally, potential therapeutic interventions that may harness E-cadherin to mitigate barrier-associated tissue damage are explored. Overall, this review highlights the crucial role of E-cadherin in systemic health, offering insights that could pave the way for strategies to reinforce/restore barrier integrity and treat related diseases.
Collapse
Affiliation(s)
- Peter Lialios
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
- Center for Biological and Biomedical Engineering, Georgetown University, Washington, DC, United States
| | - Stella Alimperti
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
- Center for Biological and Biomedical Engineering, Georgetown University, Washington, DC, United States
| |
Collapse
|
8
|
Tao L, Zhang Q, Liu L, Wang K, Liu X, Li J, Zhao P. Magnolol preserves the integrity of the intestinal epithelial barrier and mitigates intestinal injury through activation of PPAR γ in COPD rat. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119068. [PMID: 39522848 DOI: 10.1016/j.jep.2024.119068] [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: 09/26/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Magnolia officinalis Rehder & E.H. Wilson is traditionally used in the treatment of gastrointestinal disorders, diarrhea, and cough. Its main active ingredient, magnolol, exhibits protective effects on the lungs and gastrointestinal tract, including the inhibition of inflammation in these organs. AIM OF THE STUDY This work aims to explore the molecular mechanism by which magnolol suppressed Chronic obstructive pulmonary disease (COPD) intestinal damage by improving the intestinal epithelial barrier. MATERIALS AND METHODS The study focused on investigating the mitigation effect of magnolol on intestinal injury and epithelial barrier in a COPD rat. Caco-2 cells were induced with TNF-α or IL-1β to establish the barrier injury model in order to explore the direct protective effect of magnolol on the intestinal barrier and elucidate the molecular mechanism by which it activates peroxisome proliferators-activated receptors-γ (PPARγ). RESULTS Magnolol significantly improves pulmonary function and tissue damage in COPD rats by inhibiting inflammation, protease imbalance, and oxidative stress. It also suppresses colon tissue damage and inflammation, and protects colon epithelial barrier function by suppressing the decline of tight junction proteins, reducing colon epithelial permeability. In Caco-2 cells, magnolol directly reduces monolayer permeability, increases TEER, and upregulates tight junction protein expression induced by TNF-α or IL-1β. Drug Affinity Responsive Target Stability (DARTS) and thermal shift assays show that magnolol effectively binds to SRC, activating PPARγ signaling in Caco-2 cells and colon tissues of COPD rats. Furthermore, magnolol enhances the binding of PPARγ and RXRα, promoting their activation and entry into the nucleus. The PPARγ inhibitor GW9662 can reverse the effects of magnolol on PPARγ activation and tight junction protein upregulation in IL-1β or TNF-α induced Caco-2 cells. CONCLUSIONS This work demonstrates that magnolol enhances lung and intestinal functions in COPD rats, and elucidates its mechanism of action in protecting the intestinal epithelial barrier by activating PPARγ.
Collapse
Affiliation(s)
- Liuying Tao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, 450046, Henan Province, China; Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, China
| | - Qin Zhang
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, 450046, Henan Province, China; Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450000, China
| | - Lan Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, 450046, Henan Province, China; Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, China
| | - Kun Wang
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, 450046, Henan Province, China; Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, China
| | - Xuefang Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, 450046, Henan Province, China; Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, China
| | - Jiansheng Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, 450046, Henan Province, China; Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450000, China
| | - Peng Zhao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, 450046, Henan Province, China; Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450000, China.
| |
Collapse
|
9
|
Deng WQ, Ye ZH, Tang Z, Zhang XL, Lu JJ. Beyond cancer: The potential application of CD47-based therapy in non-cancer diseases. Acta Pharm Sin B 2025; 15:757-791. [PMID: 40177549 PMCID: PMC11959971 DOI: 10.1016/j.apsb.2024.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/10/2024] [Accepted: 11/22/2024] [Indexed: 04/05/2025] Open
Abstract
CD47 is an immune checkpoint widely regarded as a 'don't eat me' signal. CD47-based anti-cancer therapy has received considerable attention, with a significant number of clinical trials conducted. While anti-cancer therapies based on CD47 remain a focal point of interest among researchers, it is noteworthy that an increasing number of studies have found that CD47-based therapy ameliorated the pathological status of non-cancer diseases. This review aims to provide an overview of the recent progress in comprehending the role of CD47-based therapy in non-cancer diseases, including diseases of the circulatory system, nervous system, digestive system, and so on. Furthermore, we sought to delineate the promising mechanisms of CD47-based therapy in treating non-cancer diseases. Our findings suggest that CD47-based agents may exert their effect by regulating phagocytosis, regulating T cells, dendritic cells, and neutrophils, and regulating the secretion of cytokines and chemokines. Additionally, we put forward the orientation of further research to bring to light the potential of CD47 and its binding partners as a target in non-cancer diseases.
Collapse
Affiliation(s)
- Wei-Qing Deng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Zi-Han Ye
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Zhenghai Tang
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China
| | - Xiao-Lei Zhang
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macao 999078, China
| |
Collapse
|
10
|
Tao L, Zhang Q, Liu L, Wang K, Wang J, Liu X, Zhao P, Li J. Inhibition of AhR disrupts intestinal epithelial barrier and induces intestinal injury by activating NF-κB in COPD. FASEB J 2024; 38:e70256. [PMID: 39679871 DOI: 10.1096/fj.202402320r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/25/2024] [Accepted: 12/05/2024] [Indexed: 12/17/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is frequently associated with intestinal comorbidities. Damage to the intestinal barrier plays a crucial role in these disorders, leading to increased intestinal and systemic inflammation, and thereby promoting the progression of COPD. This study aims to investigate the mechanism of intestinal epithelial barrier damage, focusing on the roles of the Aryl hydrocarbon Receptor (AhR) and NF-κB in COPD-related intestinal damage. A COPD rat model was induced by cigarette smoke and bacterial infection, while Caco-2/HT29 intestinal epithelial cells were treated with TNF-α or IL-1β to assess intestinal disorder and the underlying mechanisms of barrier damage. COPD rats exhibited significant lung function decline, pathological damage, and inflammatory response in lung tissues. Additionally, significant intestinal injury was observed, accompanied by pronounced colonic pathological damage, an enhanced inflammatory response, and intestinal barrier disruption. This was evidenced by decreased expression of apical junction proteins and elevated serum diamine oxidase levels. Pro-inflammatory cytokines TNF-α or IL-1β significantly downregulated the expression of apical junction proteins in Caco-2/HT29 cells, reduced transepithelial electrical resistance of Caco-2 cells, and increased FD-4 permeability. Moreover, TNF-α or IL-1β induction activated NF-κB in Caco-2/HT29 cells, with a similar activation observed in the colonic tissues of COPD rats. The NF-κB inhibitor PDTC suppressed this activation and protected against intestinal epithelial barrier damage. Furthermore, AhR inhibition was observed both in vitro and in vivo. The AhR activator FICZ inhibited NF-κB activation and mitigated intestinal epithelial barrier damage, whereas the AhR inhibitor CH223191 inhibited AhR and exacerbated intestinal epithelial barrier damage by facilitating NF-κB activation. However, the NF-κB inhibitor PDTC did not significantly affect AhR. Additionally, TNF-α/IL-1β inhibited the binding of AhR and p-NF-κB. Consequently, AhR inhibition can downregulate the expression of apical junction proteins, probably through activation of NF-κB signaling leading to intestinal epithelial barrier damage. This study confirmed the presence of lesions in the lungs and intestines of COPD rats, as well as the associated damage to the intestinal epithelial barrier. The inhibition of AhR followed by the activation of NF-κB has been identified as a critical mechanism underlying the injury to the intestinal epithelial barrier.
Collapse
Affiliation(s)
- Liuying Tao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Qin Zhang
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Lan Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Kun Wang
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Juanhui Wang
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xuefang Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Peng Zhao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jiansheng Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
11
|
Zhao X, Chen X, Yue C. Rutin Ameliorates Inflammation and Oxidative Stress in Ulcerative Colitis by Inhibiting NLRP3 Inflammasome Signaling Pathway. Cell Biochem Biophys 2024; 82:3715-3726. [PMID: 39138797 PMCID: PMC11576901 DOI: 10.1007/s12013-024-01459-7] [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] [Accepted: 07/23/2024] [Indexed: 08/15/2024]
Abstract
Ulcerative colitis (UC) is an idiopathic inflammatory disease. We intend to explore the mechanism of Rutin in the therapy of UC. Disease activity index (DAI) and hematoxylin-eosin staining were employed to assess therapeutic effect of Rutin on dextran sulfate sodium-stimulated mice. The proliferation was detected by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide assay. Oxidative stress (OS) was assessed by measuring reactive oxygen species (ROS), malondialdehyde (MDA), and superoxide dismutase (SOD). Inflammatory factors were detected using enzyme-linked immunosorbent assay and immunofluorescence staining. mRNA and protein expressions were detected by real-time quantitative polymerase chain reaction and immunoblotting assay. Rutin decreased DAI scores and ameliorated pathological damage in UC mice with decreased levels of inflammatory factors. Rutin recovered the inhibited proliferation of fetal human colon cells caused by lipopolysaccharide. Rutin inhibited OS by reducing ROS and MDA, while enhancing SOD activity in LPS-induced fetal human colon cells. Rutin inhibited NLRP3 inflammasome in UC mice and cell model. Silencing NLRP3 enhanced the inhibitory effect of Rutin on OS in lipopolysaccharide-induced fetal human colon cells. Conversely, NLRP3 overexpression reversed the restraining role of Rutin in OS. Rutin ameliorates UC by inhibiting inflammation and OS through suppressing NLRP3 inflammasome.
Collapse
Affiliation(s)
- Xiangdong Zhao
- Department of Anorectal, Shenzhen Traditional Chinese Medicine Hospital, No. 1, Fuhua Road, Nanyuan Street, Futian District, Shenzhen, 518003, Guangdong, China
| | - Xiaochao Chen
- Department of Anorectal, Chengdu Anorectal Hospital, No.152, Daqiang East Street, Qingyang District, Chengdu, 610015, Sichuan, China
| | - Chaochi Yue
- Department of Traditional Chinese Medicine, The Affiliated Hospital, Southwest Medical University, No. 25, Taiping Street, Jiangyang District, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
12
|
Kang J, Xie W, Wu L, Liu Y, Xu Y, Xu Y, Mai Y, Peng L, Huang B, Guo S, Luo S. The ethanolic extract of domesticated Amauroderma rugosum alleviated DSS-induced ulcerative colitis via repairing the intestinal barrier. Food Sci Biotechnol 2024; 33:3335-3345. [PMID: 39328223 PMCID: PMC11422322 DOI: 10.1007/s10068-024-01565-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 01/21/2024] [Accepted: 03/15/2024] [Indexed: 09/28/2024] Open
Abstract
Amauroderma rugosum (Blume and T. Nees) Torrend (Ganodermataceae) (A. rugosum) has been found to have anti-inflammatory ability in previous studies. The present study aimed to verify the therapeutic benefits of A. rugosum in the treatment of ulcerative colitis and to investigate its underlying mechanism of action. Acute experimental ulcerative colitis was induced by feeding the mice drinking water supplemented with dextran sodium sulfate (DSS). The findings indicated that the ethanolic extract of domesticated A. rugosum exhibited therapeutic efficacy comparable to Salazosulfapyridine (SASP) in mitigating clinical symptoms and the pathological score of the colon. Furthermore, A. rugosum exhibited the capacity to enhance the expression of tight junction (TJ) proteins, while concurrently decreasing the levels of TNF-ɑ and IL-6. A noteworthy finding is that it exhibited the capability to diminish the nuclear translocation of NF-κB p65. In conclusion, A. rugosum attenuates DSS-induced ulcerative colitis by enhancing intestinal barrier function and inhibiting mucosal inflammation. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-024-01565-5.
Collapse
Affiliation(s)
- Jianyuan Kang
- Institute of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 1 Fuhua Road, Futian District, Shenzhen, 518033 Guangdong China
| | - Weicang Xie
- Institute of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 1 Fuhua Road, Futian District, Shenzhen, 518033 Guangdong China
| | - Lingping Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Yuanyuan Liu
- Yantian District Maternity and Child Healthcare Hospital, Shenzhen, 518081 China
| | - Youcai Xu
- Institute of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 1 Fuhua Road, Futian District, Shenzhen, 518033 Guangdong China
| | - Yifei Xu
- Institute of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 1 Fuhua Road, Futian District, Shenzhen, 518033 Guangdong China
| | - Yanzhen Mai
- Huizhou Health Sciences Polytechnic, Huizhou, 516025 China
| | - Lisheng Peng
- Institute of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 1 Fuhua Road, Futian District, Shenzhen, 518033 Guangdong China
| | - Bin Huang
- Institute of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 1 Fuhua Road, Futian District, Shenzhen, 518033 Guangdong China
| | - Shaoju Guo
- Institute of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 1 Fuhua Road, Futian District, Shenzhen, 518033 Guangdong China
| | - Shuang Luo
- Institute of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 1 Fuhua Road, Futian District, Shenzhen, 518033 Guangdong China
- Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guian New Area, Guiyang, 561113 Guizhou China
| |
Collapse
|
13
|
Liu Q, Yang Y, Pan M, Shi K, Mo D, Li Y, Wang M, Guo L, Qian Z. Camptothecin multifunctional nanoparticles effectively achieve a balance between the efficacy of breast cancer treatment and the preservation of intestinal homeostasis. Bioact Mater 2024; 41:413-426. [PMID: 39184827 PMCID: PMC11342206 DOI: 10.1016/j.bioactmat.2024.07.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/27/2024] [Accepted: 07/27/2024] [Indexed: 08/27/2024] Open
Abstract
Camptothecin (CPT) exhibits potent antitumor activity; however, its clinical application is limited by significant gastrointestinal adverse effects (GAEs). Although the severity of GAEs associated with CPT derivatives has decreased, the incidence rate of these adverse effects has remained high. CPT multifunctional nanoparticles (PCRHNs) have the potential to increase the efficacy of CPT while reducing side effects in major target organs; however, the impact of PCRHNs on the GAEs from CPT remains uncertain. Here, we investigated the therapeutic effects of PCRHNs and different doses of CPT and examined their impacts on the intestinal barrier and the intestinal microbiota. We found that the therapeutic efficacy of PCRHNs + Laser treatment was superior to that of high-dose CPT, and PCRHNs + Laser treatment also provided greater benefits by helping maintain intestinal barrier integrity, intestinal microbiota diversity, and intestinal microbiota abundance. In summary, compared to high-dose CPT treatment, PCRHNs + Laser treatment can effectively balance therapeutic effects and GAEs. A high dose of CPT promotes the enrichment of the pathogenic bacteria Escherichia-Shigella, which may be attributed to diarrhea caused by CPT, thus leading to a reduction in microbial burden; additionally, Escherichia-Shigella rapidly grows and occupies niches previously occupied by other bacteria that are lost due to diarrhea. PCRHNs + Laser treatment increased the abundance of Lactobacillus (probiotics), possibly due to the photothermal effect of the PCRHNs. This effect increased catalase activity, thus facilitating the conversion of hydrogen peroxide into oxygen within tumors and increasing oxygen levels in the body, which is conducive to the growth of facultative anaerobic bacteria.
