1
|
Wang HQ, Zhu YW, Dou ZY, Chen Z, Tong CC, He X, Ma XH, Guan J, Xu DX, Chen X. 1,25(OH) 2D 3 Ameliorates DSS-induced Intestinal Ferroptosis through the SIRT3-SOD2-mtROS Pathway. J Nutr Biochem 2025:109999. [PMID: 40513839 DOI: 10.1016/j.jnutbio.2025.109999] [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: 02/23/2025] [Revised: 05/29/2025] [Accepted: 06/09/2025] [Indexed: 06/16/2025]
Abstract
Ferroptosis has been shown to play a significant role in the pathogenesis of ulcerative colitis (UC). This study investigated the effects of 1,25(OH)2D3 supplementation on ferroptosis in dextran sulfate sodium (DSS)-evoked colitis. Intestinal VDR was reduced in UC patients. Accordingly, GPX4 was downregulated and ACSL4 was upregulated in the intestine of UC patients. Animal experiments indicated that vitamin D deficiency exacerbated DSS-induced intestinal ferroptosis in mice. Conversely, pretreatment with 1,25(OH)2D3 alleviated DSS-induced ferroptosis in mouse intestine. Similarly, 1,25(OH)2D3 supplementation inhibited DSS-induced ferroptosis in HT-29 cells. Furthermore, we found decreased intestinal SIRT3 protein and increased acetylated superoxide dismutase 2 (Ac-SOD2) in UC patients. Pretreatment with 1,25(OH)2D3 attenuated DSS-induced downregulation of SIRT3 and acetylation of SOD2 in both mouse intestine and HT-29 cells. Moreover, 1,25(OH)2D3 pretreatment inhibited mitochondrial reactive oxygen species (mtROS) in DSS-treated HT-29 cells. Finally, transfection with SIRT3 siRNA antagonized the protective effect of 1,25(OH)2D3 on ferroptosis in DSS-treated HT-29 cells. Overall, our results suggest that 1,25(OH)2D3 alleviates DSS-induced intestinal ferroptosis via the SIRT3-SOD2-mtROS pathway, further supporting the potential use of 1,25(OH)2D3 supplementation in UC treatment.
Collapse
Affiliation(s)
- Hong-Qian Wang
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Provincial Key Laboratory of Digestive Diseases, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ya-Wen Zhu
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Provincial Key Laboratory of Digestive Diseases, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zi-Yue Dou
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Provincial Key Laboratory of Digestive Diseases, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhuo Chen
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Provincial Key Laboratory of Digestive Diseases, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Cheng-Cheng Tong
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Provincial Key Laboratory of Digestive Diseases, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xue He
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Provincial Key Laboratory of Digestive Diseases, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiao-Han Ma
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Provincial Key Laboratory of Digestive Diseases, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jing Guan
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Provincial Key Laboratory of Digestive Diseases, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - De-Xiang Xu
- Department of Toxicology, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China.
| | - Xi Chen
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Provincial Key Laboratory of Digestive Diseases, First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
2
|
Gao Y, Kong D, Sun JX, Ma ZX, Wang GQ, Ma XF, Sun L, Luo HY, Xu Y, Wang KH. Intestinal barrier damage caused by addictive substance use disorder. Eur J Med Res 2025; 30:226. [PMID: 40176069 PMCID: PMC11963533 DOI: 10.1186/s40001-025-02446-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 03/10/2025] [Indexed: 04/04/2025] Open
Abstract
Addictive substance use disorder has a wide range of effects on the intestinal barrier, including damage to the biological, chemical, mechanical, and immune barriers. Damage to the intestinal barrier caused by addictive substance use disorder allows harmful substances and bacteria to cross the intestinal barrier into the circulatory system, leading to systemic inflammatory responses and immune imbalances. In addition, the interaction between the gut flora and the central nervous system is recognized as an important component of the gut-brain axis. Gut barrier damage leads to dysbiosis, which in turn affects brain function by activating immune cells and releasing inflammatory factors. This may lead to altered mood and cognitive function, increased addictive substance cravings, and dependence. Recent research has indicated that reshaping the gut-brain axis and adjusting the composition and abundance of gut microbiota holds promise in alleviating withdrawal symptoms with addictive substance dependence. This article reviews the effects of addictive substance use disorder on the intestinal barrier and explores the possibility of improving addictive substance dependence by treating gut barrier damage.
Collapse
Affiliation(s)
- Yan Gao
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, 650504, China
| | - Deshenyue Kong
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, 650504, China
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Jia-Xue Sun
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, 650504, China
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Zhong-Xu Ma
- Third People's Hospital of Kunming City, Kunming, 650041, China
| | - Guang-Qing Wang
- Drug Rehabilitation Administration of Yunnan Province, Kunming, 650032, China
| | - Xing-Feng Ma
- Drug Rehabilitation Administration of Yunnan Province, Kunming, 650032, China
| | - Liang Sun
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Hua-You Luo
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
| | - Yu Xu
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, 650504, China.
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
| | - Kun-Hua Wang
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, 650504, China.
| |
Collapse
|
3
|
Zhang XQ, Li JM, Wang FQ, Ren YH, Wu SX, Wu Y, Tang Y. The clinical significance and biological function of tropomyosin 3 in ulcerative colitis. Tissue Cell 2025; 93:102770. [PMID: 39938429 DOI: 10.1016/j.tice.2025.102770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/14/2025]
Abstract
BACKGROUND Ulcerative colitis (UC) is a lifelong chronic inflammatory disease that is characterized by the absence of specific markers for diagnosis and prognosis. TPM3 is an integral component of the thin filament, responsible for the structural stability of actin filaments and modulation of cytoskeletal function. This study investigated the regulatory role of TPM3 in UC and its potential mechanisms. METHODS At the clinical level, TPM3 levels were assessed in serum and mucosal tissues of UC and other enteric disease. At the cellular level, the effects of TMP3 overexpressing lentivirus on Caco-2 cell phenotype and the barrier of IL-1β-induced UC model were explored. At the animal level, the effects of TMP3 overexpressing lentivirus on symptoms and colonic damage in a DSS-induced UC model were explored. RESULTS TPM3 expression in serum of UC patients was significantly lower than that of other enteric disease, and TPM3 levels in the intestinal mucosa showed a negative correlation with the Mayo score of UC patients. TPM3 overexpression alleviates IL-