Collapse
Affiliation(s)
- Qingya Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yun Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meng Pan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kun Shi
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dong Mo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yicong Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meng Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Linfeng Guo
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhiyong Qian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
14
|
Liebing E, Krug SM, Neurath MF, Siegmund B, Becker C. Wall of Resilience: How the Intestinal Epithelium Prevents Inflammatory Onslaught in the Gut. Cell Mol Gastroenterol Hepatol 2024; 19:101423. [PMID: 39461590 PMCID: PMC11720114 DOI: 10.1016/j.jcmgh.2024.101423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024]
Abstract
The intestinal epithelium forms the boundary between the intestinal immune system in the lamina propria and the outside world, the intestinal lumen, which contains a diverse array of microbial and environmental antigens. Composed of specialized cells, this epithelial monolayer has an exceptional turnover rate. Differentiated epithelial cells are released into the intestinal lumen within a few days, at the villus tip, a process that requires strict regulation. Dysfunction of the epithelial barrier increases the intestinal permeability and paves the way for luminal antigens to pass into the intestinal serosa. Stem cells at the bottom of Lieberkühn crypts provide a constant supply of mature epithelial cells. Differentiated intestinal epithelial cells exhibit a diverse array of mechanisms that enable communication with surrounding cells, fortification against microorganisms, and orchestration of nutrient absorption and hormonal balance. Furthermore, tight junctions regulate paracellular permeability properties, and their disruption can lead to an impairment of the intestinal barrier, allowing inflammation to develop or further progress. Intestinal epithelial cells provide a communication platform through which they maintain homeostasis with a spectrum of entities including immune cells, neuronal cells, and connective tissue cells. This homeostasis can be disrupted in disease, such as inflammatory bowel disease. Patients suffering from inflammatory bowel disease show an impaired gut barrier, dysregulated cellular communication, and aberrant proliferation and demise of cells. This review summarizes the individual cellular and molecular mechanisms pivotal for upholding the integrity of the intestinal epithelial barrier and shows how these can be disrupted in diseases, such as inflammatory bowel disease.
Collapse
Affiliation(s)
- Eva Liebing
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Susanne M Krug
- Clinical Physiology/Nutritional Medicine, Charité-Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Markus F Neurath
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Britta Siegmund
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christoph Becker
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie, Erlangen, Germany.
| |
Collapse
|
15
|
Ariturk LA, Cilingir S, Kolgazi M, Elmas M, Arbak S, Yapislar H. Docosahexaenoic acid (DHA) alleviates inflammation and damage induced by experimental colitis. Eur J Nutr 2024; 63:2801-2813. [PMID: 39105785 PMCID: PMC11490523 DOI: 10.1007/s00394-024-03468-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 07/05/2024] [Indexed: 08/07/2024]
Abstract
PURPOSE Inflammatory bowel diseases (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), are chronic gastrointestinal disorders associated with significant morbidity and complications. This study investigates the therapeutic potential of docosahexaenoic acid (DHA) in a trinitrobenzene sulfonic acid (TNBS) induced colitis model, focusing on inflammation, oxidative stress, and intestinal membrane permeability. METHODS Wistar albino rats were divided into Control, Colitis, and Colitis + DHA groups (n = 8-10/group). The Colitis and Colitis + DHA groups received TNBS intrarectally, while the Control group received saline. DHA (600 mg/kg/day) or saline was administered via gavage for six weeks. Macroscopic and microscopic evaluations of colon tissues were conducted. Parameters including occludin and ZO-1 expressions, myeloperoxidase (MPO) activity, malondialdehyde (MDA), glutathione (GSH), total antioxidant status (TAS), total oxidant status (TOS), Interleukin-6 (IL-6), and tumor necrosis factor alpha (TNF-α) levels were measured in colon tissues. RESULTS Colitis induction led to significantly higher macroscopic and microscopic damage scores, elevated TOS levels, reduced occludin and ZO-1 intensity, decreased mucosal thickness, and TAS levels compared to the Control group (p < 0.001). DHA administration significantly ameliorated these parameters (p < 0.001). MPO, MDA, TNF-α, and IL-6 levels were elevated in the Colitis group but significantly reduced in the DHA-treated group (p < 0.001 for MPO, MDA; p < 0.05 for TNF-α and IL-6). CONCLUSION DHA demonstrated antioxidant and anti-inflammatory effects by reducing reactive oxygen species production, enhancing TAS capacity, preserving GSH content, decreasing proinflammatory cytokine levels, preventing neutrophil infiltration, reducing shedding in colon epithelium, and improving gland structure and mucosal membrane integrity. DHA also upregulated the expressions of occludin and ZO-1, critical for barrier function. Thus, DHA administration may offer a therapeutic strategy or supplement to mitigate colitis-induced adverse effects.
Collapse
Affiliation(s)
- Leman Arslan Ariturk
- Faculty of Medicine, Department of Physiology, Marmara University, Istanbul, Turkey
| | - Sumeyye Cilingir
- Faculty of Medicine, Department of Physiology, Acibadem Mehmet Ali Adinlar University, Istanbul, Turkey
| | - Meltem Kolgazi
- Faculty of Medicine, Department of Physiology, Acibadem Mehmet Ali Adinlar University, Istanbul, Turkey
| | - Merve Elmas
- Faculty of Medicine, Department of Histology&Embriology, Acibadem Mehmet Ali Adinlar University, Istanbul, Turkey
| | - Serap Arbak
- Faculty of Medicine, Department of Histology&Embriology, Acibadem Mehmet Ali Adinlar University, Istanbul, Turkey
| | - Hande Yapislar
- Faculty of Medicine, Department of Physiology, Acibadem Mehmet Ali Adinlar University, Istanbul, Turkey.
| |
Collapse
|
16
|
Górecka A, Jura-Półtorak A, Koźma EM, Szeremeta A, Olczyk K, Komosińska-Vassev K. Biochemical Modulators of Tight Junctions (TJs): Occludin, Claudin-2 and Zonulin as Biomarkers of Intestinal Barrier Leakage in the Diagnosis and Assessment of Inflammatory Bowel Disease Progression. Molecules 2024; 29:4577. [PMID: 39407507 PMCID: PMC11478261 DOI: 10.3390/molecules29194577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Considering the increasing worldwide prevalence of inflammatory bowel disease (IBD), the early diagnosis of this disease is extremely important. However, non-invasive diagnostic methods remain limited, while invasive techniques are the most commonly used in daily practice. Therefore, there is a serious need to find new non-invasive biomarkers of IBD. METHODS The serum profiles of occludin, claudin-2, and zonulin were assessed in IBD patients using the ELISA method. The levels of the analyzed biomarkers were measured before and after a year of anti-inflammatory treatment, which was a tumor necrosis factor α (TNF-α) inhibitor (adalimumab) in patients with ulcerative colitis (UC) and conventional therapy in patients with Crohn's disease (CD). RESULTS In IBD patients, the serum level of occludin (p < 0.001) decreased compared to healthy individuals, while the level of claudin-2 (p < 0.001) increased. Additionally, zonulin (p < 0.01) concentration increased in CD patients compared to the control group. The highest diagnostic ability was presented by occludin measurements with the area under the curve (AUC) of 0.959 (95% CI 0.907-1) in UC and 0.948 (95% CI 0.879-1) in CD. Claudin-2 also demonstrated very good ability in diagnosing UC and CD with AUC values of 0.864 (95% CI 0.776-0.952) and 0.896 (95% CI 0.792-0.999), respectively. The ability of zonulin to diagnose CD was estimated as good with an AUC of 0.74 (95% CI 0.598-0.881). Moreover, a significant correlation was identified between C-reactive protein (CRP), claudin-2 (r = -0.37; p < 0.05), and zonulin (r = -0.44; p < 0.05) in UC patients. Treatment with adalimumab improved the level of occludin, claudin-2, and zonulin in UC patients, while anti-inflammatory conventional therapy decreased the concentration of zonulin in CD. CONCLUSIONS Occludin and claudin-2 measurements present significant utility in diagnosing both UC and CD, while zonulin assessments may be useful in CD diagnosis. Additionally, claudin-2 and zonulin measurements may be helpful in evaluating the intensity of the inflammatory process. Anti-TNF-α treatment improved the value of occludin, claudin-2, and zonulin, indicating its beneficial effect on the integrity of tight junctions in UC.
Collapse
Affiliation(s)
- Aleksandra Górecka
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland; (A.J.-P.); (E.M.K.); (A.S.); (K.O.); (K.K.-V.)
| | | | | | | | | | | |
Collapse
|
17
|
Ren X, Shi B, Chang Z, Zhang J, Wang S, Liu R, Sang M, Dong H, Wu Q. Relationship between pathogenic E.coli O78-induced intestinal epithelial barrier damage and Zonulin expression levels in yaks. Front Cell Infect Microbiol 2024; 14:1456356. [PMID: 39376662 PMCID: PMC11456573 DOI: 10.3389/fcimb.2024.1456356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/03/2024] [Indexed: 10/09/2024] Open
Abstract
To explore whether the intestinal damage of yak colibacillosis resulted from the regulation of Zonulin expression by its pathogenic bacteria, the overexpression and interference plasmids of Zonulin were designed and cultured in Tranwell after cell transfection. Then qRT-PCR and Western blot were used to detect the results of cell transfection, 200 mL 1×105 CFU/mL E.coli O78 was added for 4 hours, transmembrane resistance was measured by transmembrane resistance meter, FD4 fluorescence concentration in the lower chamber was detected by enzyme labeling instrument, bacterial translocation was measured by CFU counting method, and epithelial mucin (MUC1, MUC2) and tight junction protein (FABP2, Occludin, ZO-1) were detected by qRT-PCR. Results The Zonulin gene overexpression and knockout cell lines were successfully constructed, the TEER value of the barrier of Zonulin overexpression cell lines began to decrease at 1 h after the addition of E.coli O78 and reached the lowest value at 4 h, and the TEER value of Zonulin interference cell lines decreased within 1-4 h after the addition of E.coli O78. At 4 h, the FD4 passing capacity of Zonulin overexpression cell lines was significantly higher than that of interfering cell lines, reaching twice as much as siRNA-1. The amount of bacterial translocation in overexpressed cell lines increased rapidly within 1-4 h, and the concentration of E.coli in the lower chamber was significantly higher than that in the siRNA-1 group at 4 h, but there was no significant change in the siRNA-1 group in the 1-4 h. There was no significant change in the mRNA level of MUC1 in Zonulin overexpression and interference cell lines after the addition of E.coli O78. In the overexpression group, the mRNA levels of MUC2, Occludin, and ZO-1 were significantly decreased, and the mRNA level of FABP2 was increased considerably. These results suggest stimulate epithelial cells to secrete Zonulin protein. Many Zonulin proteins regulate the opening of tight junction structures, reduce the transmembrane resistance of the cell barrier, and improve the permeability of the cell barrier and the amount of bacterial translocation.
Collapse
Affiliation(s)
- Xiaoli Ren
- Key Laboratory of Clinical Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| | - Bin Shi
- Key Laboratory of Clinical Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, China
- Institute of Animal Science, Xizang Academy of Agricultural and Animal Husbandry Sciences, Xizang Lhasa, China
| | - Zhenyu Chang
- Key Laboratory of Clinical Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| | - Jingbo Zhang
- Key Laboratory of Clinical Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| | - Shuo Wang
- Key Laboratory of Clinical Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| | - Ruidong Liu
- Key Laboratory of Clinical Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| | - Mudan Sang
- Key Laboratory of Clinical Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| | - Hailong Dong
- Key Laboratory of Clinical Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| | - Qingxia Wu
- Key Laboratory of Clinical Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| |
Collapse
|
18
|
Erkert L, Gamez-Belmonte R, Kabisch M, Schödel L, Patankar JV, Gonzalez-Acera M, Mahapatro M, Bao LL, Plattner C, Kühl AA, Shen J, Serneels L, De Strooper B, Neurath MF, Wirtz S, Becker C. Alzheimer's disease-related presenilins are key to intestinal epithelial cell function and gut immune homoeostasis. Gut 2024; 73:1618-1631. [PMID: 38684238 DOI: 10.1136/gutjnl-2023-331622] [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: 11/24/2023] [Accepted: 04/16/2024] [Indexed: 05/02/2024]
Abstract
OBJECTIVE Mutations in presenilin genes are the major cause of Alzheimer's disease. However, little is known about their expression and function in the gut. In this study, we identify the presenilins Psen1 and Psen2 as key molecules that maintain intestinal homoeostasis. DESIGN Human inflammatory bowel disease (IBD) and control samples were analysed for Psen1 expression. Newly generated intestinal epithelium-specific Psen1-deficient, Psen2-deficient and inducible Psen1/Psen2 double-deficient mice were used to dissect the functional role of presenilins in intestinal homoeostasis. RESULTS Psen1 expression was regulated in experimental gut inflammation and in patients with IBD. Induced deletion of Psen1 and Psen2 in mice caused rapid weight loss and spontaneous development of intestinal inflammation. Mice exhibited epithelial barrier disruption with bacterial translocation and deregulation of key pathways for nutrient uptake. Wasting disease was independent of gut inflammation and dysbiosis, as depletion of microbiota rescued Psen-deficient animals from spontaneous colitis development but not from weight loss. On a molecular level, intestinal epithelial cells lacking Psen showed impaired Notch signalling and dysregulated epithelial differentiation. CONCLUSION Overall, our study provides evidence that Psen1 and Psen2 are important guardians of intestinal homoeostasis and future targets for barrier-promoting therapeutic strategies in IBD.
Collapse
Affiliation(s)
- Lena Erkert
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
| | - Reyes Gamez-Belmonte
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
| | - Melanie Kabisch
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
| | - Lena Schödel
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
| | - Jay V Patankar
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Miguel Gonzalez-Acera
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
| | - Mousumi Mahapatro
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
| | - Li-Li Bao
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
| | - Christina Plattner
- Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Anja A Kühl
- iPATH.Berlin, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jie Shen
- Department of Neurology, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lutgarde Serneels
- VIB Center for Brain and Disease Research, KU Leuven, Leuven, Belgium
| | - Bart De Strooper
- VIB Center for Brain and Disease Research, KU Leuven, Leuven, Belgium
- UK Dementia Research Institute@UCL, University College London, London, UK
| | - Markus F Neurath
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Stefan Wirtz
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Christoph Becker
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| |
Collapse
|
19
|
Qian G, Zang H, Tang J, Zhang H, Yu J, Jia H, Zhang X, Zhou J. Lactobacillus gasseri ATCC33323 affects the intestinal mucosal barrier to ameliorate DSS-induced colitis through the NR1I3-mediated regulation of E-cadherin. PLoS Pathog 2024; 20:e1012541. [PMID: 39250508 PMCID: PMC11412683 DOI: 10.1371/journal.ppat.1012541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 09/19/2024] [Accepted: 08/29/2024] [Indexed: 09/11/2024] Open
Abstract
Inflammatory bowel disease (IBD) is an immune system disorder primarily characterized by colitis, the exact etiology of which remains unclear. Traditional treatment approaches currently yield limited efficacy and are associated with significant side effects. Extensive research has indicated the potent therapeutic effects of probiotics, particularly Lactobacillus strains, in managing colitis. However, the mechanisms through which Lactobacillus strains ameliorate colitis require further exploration. In our study, we selected Lactobacillus gasseri ATCC33323 from the intestinal microbiota to elucidate the specific mechanisms involved in modulation of colitis. Experimental findings in a DSS-induced colitis mouse model revealed that L. gasseri ATCC33323 significantly improved physiological damage in colitic mice, reduced the severity of colonic inflammation, decreased the production of inflammatory factors, and preserved the integrity of the intestinal epithelial structure and function. It also maintained the expression and localization of adhesive proteins while improving intestinal barrier permeability and restoring dysbiosis in the gut microbiota. E-cadherin, a critical adhesive protein, plays a pivotal role in this protective mechanism. Knocking down E-cadherin expression within the mouse intestinal tract significantly attenuated the ability of L. gasseri ATCC33323 to regulate colitis, thus confirming its protective role through E-cadherin. Finally, transcriptional analysis and in vitro experiments revealed that L. gasseri ATCC33323 regulates CDH1 transcription by affecting NR1I3, thereby promoting E-cadherin expression. These findings contribute to a better understanding of the specific mechanisms by which Lactobacillus strains alleviate colitis, offering new insights for the potential use of L. gasseri as an alternative therapy for IBD, particularly in dietary supplementation.
Collapse
Affiliation(s)
- Guanru Qian
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Hui Zang
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Jingtong Tang
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Hao Zhang
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Jiankang Yu
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Huibiao Jia
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Xinzhuang Zhang
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Jianping Zhou
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| |
Collapse
|
20
|
Kong C, Yang M, Yue N, Zhang Y, Tian C, Wei D, Shi R, Yao J, Wang L, Li D. Restore Intestinal Barrier Integrity: An Approach for Inflammatory Bowel Disease Therapy. J Inflamm Res 2024; 17:5389-5413. [PMID: 39161679 PMCID: PMC11330754 DOI: 10.2147/jir.s470520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/02/2024] [Indexed: 08/21/2024] Open
Abstract
The intestinal barrier maintained by various types of columnar epithelial cells, plays a crucial role in regulating the interactions between the intestinal contents (such as the intestinal microbiota), the immune system, and other components. Dysfunction of the intestinal mucosa is a significant pathophysiological mechanism and clinical manifestation of inflammatory bowel disease (IBD). However, current therapies for IBD primarily focus on suppressing inflammation, and no disease-modifying treatments specifically target the epithelial barrier. Given the side effects associated with chronic immunotherapy, effective alternative therapies that promote mucosal healing are highly attractive. In this review, we examined the function of intestinal epithelial barrier function and the mechanisms of behind its disruption in IBD. We illustrated the complex process of intestinal mucosal healing and proposed therapeutic approaches to promote mucosal healing strategies in IBD. These included the application of stem cell transplantation and organ-like tissue engineering approaches to generate new intestinal tissue. Finally, we discussed potential strategies to restore the function of the intestinal barrier as a treatment for IBD.
Collapse
Affiliation(s)
- Chen Kong
- The Second Clinical Medical College, Jinan University; Shenzhen, Guangdong, People’s Republic of China
| | - Meifeng Yang
- Department of Hematology, Yantian District People’s Hospital, Shenzhen, Guangdong, People’s Republic of China
| | - Ningning Yue
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou, Guangdong, People’s Republic of China
| | - Chengmei Tian
- Department of Emergency, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, People’s Republic of China
| | - Daoru Wei
- Department of Rehabilitation, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, People’s Republic of China
| | - Ruiyue Shi
- The Second Clinical Medical College, Jinan University; Shenzhen, Guangdong, People’s Republic of China
| | - Jun Yao
- The Second Clinical Medical College, Jinan University; Shenzhen, Guangdong, People’s Republic of China
| | - Lisheng Wang
- The Second Clinical Medical College, Jinan University; Shenzhen, Guangdong, People’s Republic of China
| | - Defeng Li
- The Second Clinical Medical College, Jinan University; Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
21
|
Kim KY, Kang YM, Lee A, Kim YJ, Kim KH, Hwang YH. Hydroethanolic Extract of Lepidium apetalum Willdenow Alleviates Dextran Sulfate Sodium-Induced Colitis by Enhancing Intestinal Barrier Integrity and Inhibiting Oxidative Stress and Inflammasome Activation. Antioxidants (Basel) 2024; 13:795. [PMID: 39061864 PMCID: PMC11273485 DOI: 10.3390/antiox13070795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
The prevalence of ulcerative colitis (UC) has surged in Asian nations recently. The limitations of traditional drug treatments, including biologics, have spurred interest in herbal medicines for managing UC. This study aimed to elucidate the protective mechanisms of hydroethanolic extract from Lepidium apetalum Willdenow (LWE) on intestinal integrity and inflammation in a dextran sodium sulfate (DSS)-induced colitis model of inflammatory bowel disease (IBD). Using UPLC-MS/MS analysis, eleven phytochemicals were identified in LWE, including catechin, vicenin-2, and quercetin. LWE restored transepithelial electrical resistance (TEER) and reduced paracellular permeability in IL-6-stimulated Caco-2 cells, increasing the expression of the tight junction proteins ZO-1 and occludin. LWE treatment alleviated DSS-induced colitis symptoms in mice, reducing body weight loss, disease activity index values, and spleen size, while improving colon length and reducing serum FITC-dextran levels, indicating enhanced intestinal barrier function. LWE suppressed NLRP3 inflammasome activation, reducing protein levels of pro-caspase-1, cleaved-caspase-1, ASC, and NLRP3, as well as mRNA levels of IL-1β, IL-6, and TNF-α. LWE treatment upregulated activity and mRNA levels of the antioxidant enzymes SOD1 and NQO1. Additionally, LWE modulated the Nrf2/Keap1 pathway, increasing p-Nrf2 levels and decreasing Keap1 levels. LWE also restored goblet cell numbers and reduced fibrosis in DSS-induced chronic colitis mice, increasing gene and protein expressions of ZO-1 and occludin. In summary, LWE shows promise as a therapeutic intervention for reducing tissue damage and inflammation by enhancing intestinal barrier function and inhibiting colonic oxidative stress-induced inflammasome activation.
Collapse
Affiliation(s)
- Kwang-Youn Kim
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 41062, Republic of Korea; (Y.-M.K.); (Y.-J.K.); (K.-H.K.)
| | - Yun-Mi Kang
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 41062, Republic of Korea; (Y.-M.K.); (Y.-J.K.); (K.-H.K.)
| | - Ami Lee
- Herbal Medicine Research Division, Korea Institution of Oriental Medicine (KIOM), Daejeon 34054, Republic of Korea;
- Korean Convergence Medical Science Major, KIOM School, University of Science & Technology (UST), Daejeon 34054, Republic of Korea
| | - Yeon-Ji Kim
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 41062, Republic of Korea; (Y.-M.K.); (Y.-J.K.); (K.-H.K.)
| | - Kyung-Ho Kim
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 41062, Republic of Korea; (Y.-M.K.); (Y.-J.K.); (K.-H.K.)
- Korean Convergence Medical Science Major, KIOM School, University of Science & Technology (UST), Daejeon 34054, Republic of Korea
| | - Youn-Hwan Hwang
- Herbal Medicine Research Division, Korea Institution of Oriental Medicine (KIOM), Daejeon 34054, Republic of Korea;
- Korean Convergence Medical Science Major, KIOM School, University of Science & Technology (UST), Daejeon 34054, Republic of Korea
| |
Collapse
|
22
|
Alqudah A, Qnais E, Gammoh O, Bseiso Y, Wedyan M, Alqudah M, Oqal M, Abudalo R, Abdalla SS. Exploring the therapeutic potential of Anastatica hierochuntica essential oil in DSS-induced colitis. Inflammopharmacology 2024; 32:2035-2048. [PMID: 38520575 DOI: 10.1007/s10787-024-01449-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 02/23/2024] [Indexed: 03/25/2024]
Abstract
The aim of this investigation was to explore the protective impacts and mechanisms of Anastatica hierochuntica essential oil (EOAH) against dextran sulfate sodium (DSS)-induced experimental colitis in mice. EOAH demonstrated a reduction in DSS-induced body weight decline, disease activity index (DAI), colon length reduction, colonic tissue damage, and myeloperoxidase (MPO) activity. The essential oil significantly mitigated the production of pro-inflammatory agents including TNF-α, IL-1β, and IL-12. Further analysis revealed that EOAH's anti-inflammatory effects involved the regulation of NF-κB and PPARγ pathways, as well as the inhibition of NLRP3 activation in colitis mice. Notably, EOAH treatment elevated the levels of beneficial commensal bacteria such as Lactobacillus and Bifidobacteria, while reducing Escherichia coli levels in the mice's feces. In addition, EOAH restored the expression of occludin and ZO-1 proteins in colonic tissues affected by ulcerative colitis (UC). These findings indicate that supplementing with EOAH might offer a novel therapeutic approach for UC prevention.
Collapse
Affiliation(s)
- Abdelrahim Alqudah
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa, Jordan.
| | - Esam Qnais
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Omar Gammoh
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Yousra Bseiso
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Mohammed Wedyan
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Mohammed Alqudah
- Physiology Department, School of Medicine and Biomedical Sciences, Arabian Gulf University, Manama, Bahrain
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Muna Oqal
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa, Jordan
| | - Rawan Abudalo
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa, Jordan
| | - Shtaywy S Abdalla
- Department of Biological Sciences, Faculty of Science, University of Jordan, Amman, Jordan
| |
Collapse
|
23
|
Zhang Z, Huang J, Li C, Zhao Z, Cui Y, Yuan X, Wang X, Liu Y, Zhou Y, Zhu Z. The gut microbiota contributes to the infection of bovine viral diarrhea virus in mice. J Virol 2024; 98:e0203523. [PMID: 38299844 PMCID: PMC10878277 DOI: 10.1128/jvi.02035-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 01/10/2024] [Indexed: 02/02/2024] Open
Abstract
Bovine viral diarrhea virus (BVDV) is prevalent worldwide and causes significant economic losses. Gut microbiota is a large microbial community and has a variety of biological functions. However, whether there is a correlation between gut microbiota and BVDV infection and what kind of relation between them have not been reported. Here, we found that gut microbiota composition changed in normal mice after infecting with BVDV, but mainly the low abundance microbe was affected. Interestingly, BVDV infection significantly reduced the diversity of gut microbiota and changed its composition in gut microbiota-dysbiosis mice. Furthermore, compared with normal mice of BVDV infection, there were more viral loads in the duodenum, jejunum, spleen, and liver of the gut microbiota-dysbiosis mice. However, feces microbiota transplantation (FMT) reversed these effects. The data above indicated that the dysbiosis of gut microbiota was a key factor in the high infection rate of BVDV. It is found that the IFN-I signal was involved by investigating the underlying mechanisms. The inhibition of the proliferation and increase in the apoptosis of peripheral blood lymphocytes (PBL) were also observed. However, FMT treatment reversed these changes by regulating PI3K/Akt, ERK, and Caspase-9/Caspase-3 pathways. Furthermore, the involvement of butyrate in the pathogenesis of BVDV was also further confirmed. Our results showed for the first time that gut microbiota acts as a key endogenous defense mechanism against BVDV infection; moreover, targeting regulation of gut microbiota structure and abundance may serve as a new strategy to prevent and control the disease.IMPORTANCEWhether the high infection rate of BVDV is related to gut microbiota has not been reported. In addition, most studies on BVDV focus on in vitro experiments, which limits the study of its prevention and control strategy and its pathogenic mechanism. In this study, we successfully confirmed the causal relationship between gut microbiota and BVDV infection as well as the potential molecular mechanism based on a mouse model of BVDV infection and a mouse model of gut microbiota dysbiosis. Meanwhile, a mouse model which is more susceptible to BVDV provided in this study lays an important foundation for further research on prevention and control strategy of BVDV and its pathogenesis. In addition, the antiviral effect of butyrate, the metabolites of butyrate-producing bacteria, has been further revealed. Overall, our findings provide a promising prevention and control strategy to treat this infectious disease which is distributed worldwide.
Collapse
Affiliation(s)
- Zecai Zhang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
- Engineering Research Center for Prevention and Control of Cattle Diseases, Heilongjiang Province, Daqing, China
- Heilongjiang Province Cultivating Collaborative Innovation Center for The Beidahuang Modern Agricultural Industry Technology, Daqing, China
| | - Jiang Huang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Agriculture and Rural Bureau of Sinan County, Sinan County, Guizhou, China
- Animal Health Supervision Institute of Sinan County, Sinan County, Guizhou, China
| | - Chuang Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
| | - Zhicheng Zhao
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
| | - Yueqi Cui
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
| | - Xueying Yuan
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
| | - Xue Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
| | - Yu Liu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
- Engineering Research Center for Prevention and Control of Cattle Diseases, Heilongjiang Province, Daqing, China
- Heilongjiang Province Cultivating Collaborative Innovation Center for The Beidahuang Modern Agricultural Industry Technology, Daqing, China
| | - Yulong Zhou
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
- Engineering Research Center for Prevention and Control of Cattle Diseases, Heilongjiang Province, Daqing, China
- Heilongjiang Province Cultivating Collaborative Innovation Center for The Beidahuang Modern Agricultural Industry Technology, Daqing, China
| | - Zhanbo Zhu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
- Engineering Research Center for Prevention and Control of Cattle Diseases, Heilongjiang Province, Daqing, China
- Heilongjiang Province Cultivating Collaborative Innovation Center for The Beidahuang Modern Agricultural Industry Technology, Daqing, China
| |
Collapse
|
24
|
Zhang J, Ren X, Wang S, Liu R, Shi B, Dong H, Wu Q. Microbial interventions in yak colibacillosis: Lactobacillus-mediated regulation of intestinal barrier. Front Cell Infect Microbiol 2024; 14:1337439. [PMID: 38390621 PMCID: PMC10883308 DOI: 10.3389/fcimb.2024.1337439] [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: 11/13/2023] [Accepted: 01/17/2024] [Indexed: 02/24/2024] Open
Abstract
Introduction The etiology of Escherichia coli in yaks, along with its drug resistance, results in economic losses within the yak breeding industry. The utilization of lactic acid bacteria treatment has emerged as a viable alternative to antibiotics in managing colibacillosis. Methods To elucidate the therapeutic mechanisms of Lactobacillus against Escherichia coli-induced intestinal barrier damage in yaks, we employed yak epithelial cells as the experimental model and established a monolayer epithelial barrier using Transwell. The study encompassed four groups: a control group, a model group (exposed to E. coli O78), a low-dose Lactobacillus group (E. coli O78 + 1 × 105CFU LAB), and a high-dose Lactobacillus group (E. coli O78 + 1 × 107CFU LAB). Various techniques, including transmembrane resistance measurement, CFU counting, RT-qPCR, and Western Blot, were employed to assess indicators related to cell barrier permeability and tight junction integrity. Results In the Model group, Escherichia coli O78 significantly compromised the permeability and tight junction integrity of the yak epithelial barrier. It resulted in decreased transmembrane resistance, elevated FD4 flux, and bacterial translocation. Furthermore, it downregulated the mRNA and protein expression of MUC2, Occludin, and ZO-1, while upregulating the mRNA expression and protein expression of FABP2 and Zonulin, thereby impairing intestinal barrier function. Contrastingly, Lactobacillus exhibited a remarkable protective effect. It substantially increased transmembrane resistance, mitigated FD4 flux, and reduced bacterial translocation. Moreover, it significantly upregulated the mRNA and protein expression of MUC2, Occludin, and ZO-1, while downregulating the mRNA and protein expression of FABP2 and Zonulin. Notably, high-dose LAB demonstrated superior regulatory effects compared to the low-dose LAB group. Discussion In conclusion, our findings suggest that Lactobacillus holds promise in treating yak colibacillosis by enhancing mucin and tight junction protein expression. Furthermore, we propose that Lactobacillus achieves these effects through the regulation of Zonulin.
Collapse
Affiliation(s)
- Jingbo Zhang
- College of Animal Science, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| | - Xiaoli Ren
- College of Animal Science, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| | - Shuo Wang
- College of Animal Science, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| | - Ruidong Liu
- College of Animal Science, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| | - Bin Shi
- College of Animal Science, Tibet Agriculture and Animal Husbandry University, Linzhi, China
- Institute of Animal Husbandry and Veterinary Medicine, Tibet Autonomous Region Academy of Agriculture and Animal Science, Lhasa, China
| | - Hailong Dong
- College of Animal Science, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| | - Qingxia Wu
- College of Animal Science, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| |
Collapse
|
25
|
Koufou EE, Assimakopoulos SF, Bosgana P, de Lastic AL, Grypari IM, Georgopoulou GA, Antonopoulou S, Mouzaki A, Kourea HP, Thomopoulos K, Davlouros P. Altered Expression of Intestinal Tight Junction Proteins in Heart Failure Patients with Reduced or Preserved Ejection Fraction: A Pathogenetic Mechanism of Intestinal Hyperpermeability. Biomedicines 2024; 12:160. [PMID: 38255265 PMCID: PMC10813326 DOI: 10.3390/biomedicines12010160] [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: 12/19/2023] [Revised: 01/07/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Although intestinal microbiota alterations (dysbiosis) have been described in heart failure (HF) patients, the possible mechanisms of intestinal barrier dysfunction leading to endotoxemia and systemic inflammation are not fully understood. In this study, we investigated the expression of the intestinal tight junction (TJ) proteins occludin and claudin-1 in patients with HF with reduced (HFrEF) or preserved ejection fraction (HFpEF) and their possible association with systemic endotoxemia and inflammation. Ten healthy controls and twenty-eight patients with HF (HFrEF (n = 14), HFpEF (n = 14)) underwent duodenal biopsy. Histological parameters were recorded, intraepithelial CD3+ T-cells and the expression of occludin and claudin-1 in enterocytes were examined using immunohistochemistry, circulating endotoxin concentrations were determined using ELISA, and concentrations of cytokines were determined using flow cytometry. Patients with HFrEF or HFpEF had significantly higher serum endotoxin concentrations (p < 0.001), a significantly decreased intestinal occludin and claudin-1 expression (in HfrEF p < 0.01 for occludin, p < 0.05 for claudin-1, in HfpEF p < 0.01 occludin and claudin-1), and significantly increased serum concentrations of IL-6, IL-8, and IL-10 (for IL-6 and IL-10, p < 0.05 for HFrEF and p < 0.001 for HFpEF; and for IL-8, p < 0.05 for both groups) compared to controls. Occludin and claudin-1 expression inversely correlated with systemic endotoxemia (p < 0.05 and p < 0.01, respectively). Heart failure, regardless of the type of ejection fraction, results in a significant decrease in enterocytic occludin and claudin-1 expression, which may represent an important cellular mechanism for the intestinal barrier dysfunction causing systemic endotoxemia and inflammatory response.
Collapse
Affiliation(s)
| | - Stelios F. Assimakopoulos
- Department of Internal Medicine and Division of Infectious Diseases, University of Patras Medical School, 26504 Patras, Greece;
| | - Pinelopi Bosgana
- Department of Pathology, Medical School of Patras, 26504 Patras, Greece; (P.B.); (H.P.K.)
| | - Anne-Lise de Lastic
- Laboratory of Immunohematology, Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, 26504 Patras, Greece; (A.-L.d.L.); (A.M.)
| | - Ioanna-Maria Grypari
- Cytology Department, Aretaieion University Hospital, National Kapodistrian University of Athens, 11528 Athens, Greece;
| | | | | | - Athanasia Mouzaki
- Laboratory of Immunohematology, Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, 26504 Patras, Greece; (A.-L.d.L.); (A.M.)
| | - Helen P. Kourea
- Department of Pathology, Medical School of Patras, 26504 Patras, Greece; (P.B.); (H.P.K.)
| | - Konstantinos Thomopoulos
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University of Patras, University Hospital of Patras, 26504 Patras, Greece;
| | - Periklis Davlouros
- Department of Cardiology, Patras University Hospital, 26504 Patras, Greece;
| |
Collapse
|
26
|
Coutry N, Gasmi I, Herbert F, Jay P. Mechanisms of intestinal dysbiosis: new insights into tuft cell functions. Gut Microbes 2024; 16:2379624. [PMID: 39042424 PMCID: PMC11268228 DOI: 10.1080/19490976.2024.2379624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 07/08/2024] [Indexed: 07/24/2024] Open
Abstract
Symbiosis between the host and intestinal microbial communities is essential for human health. Disruption in this symbiosis is linked to gastrointestinal diseases, including inflammatory bowel diseases, as well as extra-gastrointestinal diseases. Unbalanced gut microbiome or gut dysbiosis contributes in multiple ways to disease frequency, severity and progression. Microbiome taxonomic profiling and metabolomics approaches greatly improved our understanding of gut dysbiosis features; however, the precise mechanisms involved in gut dysbiosis establishment still need to be clarified. The aim of this review is to present new actors and mechanisms underlying gut dysbiosis formation following parasitic infection or in a context of altered Paneth cells, revealing the existence of a critical crosstalk between Paneth and tuft cells to control microbiome composition.
Collapse
Affiliation(s)
- Nathalie Coutry
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Imène Gasmi
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Fabien Herbert
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Philippe Jay
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| |
Collapse
|
27
|
Prame Kumar K, Ooi JD, Goldberg R. The interplay between the microbiota, diet and T regulatory cells in the preservation of the gut barrier in inflammatory bowel disease. Front Microbiol 2023; 14:1291724. [PMID: 38107848 PMCID: PMC10722198 DOI: 10.3389/fmicb.2023.1291724] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/06/2023] [Indexed: 12/19/2023] Open
Abstract
Inflammatory bowel disease (IBD) is becoming more common in the Western world due to changes in diet-related microbial dysbiosis, genetics and lifestyle. Incidences of gut permeability can predate IBD and continued gut barrier disruptions increase the exposure of bacterial antigens to the immune system thereby perpetuating chronic inflammation. Currently, most of the approved IBD therapies target individual pro-inflammatory cytokines and pathways. However, they fail in approximately 50% of patients due to their inability to overcome the redundant pro inflammatory immune responses. There is increasing interest in the therapeutic potential of T regulatory cells (Tregs) in inflammatory conditions due to their widespread capability to dampen inflammation, promote tolerance of intestinal bacteria, facilitate healing of the mucosal barrier and ability to be engineered for more targeted therapy. Intestinal Treg populations are inherently shaped by dietary molecules and gut microbiota-derived metabolites. Thus, understanding how these molecules influence Treg-mediated preservation of the intestinal barrier will provide insights into immune tolerance-mediated mucosal homeostasis. This review comprehensively explores the interplay between diet, gut microbiota, and immune system in influencing the intestinal barrier function to attenuate the progression of colitis.
Collapse
Affiliation(s)
- Kathryn Prame Kumar
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Monash Medical Centre, Monash University, Clayton, VIC, Australia
| | | | | |
Collapse
|
28
|
Li L, He Y, Wang N, Li Y, Du Y, He N, Wang B, Zhang T. Atractylone in the Atractylodes macrocephala Rhizoma Essential Oil and Its Anti-Inflammatory Activity. Molecules 2023; 28:7340. [PMID: 37959758 PMCID: PMC10648463 DOI: 10.3390/molecules28217340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/21/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
The aim of this study was to conduct a screening of potential therapeutic compounds found in the Atractylodes macrocephala rhizoma essential oil (AO) and explore its mechanism of action in the treatment of ulcerative colitis (UC). An inflammation cell model was employed in conjunction with phospho-antibody array technology to explore potential therapeutic compounds of AO and their anti-inflammatory and antioxidant effects. Furthermore, we assessed their efficacy and mechanisms of action in treating dextran sulfate sodium (DSS)-induced colitis in mice. Via the screening process, we identified atractylone (ATR) as the primary active compound in AO. It has been demonstrated that ATR can both decrease the levels of tumor necrosis factor (TNF)-α and reactive oxygen species (ROS) and increase the expression of adhesion proteins such as claudin, ZO-1, and occludin in vitro. Moreover, ATR has been shown to improve UC symptoms in vivo. Via a non-targeted metabolomics analysis of colon tissue, we identified 57 distinct metabolites that responded to ATR treatment. Subsequent analysis of the metabolic pathways revealed that the action of ATR was primarily focused on the amino acid metabolism pathway. In summary, ATR may alleviate the symptoms of UC by regulating multiple signaling pathways. Additionally, ATR has a comprehensive function in anti-inflammation, antioxidative stress, and intestinal injury reduction.
Collapse
Affiliation(s)
- Ling Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (L.L.); (Y.H.); (N.W.); (Y.D.)
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China;
| | - Yihao He
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (L.L.); (Y.H.); (N.W.); (Y.D.)
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Nan Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (L.L.); (Y.H.); (N.W.); (Y.D.)
| | - Yuting Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China;
| | - Yaoyao Du
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (L.L.); (Y.H.); (N.W.); (Y.D.)
| | - Ning He
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China;
| | - Bing Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (L.L.); (Y.H.); (N.W.); (Y.D.)
| |
Collapse
|
29
|
Kollmann C, Buerkert H, Meir M, Richter K, Kretzschmar K, Flemming S, Kelm M, Germer CT, Otto C, Burkard N, Schlegel N. Human organoids are superior to cell culture models for intestinal barrier research. Front Cell Dev Biol 2023; 11:1223032. [PMID: 37849736 PMCID: PMC10577213 DOI: 10.3389/fcell.2023.1223032] [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: 05/15/2023] [Accepted: 09/19/2023] [Indexed: 10/19/2023] Open
Abstract
Loss of intestinal epithelial barrier function is a hallmark in digestive tract inflammation. The detailed mechanisms remain unclear due to the lack of suitable cell-based models in barrier research. Here we performed a detailed functional characterization of human intestinal organoid cultures under different conditions with the aim to suggest an optimized ex-vivo model to further analyse inflammation-induced intestinal epithelial barrier dysfunction. Differentiated Caco2 cells as a traditional model for intestinal epithelial barrier research displayed mature barrier functions which were reduced after challenge with cytomix (TNFα, IFN-γ, IL-1ß) to mimic inflammatory conditions. Human intestinal organoids grown in culture medium were highly proliferative, displayed high levels of LGR5 with overall low rates of intercellular adhesion and immature barrier function resembling conditions usually found in intestinal crypts. WNT-depletion resulted in the differentiation of intestinal organoids with reduced LGR5 levels and upregulation of markers representing the presence of all cell types present along the crypt-villus axis. This was paralleled by barrier maturation with junctional proteins regularly distributed at the cell borders. Application of cytomix in immature human intestinal organoid cultures resulted in reduced barrier function that was accompanied with cell fragmentation, cell death and overall loss of junctional proteins, demonstrating a high susceptibility of the organoid culture to inflammatory stimuli. In differentiated organoid cultures, cytomix induced a hierarchical sequence of changes beginning with loss of cell adhesion, redistribution of junctional proteins from the cell border, protein degradation which was accompanied by loss of epithelial barrier function. Cell viability was observed to decrease with time but was preserved when initial barrier changes were evident. In summary, differentiated intestinal organoid cultures represent an optimized human ex-vivo model which allows a comprehensive reflection to the situation observed in patients with intestinal inflammation. Our data suggest a hierarchical sequence of inflammation-induced intestinal barrier dysfunction starting with loss of intercellular adhesion, followed by redistribution and loss of junctional proteins resulting in reduced barrier function with consecutive epithelial death.
Collapse
Affiliation(s)
- Catherine Kollmann
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Hannah Buerkert
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Michael Meir
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Konstantin Richter
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Kai Kretzschmar
- Mildred-Scheel Early Career Centre (MSNZ) for Cancer Research, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Sven Flemming
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Matthias Kelm
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Christoph-Thomas Germer
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Christoph Otto
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Natalie Burkard
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Nicolas Schlegel
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
30
|
Hu Y, He Z, Zhang J, Zhang C, Wang Y, Zhang W, Zhang F, Zhang W, Gu F, Hu W. Effect of Piper nigrum essential oil in dextran sulfate sodium (DSS)-induced colitis and its potential mechanisms. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 119:155024. [PMID: 37597364 DOI: 10.1016/j.phymed.2023.155024] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/07/2023] [Accepted: 08/12/2023] [Indexed: 08/21/2023]
Abstract
BACKGROUND Piper nigrum essential oil (PnEO) possesses pleasant aroma, unique flavor, and various bioactivities; however, its role against colitis remains unclear. PURPOSE In this study, we investigated the role of PnEO in relieving colitis and explored its potential mechanisms in a mouse model of dextran sulfate sodium (DSS)-induced colitis. METHODS Initially, we identified and quantified the components of PnEO by gas chromatography-mass spectrometry (GC-MS). Subsequently, we investigated the protective role of PnEO (50 and 200 mg/kg) in DSS-induced colitis in mice by evaluating disease activity index (DAI) scores and colon length, and performing histological analyses. Eyeball blood was collected and cytokines were determined using ELISA kits. The anti-inflammatory mechanisms of PnEO were analyzed by western blot (WB) and immunohistochemistry (IHC). The intestinal barrier function was evaluated according to tight junction (TJ) protein mRNA levels. We used 16S rRNA gene sequencing to analyze the intestinal microflora of mouse cecal contents. RESULTS Supplementation with PnEO (50 and 200 mg/kg) increased colon length and improved colon histopathology. PnEO regulated inflammatory responses by downregulating TLR4/MAPKs activation, thereby reducing the release of cytokines and mediators. Moreover, it also protected the intestinal barrier through enhancing the expression of claudin-1, claudin-3, occludin, ZO-1, and mucin 2. 16S rRNA gene sequencing revealed that PnEO (200 mg/kg) decreased the abundance of Akkermansia in the gut microbiome. CONCLUSION PnEO treatment (50 and 200 mg/kg) relieved DSS-induced colitis by inhibiting TLR4/MAPK pathway and protecting intestinal barrier, and high-dose PnEO exhibited better effects. Moreover, PnEO (200 mg/kg) regulated key compositions of the gut microbiome, which indicated that it had therapeutic potential for sustaining gut health to lower the risk of colitis.
Collapse
Affiliation(s)
- Yeye Hu
- Spice and Beverage Research Institute, Sanya Research Institute, Chinese Academy of Tropical Agricultural Sciences, Hainan 572025, China; Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China
| | - Ziliang He
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China
| | - Ji Zhang
- School of Life Sciences, Huaiyin Normal University, Huaian 223300, China
| | - Chaohua Zhang
- Spice and Beverage Research Institute, Sanya Research Institute, Chinese Academy of Tropical Agricultural Sciences, Hainan 572025, China
| | - Yanting Wang
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China
| | - Wei Zhang
- School of Life Sciences, Huaiyin Normal University, Huaian 223300, China
| | - Fenglun Zhang
- Nanjing Institute for the Comprehensive Utilization of Wild Plants, Nanjing 211111, China
| | - Weiming Zhang
- Nanjing Institute for the Comprehensive Utilization of Wild Plants, Nanjing 211111, China
| | - Fenglin Gu
- Spice and Beverage Research Institute, Sanya Research Institute, Chinese Academy of Tropical Agricultural Sciences, Hainan 572025, China; Key Laboratory of Processing Suitability and Quality Control of the Special Tropical Crops of Hainan Province, Wanning, Hainan 571533, China.
| | - Weicheng Hu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, School of Medicine, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
31
|
Zhu MZ, Yang MF, Song Y, Xu HM, Xu J, Yue NN, Zhang Y, Tian CM, Shi RY, Liang YJ, Yao J, Wang LS, Nie YQ, Li DF. Exploring the efficacy of herbal medicinal products as oral therapy for inflammatory bowel disease. Biomed Pharmacother 2023; 165:115266. [PMID: 37541177 DOI: 10.1016/j.biopha.2023.115266] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/30/2023] [Accepted: 07/31/2023] [Indexed: 08/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) encompasses a collection of idiopathic diseases characterized by chronic inflammation in the gastrointestinal (GI) tract. Patients diagnosed with IBD often experience necessitate long-term pharmacological interventions. Among the multitude of administration routes available for treating IBD, oral administration has gained significant popularity owing to its convenience and widespread utilization. In recent years, there has been extensive evaluation of the efficacy of orally administered herbal medicinal products and their extracts as a means of treating IBD. Consequently, substantial evidence has emerged, supporting their effectiveness in IBD treatment. This review aimed to provide a comprehensive summary of recent studies evaluating the effects of herbal medicinal products in the treatment of IBD. We delved into the regulatory role of these products in modulating immunity and maintaining the integrity of the intestinal epithelial barrier. Additionally, we examined their impact on antioxidant activity, anti-inflammatory properties, and the modulation of intestinal flora. By exploring these aspects, we aimed to emphasize the significant advantages associated with the use of oral herbal medicinal products in the treatment of IBD. Of particular note, this review introduced the concept of herbal plant-derived exosome-like nanoparticles (PDENs) as the active ingredient in herbal medicinal products for the treatment of IBD. The inclusion of PDENs offers distinct advantages, including enhanced tissue penetration and improved physical and chemical stability. These unique attributes not only demonstrate the potential of PDENs but also pave the way for the modernization of herbal medicinal products in IBD treatment.
Collapse
Affiliation(s)
- Min-Zheng Zhu
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, Guangdong, China
| | - Mei-Feng Yang
- Department of Hematology, Yantian District People's Hospital, Shenzhen 518020, Guangdong, China
| | - Yang Song
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China; Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China
| | - Hao-Ming Xu
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, Guangdong, China
| | - Jing Xu
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, Guangdong, China
| | - Ning-Ning Yue
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University), Shenzhen 518020, Guangdong, China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou 516000, Guangdong, China
| | - Cheng-Mei Tian
- Department of Emergency, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Rui-Yue Shi
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China; Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China
| | - Yu-Jie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen 518020, Guangdong, China.
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China; Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China.
| | - Li-Sheng Wang
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China; Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China.
| | - Yu-Qiang Nie
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, Guangdong, China.
| | - De-Feng Li
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China; Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China.
| |
Collapse
|
32
|
Xie MM, Dai B, Hackney JA, Sun T, Zhang J, Jackman JK, Jeet S, Irizarry-Caro RA, Fu Y, Liang Y, Bender H, Shamir ER, Keir ME, Bevers J, Nakamura G, Townsend MJ, Fox DA, Scherl A, Lee WP, Martin F, Godowski PJ, Pappu R, Yi T. An agonistic anti-signal regulatory protein α antibody for chronic inflammatory diseases. Cell Rep Med 2023; 4:101130. [PMID: 37490914 PMCID: PMC10439247 DOI: 10.1016/j.xcrm.2023.101130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 05/23/2023] [Accepted: 06/30/2023] [Indexed: 07/27/2023]
Abstract
Signal regulatory protein (SIRPα) is an immune inhibitory receptor expressed by myeloid cells to inhibit immune cell phagocytosis, migration, and activation. Despite the progress of SIRPα and CD47 antagonist antibodies to promote anti-cancer immunity, it is not yet known whether SIRPα receptor agonism could restrain excessive autoimmune tissue inflammation. Here, we report that neutrophil- and monocyte-associated genes including SIRPA are increased in inflamed tissue biopsies from patients with rheumatoid arthritis and inflammatory bowel diseases, and elevated SIRPA is associated with treatment-refractory ulcerative colitis. We next identify an agonistic anti-SIRPα antibody that exhibits potent anti-inflammatory effects in reducing neutrophil and monocyte chemotaxis and tissue infiltration. In preclinical models of arthritis and colitis, anti-SIRPα agonistic antibody ameliorates autoimmune joint inflammation and inflammatory colitis by reducing neutrophils and monocytes in tissues. Our work provides a proof of concept for SIRPα receptor agonism for suppressing excessive innate immune activation and chronic inflammatory disease treatment.
Collapse
Affiliation(s)
- Markus M Xie
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA
| | - Bingbing Dai
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA
| | - Jason A Hackney
- Department of OMNI Biomarker Development, Genentech, Inc., South San Francisco, CA, USA
| | - Tianhe Sun
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA
| | - Juan Zhang
- Department of Translational Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Janet K Jackman
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA
| | - Surinder Jeet
- Department of Translational Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Ricardo A Irizarry-Caro
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA; Department of Human Pathobiology and OMNI Reverse Translation, Genentech, Inc., South San Francisco, CA, USA
| | - Yongyao Fu
- Department of Discovery Oncology, Genentech, Inc., South San Francisco, CA, USA
| | - Yuxin Liang
- Department of Microchemistry, Proteomics, and Lipidomics and Next Generation Sequencing, Genentech, Inc., South San Francisco, CA, USA
| | - Hannah Bender
- Department of Pathology, Genentech, Inc., South San Francisco, CA, USA
| | - Eliah R Shamir
- Department of Pathology, Genentech, Inc., South San Francisco, CA, USA
| | - Mary E Keir
- Department of Human Pathobiology and OMNI Reverse Translation, Genentech, Inc., South San Francisco, CA, USA
| | - Jack Bevers
- Department of Antibody Engineering, Genentech, Inc., South San Francisco, CA, USA
| | - Gerald Nakamura
- Department of Antibody Engineering, Genentech, Inc., South San Francisco, CA, USA
| | - Michael J Townsend
- Department of Human Pathobiology and OMNI Reverse Translation, Genentech, Inc., South San Francisco, CA, USA
| | - David A Fox
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Alexis Scherl
- Department of Pathology, Genentech, Inc., South San Francisco, CA, USA
| | - Wyne P Lee
- Department of Translational Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Flavius Martin
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA
| | - Paul J Godowski
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA.
| | - Rajita Pappu
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA.
| | - Tangsheng Yi
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA.
| |
Collapse
|
33
|
Rath T, Atreya R, Neurath MF. A spotlight on intestinal permeability and inflammatory bowel diseases. Expert Rev Gastroenterol Hepatol 2023; 17:893-902. [PMID: 37606514 DOI: 10.1080/17474124.2023.2242772] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 07/27/2023] [Indexed: 08/23/2023]
Abstract
INTRODUCTION The intestinal barrier is a multi-faced structure lining the surface of the intestinal mucosa of the GI tract. To exert its main functions as a physical and immunological defense barrier, several components of the intestinal barrier act in a concerted and cooperative manner. AREAS COVERED Herein, we first introduce to the basic organization of the intestinal barrier and then summarize different methods to assess barrier function in and ex vivo. Finally, we provide an in-depth overview of the relevance of intestinal barrier dysfunction in inflammatory bowel diseases. EXPERT OPINION In parallel to a more fundamental understanding of the intestinal barrier as a key component for intestinal integrity is the notion that intestinal barrier defects are associated with a variety of diseases such as inflammatory bowel diseases. Recent research has fueled and perpetuated the concept that barrier defects are critical components of disease development, disease behavior, and potentially also an area of therapeutic intervention in IBD patients. Although being far away from standard, new technologies can be used to easily assess barrier healing in IBD and to derive clinical consequences from these findings such as more accurate forecasting of future disease behavior or the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Timo Rath
- Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | - Raja Atreya
- Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | - Markus F Neurath
- Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany
- Deutsches Zentrum Für Immuntherapie DZI, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany
| |
Collapse
|
34
|
Cai X, Gao C, Ma L, Li C. Genome-wide identification, evolution and expression analysis of tight junction gene family and the immune roles of claudin5 gene in turbot (Scophthalmus maximus L.). Gene 2023:147541. [PMID: 37301449 DOI: 10.1016/j.gene.2023.147541] [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: 04/11/2023] [Revised: 05/11/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023]
Abstract
Tight junction proteins (TJs) are important component proteins that maintaining the structure and function of TJs, connecting to each other to form a TJ complex between cells, maintaining the biological homeostasis of the internal environment. In this study, a total of 103 TJ genes were identified in turbot according to our whole-transcriptome database. Transmembrane TJs were divided into seven subfamilies, including claudin (CLDN), occludin (OCLD), tricellulin (MARVELD2), MARVEL domain containing 3 (MARVELD3), junctional adhesion molecules (JAM), immunoglobulin superfamily member 5 (IGSF5/JAM4), blood vessel epicardial substance (BVEs). Moreover, the majority of homologous pairs of TJ genes showed highly conserved alongside length, exon/intron number and motifs. As for phylogenetic analysis for 103 TJ genes, eight of them have undergone a positive selection and JAMB-like has undergone the most neutral evolution. The expression patterns of several TJ genes showed the lowest expression levels in blood, while the highest expression levels were detected in intestine, gill and skin, which all belong to mucosal tissues. Meanwhile, most examined TJ genes showed down-regulated expression patterns during bacterial infection, while several TJ genes exhibited up-regulated expression patterns at a later stage (24 h). At the same time, several potential candidate genes (such as CLDN-15, CLDN-3, CLDN-12, CLDN-5 and OCLD) were significantly down-regulated, which may indicate their important functions that involved in the regulation of bacterial infection. Currently, there is little research on CLDN5 in the intestine, but it is highly expressed in the intestine and has significant changes in intestinal expression after bacterial infection. Thus, we knocked down CLDN5 by the method of lentiviral infection. The result showed CLDN5 was related to cell migration (wound healing) and apoptosis, and the method of dualluciferasereporterassay showed that the functions of CLDN5 could be regulated by miR-24. The study of TJs may lead to a better understanding of the function of TJs in teleost.
Collapse
Affiliation(s)
- Xin Cai
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China; Centre for Sustainable Aquatic Ecosystems, Harry Butler Institute, Murdoch University, Murdoch WA 6150, Australia
| | - Chengbin Gao
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China; Centre for Sustainable Aquatic Ecosystems, Harry Butler Institute, Murdoch University, Murdoch WA 6150, Australia
| | - Le Ma
- Centre for Sustainable Aquatic Ecosystems, Harry Butler Institute, Murdoch University, Murdoch WA 6150, Australia
| | - Chao Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
35
|
Xu Y, Xie Y, Wu Z, Wang H, Chen Z, Wang J, Bao W. Protective effects of melatonin on deoxynivalenol-induced oxidative stress and autophagy in IPEC-J2 cells. Food Chem Toxicol 2023; 177:113803. [PMID: 37120089 DOI: 10.1016/j.fct.2023.113803] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/01/2023]
Abstract
This study explored protective effects of melatonin (MEL) on deoxynivalenol (DON)-induced toxicity in porcine jejunum epithelial cells (IPEC-J2). Cells were preexposed to MEL and then exposed to DON to detect cell viability, apoptosis, and oxidative stress indicators. Compared to DON treatment, pretreatment with MEL significantly increased cell proliferation. (P < 0.01), intracellular catalase (CAT) and superoxide dismutase (SOD) levels (P < 0.05), decreased apoptosis and oxidative stress, and significantly attenuated the inflammatory response. RNA-seq analysis revealed that MEL protects IPEC-J2 from the adverse effects of DON by affecting the expression of tight junction and autophagy pathway-related genes. Similarly, further experiments revealed that MEL partly prevented DON-induced disruption of intestinal barrier function and decreased autophagy induced by DON via activation of the AKT/mTOR pathway. In conclusion, these results demonstrated the preventive properties of MEL against DON-induced cell damage by activating the antioxidant system and Inhibition of autophagy.
Collapse
Affiliation(s)
- Yafei Xu
- Key Laboratory for Animal Genetic, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China; Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Yunxiao Xie
- Key Laboratory for Animal Genetic, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Zhengchang Wu
- Key Laboratory for Animal Genetic, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Haifei Wang
- Key Laboratory for Animal Genetic, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Zhenhai Chen
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Jingneng Wang
- Shanghai Xiongtu Biotechnology Co., Ltd., Shanghai, 200000, China
| | - Wenbin Bao
- Key Laboratory for Animal Genetic, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
36
|
Kil BJ, Pyung YJ, Park H, Kang JW, Yun CH, Huh CS. Probiotic potential of Saccharomyces cerevisiae GILA with alleviating intestinal inflammation in a dextran sulfate sodium induced colitis mouse model. Sci Rep 2023; 13:6687. [PMID: 37095161 PMCID: PMC10125971 DOI: 10.1038/s41598-023-33958-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 04/21/2023] [Indexed: 04/26/2023] Open
Abstract
Recently, several probiotic products have been developed; however, most probiotic applications focused on prokaryotic bacteria whereas eukaryotic probiotics have received little attention. Saccharomyces cerevisiae yeast strains are eukaryotes notable for their fermentation and functional food applications. The present study investigated the novel yeast strains isolated from Korean fermented beverages and examined their potential probiotic characteristics. We investigated seven strains among 100 isolates with probiotic characteristics further. The strains have capabilities such as auto-aggregation tendency, co-aggregation with a pathogen, hydrophobicity with n-hexadecane,1,1-diphenyl-2-picrylhydrazyl scavenging effect, survival in simulated gastrointestinal tract conditions and the adhesion ability of the strains to the Caco-2 cells. Furthermore, all the strains contained high cell wall glucan content, a polysaccharide with immunological effects. Internal transcribed spacer sequencing identified the Saccharomyces strains selected in the present study as probiotics. To examine the effects of alleviating inflammation in cells, nitric oxide generation in raw 264.7 cells with S. cerevisiae showed that S. cerevisiae GILA could be a potential probiotic strain able to alleviate inflammation. Three probiotics of S. cerevisiae GILA strains were chosen by in vivo screening with a dextran sulfate sodium-induced colitis murine model. In particular, GILA 118 down-regulates neutrophil-lymphocyte ratio and myeloperoxidase in mice treated with DSS. The expression levels of genes encoding tight junction proteins in the colon were upregulated, cytokine interleukin-10 was significantly increased, and tumor necrosis factor-α was reduced in the serum.
Collapse
Affiliation(s)
- Bum Ju Kil
- Biomodulation Major, and Center for Food and Bioconvergence, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Young Jin Pyung
- Biomodulation Major, and Center for Food and Bioconvergence, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyunjoon Park
- Research Institute of Eco-Friendly Livestock Science, Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, 25354, Republic of Korea
| | - Jun-Won Kang
- Department of Food Science and Biotechnology, Dongguk University-Seoul, 32, Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do, 10326, Republic of Korea
| | - Cheol-Heui Yun
- Biomodulation Major, and Center for Food and Bioconvergence, Seoul National University, Seoul, 08826, Republic of Korea.
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
- Research Institute of Eco-Friendly Livestock Science, Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, 25354, Republic of Korea.
| | - Chul Sung Huh
- Research Institute of Eco-Friendly Livestock Science, Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, 25354, Republic of Korea.
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang-gun, 25354, Republic of Korea.
| |
Collapse
|
37
|
Zhang S, Chen A, Jiang L, Liu X, Chai L. Copper-mediated shifts in transcriptomic responses of intestines in Bufo gargarizans tadpoles to lead stress. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:50144-50161. [PMID: 36790706 DOI: 10.1007/s11356-023-25801-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 02/04/2023] [Indexed: 04/16/2023]
Abstract
The differential transcriptomic responses of intestines in Bufo gargarizans tadpoles to Pb alone or in the presence of Cu were evaluated. Tadpoles were exposed to 30 μg/L Pb individually and in combination with Cu at 16 or 64 μg/L from Gosner stage (Gs) 26 to Gs 38. After de novo assembly, 105,107 unigenes were generated. Compared to the control group, 7387, 6937, and 11139 differentially expressed genes (DEGs) were identified in the treatment of Pb + Cu0, Pb + Cu16, and Pb + Cu64, respectively. In addition, functional annotation and enrichment analysis of DEGs revealed substantial transcriptional reprogramming of diverse molecular and biological pathways were induced in all heavy metal treatments. The relative expression levels of genes associated with intestinal epithelial barrier and bile acids (BAs) metabolism, such as mucin2, claudin5, ZO-1, Asbt, and Ost-β, were validated by qPCR. This study demonstrated that Pb exposure induced transcriptional responses in tadpoles, and the responses could be modulated by Cu.
Collapse
Affiliation(s)
- Siliang Zhang
- School of Water and Environment, Chang'an University, Xi'an, 710054, People's Republic of China
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an, 710054, People's Republic of China
| | - Aixia Chen
- School of Water and Environment, Chang'an University, Xi'an, 710054, People's Republic of China
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an, 710054, People's Republic of China
| | - Ling Jiang
- School of Water and Environment, Chang'an University, Xi'an, 710054, People's Republic of China
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an, 710054, People's Republic of China
| | - Xiaoli Liu
- School of Water and Environment, Chang'an University, Xi'an, 710054, People's Republic of China
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an, 710054, People's Republic of China
| | - Lihong Chai
- School of Water and Environment, Chang'an University, Xi'an, 710054, People's Republic of China.
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an, 710054, People's Republic of China.
| |
Collapse
|
38
|
Tang S, Zhong W, Li T, Li Y, Song G. Isochlorogenic acid A alleviates dextran sulfate sodium-induced ulcerative colitis in mice through STAT3/NF-кB pathway. Int Immunopharmacol 2023; 118:109989. [PMID: 36958213 DOI: 10.1016/j.intimp.2023.109989] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/26/2023] [Accepted: 03/02/2023] [Indexed: 03/25/2023]
Abstract
Isochlorogenic acid A (ICGA-A) is a dicaffeoylquinic acid widely found in various medicinal plants or vegetables, such as Lonicerae japonicae Flos and chicory, and multiple properties of ICGA-A have been reported. However, the therapeutic effect of ICGA-A on colitis is not clear, and thus were investigated in our present study, as well as the underlying mechanisms. Here we found that ICGA-A alleviated clinical symptoms of dextran sodium sulfate (DSS) induced colitis model mice, including disease activity index (DAI) and histological damage. In addition, DSS-induced inflammation was significantly attenuated in mice given ICGA-A supplementation. ICGA-A reduced the fraction of neutrophils in peripheral blood and the infiltration of neutrophils and macrophages in colon tissue, and reduced pro-inflammatory cytokine production and tight junctions in mouse models. Furthermore, ICGA-A down-regulated expression of STAT3 and up-regulated the protein level of IκBα. Our in vitro studies confirmed that ICGA-A inhibited the mRNA expression of pro-inflammatory cytokines. ICGA-A blocked the phosphorylation of STAT3, p65, and IκBα, suppressed the expression STAT3 and p65. In addition, the present study also demonstrated that ICGA-A had no obvious toxicity on normal cells and organs. Taken together, we conclude that ICGA-A mitigates experimental ulcerative colitis (UC) at least in part by inhibiting the STAT3/NF-кB signaling pathways. Hence, ICGA-A may be a promising and effective drug for treating UC.
Collapse
Affiliation(s)
- Shaoshuai Tang
- Fisheries College of Jimei University, Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-environment, Xiamen, China
| | - Wei Zhong
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Tingting Li
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Yuanyue Li
- Fisheries College of Jimei University, Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-environment, Xiamen, China.
| | - Gang Song
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
39
|
Wang Y, Xu Y, Cao G, Zhou X, Wang Q, Fu A, Zhan X. Bacillus subtilis DSM29784 attenuates Clostridium perfringens-induced intestinal damage of broilers by modulating intestinal microbiota and the metabolome. Front Microbiol 2023; 14:1138903. [PMID: 37007491 PMCID: PMC10060821 DOI: 10.3389/fmicb.2023.1138903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/22/2023] [Indexed: 03/18/2023] Open
Abstract
Necrotic enteritis (NE), especially subclinical NE (SNE), without clinical symptoms, in chicks has become one of the most threatening problems to the poultry industry. Therefore, increasing attention has been focused on the research and application of effective probiotic strains as an alternative to antibiotics to prevent SNE in broilers. In the present study, we evaluated the effects of Bacillus subtilis DSM29784 (BS) on the prevention of subclinical necrotic enteritis (SNE) in broilers. A total of 480 1-day-old broiler chickens were randomly assigned to four dietary treatments, each with six replicates pens of twenty birds for 63 d. The negative (Ctr group) and positive (SNE group) groups were only fed a basal diet, while the two treatment groups received basal diets supplemented with BS (1 × 109 colony-forming units BS/kg) (BS group) and 10mg/kg enramycin (ER group), respectively. On days 15, birds except those in the Ctr group were challenged with 20-fold dose coccidiosis vaccine, and then with 1 ml of C. perfringens (2 × 108) at days 18 to 21 for SNE induction. BS, similar to ER, effectively attenuated CP-induced poor growth performance. Moreover, BS pretreatment increased villi height, claudin-1 expression, maltase activity, and immunoglobulin abundance, while decreasing lesional scores, as well as mucosal IFN-γ and TNF-α concentrations. In addition, BS pretreatment increased the relative abundance of beneficial bacteria and decreased that of pathogenic species; many lipid metabolites were enriched in the cecum of treated chickens. These results suggest that BS potentially provides active ingredients that may serve as an antibiotic substitute, effectively preventing SNE-induced growth decline by enhancing intestinal health in broilers.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, College of Animal Sciences, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Yibin Xu
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, College of Animal Sciences, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | | | - Xihong Zhou
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Qian Wang
- Yancheng Biological Engineering Higher Vocational Technology School, Yancheng, China
| | - Aikun Fu
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, College of Animal Sciences, Institute of Feed Science, Zhejiang University, Hangzhou, China
- *Correspondence: Xiuan Zhan, ; Aikun Fu,
| | - Xiuan Zhan
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, College of Animal Sciences, Institute of Feed Science, Zhejiang University, Hangzhou, China
- *Correspondence: Xiuan Zhan, ; Aikun Fu,
| |
Collapse
|
40
|
Azcutia V, Kelm M, Fink D, Cummings RD, Nusrat A, Parkos CA, Brazil JC. Sialylation regulates neutrophil transepithelial migration, CD11b/CD18 activation, and intestinal mucosal inflammatory function. JCI Insight 2023; 8:e167151. [PMID: 36719745 PMCID: PMC10077474 DOI: 10.1172/jci.insight.167151] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/25/2023] [Indexed: 02/01/2023] Open
Abstract
Polymorphonuclear neutrophils (PMNs) play a critical role in clearing invading microbes and promoting tissue repair following infection/injury. However, dysregulated PMN trafficking and associated tissue damage is pathognomonic of numerous inflammatory mucosal diseases. The final step in PMN influx into mucosal lined organs (including the lungs, kidneys, skin, and gut) involves transepithelial migration (TEpM). The β2-integrin CD11b/CD18 plays an important role in mediating PMN intestinal trafficking, with recent studies highlighting that terminal fucose and GlcNAc glycans on CD11b/CD18 can be targeted to reduce TEpM. However, the role of the most abundant terminal glycan, sialic acid (Sia), in regulating PMN epithelial influx and mucosal inflammatory function is not well understood. Here we demonstrate that inhibiting sialidase-mediated removal of α2-3-linked Sia from CD11b/CD18 inhibits PMN migration across intestinal epithelium in vitro and in vivo. Sialylation was also found to regulate critical PMN inflammatory effector functions, including degranulation and superoxide release. Finally, we demonstrate that sialidase inhibition reduces bacterial peptide-mediated CD11b/CD18 activation in PMN and blocks downstream intracellular signaling mediated by spleen tyrosine kinase (Syk) and p38 MAPK. These findings suggest that sialylated glycans on CD11b/CD18 represent potentially novel targets for ameliorating PMN-mediated tissue destruction in inflammatory mucosal diseases.
Collapse
Affiliation(s)
- Veronica Azcutia
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthias Kelm
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Dylan Fink
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Richard D. Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Asma Nusrat
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Charles A. Parkos
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jennifer C. Brazil
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
41
|
Martínez-Sánchez LDC, Ngo PA, Pradhan R, Becker LS, Boehringer D, Soteriou D, Kubankova M, Schweitzer C, Koch T, Thonn V, Erkert L, Stolzer I, Günther C, Becker C, Weigmann B, Klewer M, Daniel C, Amann K, Tenzer S, Atreya R, Bergo M, Brakebusch C, Watson AJM, Guck J, Fabry B, Atreya I, Neurath MF, López-Posadas R. Epithelial RAC1-dependent cytoskeleton dynamics controls cell mechanics, cell shedding and barrier integrity in intestinal inflammation. Gut 2023; 72:275-294. [PMID: 35241625 PMCID: PMC9872254 DOI: 10.1136/gutjnl-2021-325520] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 01/29/2022] [Indexed: 01/27/2023]
Abstract
OBJECTIVE Increased apoptotic shedding has been linked to intestinal barrier dysfunction and development of inflammatory bowel diseases (IBD). In contrast, physiological cell shedding allows the renewal of the epithelial monolayer without compromising the barrier function. Here, we investigated the role of live cell extrusion in epithelial barrier alterations in IBD. DESIGN Taking advantage of conditional GGTase and RAC1 knockout mice in intestinal epithelial cells (Pggt1b iΔIEC and Rac1 iΔIEC mice), intravital microscopy, immunostaining, mechanobiology, organoid techniques and RNA sequencing, we analysed cell shedding alterations within the intestinal epithelium. Moreover, we examined human gut tissue and intestinal organoids from patients with IBD for cell shedding alterations and RAC1 function. RESULTS Epithelial Pggt1b deletion led to cytoskeleton rearrangement and tight junction redistribution, causing cell overcrowding due to arresting of cell shedding that finally resulted in epithelial leakage and spontaneous mucosal inflammation in the small and to a lesser extent in the large intestine. Both in vivo and in vitro studies (knockout mice, organoids) identified RAC1 as a GGTase target critically involved in prenylation-dependent cytoskeleton dynamics, cell mechanics and epithelial cell shedding. Moreover, inflamed areas of gut tissue from patients with IBD exhibited funnel-like structures, signs of arrested cell shedding and impaired RAC1 function. RAC1 inhibition in human intestinal organoids caused actin alterations compatible with arresting of cell shedding. CONCLUSION Impaired epithelial RAC1 function causes cell overcrowding and epithelial leakage thus inducing chronic intestinal inflammation. Epithelial RAC1 emerges as key regulator of cytoskeletal dynamics, cell mechanics and intestinal cell shedding. Modulation of RAC1 might be exploited for restoration of epithelial integrity in the gut of patients with IBD.
Collapse
Affiliation(s)
- Luz del Carmen Martínez-Sánchez
- Department of Medicine 1, Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Bayern, Germany,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Phuong Anh Ngo
- Department of Medicine 1, Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Bayern, Germany,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Rashmita Pradhan
- Department of Medicine 1, Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Bayern, Germany,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Lukas-Sebastian Becker
- Department of Medicine 1, Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Bayern, Germany,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - David Boehringer
- Department of Physics, University of Erlangen Nuremberg, Erlangen, Bayern, Germany
| | - Despina Soteriou
- Max-Planck Zentrum für Physik und Medizin, Erlangen, Germany,Max Planck Institute for the Science of Light, Erlangen, Bayern, Germany
| | - Marketa Kubankova
- Max-Planck Zentrum für Physik und Medizin, Erlangen, Germany,Max Planck Institute for the Science of Light, Erlangen, Bayern, Germany
| | - Christine Schweitzer
- Max-Planck Zentrum für Physik und Medizin, Erlangen, Germany,Max Planck Institute for the Science of Light, Erlangen, Bayern, Germany
| | - Tatyana Koch
- Department of Medicine 1, Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Bayern, Germany
| | - Veronika Thonn
- Department of Medicine 1, Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Bayern, Germany,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Lena Erkert
- Department of Medicine 1, Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Bayern, Germany,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Iris Stolzer
- Department of Medicine 1, Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Bayern, Germany,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Claudia Günther
- Department of Medicine 1, Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Bayern, Germany,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Christoph Becker
- Department of Medicine 1, Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Bayern, Germany,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Benno Weigmann
- Department of Medicine 1, Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Bayern, Germany,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Monika Klewer
- Department of Nephropathology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
| | - Christoph Daniel
- Department of Nephropathology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
| | - Kerstin Amann
- Department of Nephropathology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
| | - Stefan Tenzer
- University Medical Center Mainz, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Rheinland-Pfalz, Germany
| | - Raja Atreya
- Department of Medicine 1, Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Bayern, Germany,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Martin Bergo
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Cord Brakebusch
- Biotech Research & Innovation Centre, University of Copenhagen, Kobenhavn, Hovedstaden, Denmark
| | | | - Jochen Guck
- Department of Physics, University of Erlangen Nuremberg, Erlangen, Bayern, Germany,Max-Planck Zentrum für Physik und Medizin, Erlangen, Germany,Max Planck Institute for the Science of Light, Erlangen, Bayern, Germany
| | - Ben Fabry
- Department of Physics, University of Erlangen Nuremberg, Erlangen, Bayern, Germany
| | - Imke Atreya
- Department of Medicine 1, Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Bayern, Germany,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Bayern, Germany,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Rocío López-Posadas
- Department of Medicine 1, Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Bayern, Germany .,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| |
Collapse
|
42
|
Chen X, Zhao H, Lu Y, Meng F, Lu Z, Lu Y. Surfactin Mitigates Dextran Sodium Sulfate-Induced Colitis and Behavioral Disorders in Mice by Mediating Gut-Brain-Axis Balance. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:1577-1592. [PMID: 36634244 DOI: 10.1021/acs.jafc.2c07369] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Ulcerative colitis (UC) is associated with brain neurotransmitter disorders and intestinal dysbiosis. Bacillus amyloliquefaciens fmb50 produces the lipopeptide surfactin, which has a wide range of biological activities. However, the effects of surfactin on DSS-induced colitis have not been reported. In the present study, oral surfactin significantly ameliorated colitis in a mouse model and reduced depression-like behavior, such as slowed walking speed, shortened movement distance in the open field test, and weakened exploration ability in the light-dark shuttle test. Surfactin noticeably improved gut microbial dysbiosis, intestinal barrier dysfunction in the colon, and blood-brain barrier dysfunction in the brain. Furthermore, the colon levels of occludin were upregulated by 68.51%, and the brain levels of occludin and ZO-1 were upregulated by 77.81% and 36.42%, respectively. Surfactin supplementation also inhibited inflammatory responses by inactivating the tumor necrosis factor-α (TNF-α), nuclear factor kappa-B (NF-κB), and NLRP3 signaling pathways in the colon and brain. Thus, we believe that surfactin improved the behavioral disorders by upregulating the levels of 5-hydroxytryptamine (5-HT), 5-hydroxyindoleacetic acid (5-HIAA), norepinephrine (NE), and brain-derived neurotrophic factor (BDNF), suppressing the inflammatory responses, and improving the blood-brain barrier dysfunction. Surfactin also reduced the abundances of gut microbes that are related to colitis, especially targeting facultative anaerobes of the phylum Proteobacteria, and it increased the abundance of beneficial bacteria such as Lactobacillus and unidentified Prevotella. Combined with its nontoxic nature observed in this long-term study in mice, oral surfactin might be a promising intervention strategy for preventing colitis by acting on the microbiota-gut-brain axis.
Collapse
Affiliation(s)
- Xiaoyu Chen
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu Province210095, China
| | - Hongyuan Zhao
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu Province210095, China
| | - Yajun Lu
- College of Chemistry and Chemical, Nanjing Tech University, Nanjing211816, China
| | - Fanqiang Meng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu Province210095, China
| | - Zhaoxin Lu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu Province210095, China
| | - Yingjian Lu
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, Jiangsu Province210023, China
| |
Collapse
|
43
|
Shum TF, Wang L, Chiou J. Impact of Plasticizer on the Intestinal Epithelial Integrity and Tissue-Repairing Ability within Cells in the Proximity of the Human Gut Microbiome. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:2152. [PMID: 36767519 PMCID: PMC9915929 DOI: 10.3390/ijerph20032152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/22/2023] [Accepted: 01/22/2023] [Indexed: 06/18/2023]
Abstract
Toxicological research into the impact of plasticizer on different organs has been reported in the past few decades, while their effects on shifting the gut microbiota and immune cells homeostasis in zebrafish were only studied recently. However, studies on the impact of plasticizer on human gut microbiota are scarce. In this study, we co-incubated healthy human fecal microbiota with different concentrations of Di(2-ethylhexyl) phthalate (DEHP) and di-iso-nonyl phthalate (DINP), analyzed microbial composition by 16S rDNA sequencing, and compared the influence of their derived microbiomes on the human enterocyte (HT-29) and murine macrophage (RAW264.7) cell lines. Microbial diversity is reduced by DEHP treatment in a dose-dependent manner. DEHP treatment reduced the phyla Firmicutes/Bacteroidetes ratio, while DINP treatment promoted Proteobacteria. Expressions of tight/adherens junction genes in HT-29 and anti-inflammatory genes in RAW264.7 were down-regulated by plasticizer-co-incubated microbiota derived metabolites. Overall, it is observed that selected plasticizers at high dosages can induce compositional changes in human microbiota. Metabolites from such altered microbiota could affect the tight junction integrity of the intestinal epithelium and upset macrophage differentiation homeostasis in proximity. Chronic exposure to these plasticizers may promote risks of dysbiosis, leaky gut or the exacerbation of intestinal inflammation.
Collapse
Affiliation(s)
- Tim-Fat Shum
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Liwen Wang
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Jiachi Chiou
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
- Research Institute for Future Food, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| |
Collapse
|
44
|
Kim JE, Song HJ, Choi YJ, Jin YJ, Roh YJ, Seol A, Park SH, Park JM, Kang HG, Hwang DY. Improvement of the intestinal epithelial barrier during laxative effects of phlorotannin in loperamide-induced constipation of SD rats. Lab Anim Res 2023; 39:1. [PMID: 36597137 PMCID: PMC9808941 DOI: 10.1186/s42826-022-00152-1] [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: 07/01/2022] [Revised: 12/12/2022] [Accepted: 12/16/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Disruptions of the intestinal epithelial barrier (IEB) are frequently observed in various digestive diseases, including irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD). This study assessed the improvement in the IEB during the laxative activity of phlorotannin (Pt) harvested from Ecklonia cava in constipation by examining the changes in the expression of the regulatory proteins for the tight junction (TJ) and adherens junction (AJ), and inflammatory cytokines in Sprague Dawley (SD) rats with loperamide (Lm)-induced constipation after a Pt treatment. RESULTS The Pt treatment induced laxative activity, including the improvement of feces-related parameters, gastrointestinal transit rate, and histological structure of the mid colon in Lm-treated SD rats. In addition, significant recovery effects were detected in the histology of IEB, including the mucus layer, epithelial cells, and lamina propria in the mid colon of Lm + Pt treated SD rats. The expression levels of E-cadherin and p120-catenin for AJ and the ZO-1, occludin, and Claudin-1 genes for TJ in epithelial cells were improved remarkably after the Pt treatment, but the rate of increase was different. Furthermore, the Pt treatment increased the expression level of several inflammatory cytokines, such as TNF-α, IL-6, IL-1β, IL-13, and IL-4 in Lm + Pt treated SD rats. CONCLUSIONS These results provide the first evidence that the laxative activity of Pt in SD rats with Lm-induced constipation phenotypes involve improvements in the IEB.
Collapse
Affiliation(s)
- Ji Eun Kim
- grid.262229.f0000 0001 0719 8572Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute/Laboratory Animal Resources Center, Pusan National University, Miryang, 50463 Korea
| | - Hee Jin Song
- grid.262229.f0000 0001 0719 8572Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute/Laboratory Animal Resources Center, Pusan National University, Miryang, 50463 Korea
| | - Yun Ju Choi
- grid.262229.f0000 0001 0719 8572Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute/Laboratory Animal Resources Center, Pusan National University, Miryang, 50463 Korea
| | - You Jeong Jin
- grid.262229.f0000 0001 0719 8572Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute/Laboratory Animal Resources Center, Pusan National University, Miryang, 50463 Korea
| | - Yu Jeong Roh
- grid.262229.f0000 0001 0719 8572Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute/Laboratory Animal Resources Center, Pusan National University, Miryang, 50463 Korea
| | - Ayun Seol
- grid.262229.f0000 0001 0719 8572Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute/Laboratory Animal Resources Center, Pusan National University, Miryang, 50463 Korea
| | - So Hae Park
- grid.262229.f0000 0001 0719 8572Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute/Laboratory Animal Resources Center, Pusan National University, Miryang, 50463 Korea
| | - Ju Min Park
- grid.262229.f0000 0001 0719 8572Department of Food Science and Nutrition, College of Human Ecology, Pusan National University, Busan, 46241 Korea
| | - Hyun Gu Kang
- grid.254229.a0000 0000 9611 0917Veterinary Medical Center, Department of Veterinary Theriogenology, College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644 Korea
| | - Dae Youn Hwang
- grid.262229.f0000 0001 0719 8572Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute/Laboratory Animal Resources Center, Pusan National University, Miryang, 50463 Korea
| |
Collapse
|
45
|
Glutathione Protects against Paraquat-Induced Oxidative Stress by Regulating Intestinal Barrier, Antioxidant Capacity, and CAR Signaling Pathway in Weaned Piglets. Nutrients 2022; 15:nu15010198. [PMID: 36615853 PMCID: PMC9823711 DOI: 10.3390/nu15010198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/29/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023] Open
Abstract
Endogenous glutathione (GSH) effectively regulates redox homeostasis in the body. This study aimed to investigate the regulatory mechanism of different dietary levels of GSH supplementation on the intestinal barrier and antioxidant function in a paraquat-induced stress-weaned piglet model. Our results showed that dietary 0.06% GSH supplementation improved the growth performance of weaned piglets under normal and stressful conditions to some degree and decreased the diarrhea rate throughout. Exogenous GSH improved paraquat-induced changes in intestinal morphology, organelle, and permeability and reduced intestinal epithelial cell apoptosis. Moreover, GSH treatment alleviated intestinal oxidative stress damage by upregulating antioxidant (GPX4, CnZnSOD, GCLC, and GCLM) and anti-inflammatory (IL-10) gene expression and downregulating inflammatory cytokines (IFN-γ and IL-12) gene expression. Furthermore, GSH significantly reduced the expression levels of constitutive androstane receptor (CAR), RXRα, HSP90, PP2Ac, CYP2B22, and CYP3A29, and increased the expression levels of GSTA1 and GSTA2 in the jejunum and ileum of paraquat-induced piglets. We conclude that exogenous GSH protects against oxidative stress damage by regulating the intestinal barrier, antioxidant capacity, and CAR signaling pathway.
Collapse
|
46
|
Gut Microbiota in Non-Alcoholic Fatty Liver Disease Patients with Inflammatory Bowel Diseases: A Complex Interplay. Nutrients 2022; 14:nu14245323. [PMID: 36558483 PMCID: PMC9785319 DOI: 10.3390/nu14245323] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
The intestinal microbiota represents the microbial community that colonizes the gastrointestinal tract and constitutes the most complex ecosystem present in nature. The main intestinal microbial phyla are Firmicutes, Bacteroidetes, Actinobacteria, Proteobacteria, Fusobacteria, and Verrucromicrobia, with a clear predominance of the two phyla Firmicutes and Bacteroidetes which account for about 90% of the intestinal phyla. Intestinal microbiota alteration, or dysbiosis, has been proven to be involved in the development of various syndromes, such as non-alcoholic fatty liver disease, Crohn's disease, and ulcerative colitis. The present review underlines the most recurrent changes in the intestinal microbiota of patients with NAFLD, Crohn's disease, and ulcerative colitis.
Collapse
|
47
|
Lv X, Chen L, Zhou C, Guo Y, Zhang G, Kang J, Tan Z, Tang S, Liu Z. Dietary tea tree ( Melaleuca alternifolia) oil supplementation enhances the expressions of amino acid transporters in goat ileal mucosa and improves intestinal immunity. Food Sci Nutr 2022; 10:3749-3758. [PMID: 36348789 PMCID: PMC9632209 DOI: 10.1002/fsn3.2972] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/09/2022] [Accepted: 06/17/2022] [Indexed: 09/08/2024] Open
Abstract
Tea tree oil (TTO) is a plant-derived additive with anti-inflammatory, bactericidal, and growth-promoting properties. However, little is known about the effects of TTO on intestinal amino acid transport and immune function in goats. Twenty-four Ganxi goats (initial body weight of 13.5 ± 0.70 kg) were randomly allotted two treatments and fed either control (CON) or CON+TTO (0.2 ml/kg) diet. The addition of TTO to the diet significantly decreased (p < .05) tumor necrosis factor-α content and increased (p < .05) interleukin-2 (IL-2) content in goat serum; significantly decreased (p < .05) IL-12, and increased (p < .05) IL-2 content in goat ileal mucosa; significantly increased (p < .05) secreted IgA content in the jejunal and ileal mucosa; significantly upregulated (p < .05) IL-2 and downregulated (p < .05) IL-12 at the mRNA level in the ileal mucosa; significantly elevated the levels of serine, arginine, and total amino acids in the ileal mucosa (p < .05); significantly upregulated (p < .05) SLC1A1 and SLC7A1 in the ileum; and significantly enhanced (p < .05) the protein expression of Claudin-1 in the ileal mucosa. In summary, adding 0.2 ml/kg of TTO to the diet enhanced SLC1A1 and SLC7A1 mRNA expression in the ileal mucosa, and SLC1A1 and SLC7A1 could transport serine and arginine from the chyme to the ileal mucosa. Thus, increased serine and arginine content in the mucosa could improve intestinal immunity. TTO supplementation upregulated the expression of IL-2 and Claudin-1 in goat ileal mucosa, and enhanced immune function in the intestine.
Collapse
Affiliation(s)
- Xiaokang Lv
- CAS Key Laboratory for Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic ProcessInstitute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
- College of Advanced AgriculturalUniversity of Chinese Academy of SciencesBeijingChina
| | - Liang Chen
- Shenyang Agricultural UniversityInstitute of Rural Revitalization StrategyShenyangChina
| | - Chuanshe Zhou
- CAS Key Laboratory for Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic ProcessInstitute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
- College of Advanced AgriculturalUniversity of Chinese Academy of SciencesBeijingChina
- School of AgricultureNingxia UniversityYinchuanChina
| | - Yibing Guo
- CAS Key Laboratory for Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic ProcessInstitute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Guijie Zhang
- School of AgricultureNingxia UniversityYinchuanChina
| | - Jinhe Kang
- CAS Key Laboratory for Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic ProcessInstitute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Zhiliang Tan
- CAS Key Laboratory for Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic ProcessInstitute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Shaoxun Tang
- CAS Key Laboratory for Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic ProcessInstitute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Zixin Liu
- CAS Key Laboratory for Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic ProcessInstitute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
- College of Advanced AgriculturalUniversity of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
48
|
DNA methyltransferase 3A controls intestinal epithelial barrier function and regeneration in the colon. Nat Commun 2022; 13:6266. [PMID: 36271073 PMCID: PMC9587301 DOI: 10.1038/s41467-022-33844-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 10/05/2022] [Indexed: 12/25/2022] Open
Abstract
Genetic variants in the DNA methyltransferase 3 A (DNMT3A) locus have been associated with inflammatory bowel disease (IBD). DNMT3A is part of the epigenetic machinery physiologically involved in DNA methylation. We show that DNMT3A plays a critical role in maintaining intestinal homeostasis and gut barrier function. DNMT3A expression is downregulated in intestinal epithelial cells from IBD patients and upon tumor necrosis factor treatment in murine intestinal organoids. Ablation of DNMT3A in Caco-2 cells results in global DNA hypomethylation, which is linked to impaired regenerative capacity, transepithelial resistance and intercellular junction formation. Genetic deletion of Dnmt3a in intestinal epithelial cells (Dnmt3aΔIEC) in mice confirms the phenotype of an altered epithelial ultrastructure with shortened apical-junctional complexes, reduced Goblet cell numbers and increased intestinal permeability in the colon in vivo. Dnmt3aΔIEC mice suffer from increased susceptibility to experimental colitis, characterized by reduced epithelial regeneration. These data demonstrate a critical role for DNMT3A in orchestrating intestinal epithelial homeostasis and response to tissue damage and suggest an involvement of impaired epithelial DNMT3A function in the etiology of IBD.
Collapse
|
49
|
Lim R, Banerjee A, Biswas R, Chari AN, Raghavan S. Mechanotransduction through adhesion molecules: Emerging roles in regulating the stem cell niche. Front Cell Dev Biol 2022; 10:966662. [PMID: 36172276 PMCID: PMC9511051 DOI: 10.3389/fcell.2022.966662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 07/18/2022] [Indexed: 11/23/2022] Open
Abstract
Stem cells have been shown to play an important role in regenerative medicine due to their proliferative and differentiation potential. The challenge, however, lies in regulating and controlling their potential for this purpose. Stem cells are regulated by growth factors as well as an array of biochemical and mechanical signals. While the role of biochemical signals and growth factors in regulating stem cell homeostasis is well explored, the role of mechanical signals has only just started to be investigated. Stem cells interact with their niche or to other stem cells via adhesion molecules that eventually transduce mechanical cues to maintain their homeostatic function. Here, we present a comprehensive review on our current understanding of the influence of the forces perceived by cell adhesion molecules on the regulation of stem cells. Additionally, we provide insights on how this deeper understanding of mechanobiology of stem cells has translated toward therapeutics.
Collapse
Affiliation(s)
- Ryan Lim
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Avinanda Banerjee
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Ritusree Biswas
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore, India
- Sastra University, Thanjavur, TN, India
| | - Anana Nandakumar Chari
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Srikala Raghavan
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore, India
| |
Collapse
|
50
|
Kirupananthan D, Bertolo RF, Brunton JA. Lysine Dipeptide Enhances Gut Structure and Whole-Body Protein Synthesis in Neonatal Piglets with Intestinal Atrophy. J Nutr 2022; 152:1843-1850. [PMID: 35481706 DOI: 10.1093/jn/nxac095] [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: 11/24/2021] [Revised: 03/28/2022] [Accepted: 04/21/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Parenteral nutrition (PN) is often a necessity for preterm infants; however, prolonged PN leads to gut atrophy, weakened gut barrier function, and a higher risk of intestinal infections. Peptide transporter-1 (PepT1) is a di- or tripeptide transporter in the gut and, unlike other nutrient transporters, its activity is preserved with the onset of intestinal atrophy from PN. As such, enteral amino acids in the form of dipeptides may be more bioavailable than free amino acids when atrophy is present. OBJECTIVES In Yucatan miniature piglets with PN-induced intestinal atrophy, we sought to determine the structural and functional effects of enteral refeeding with lysine as a dipeptide, compared to free L-lysine. METHODS Piglets aged 7-8 days were PN-fed for 4 days to induce intestinal atrophy, then were refed with enteral diets with equimolar lysine supplied as lysyl-lysine (Lys-Lys; n = 7), free lysine (n = 7), or Lys-Lys with glycyl-sarcosine (n = 6; to determine whether competitive inhibition of Lys-Lys uptake would abolish PepT1-mediated effects). The diets provided lysine at 75% of the requirement and were gastrically delivered for a total of 18 hours. Whole-body and tissue-specific protein synthesis, as well as indices for gut structure and barrier function, were measured. RESULTS The villus height, mucosal weight, and free lysine concentration were higher in the Lys-Lys group compared to the other 2 groups (P < 0.05). Lysyl-lysine led to greater whole-body protein synthesis compared to free lysine (P < 0.05). Mucosal myeloperoxidase activity was lower in the Lys-Lys group (P < 0.05), suggesting less inflammation. The inclusion of glycyl-sarcosine with Lys-Lys abolished the dipeptide effects on whole-body and tissue-specific protein synthesis (P < 0.05), suggesting that improved lysine availability was mediated by PepT1. CONCLUSIONS Improved intestinal structure and whole-body protein synthesis suggests that feeding strategies designed to exploit PepT1 may help to avoid adverse effects when enteral nutrition is reintroduced into the compromised guts of neonatal piglets.
Collapse
Affiliation(s)
| | - Robert F Bertolo
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Janet A Brunton
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL, Canada
| |
Collapse
|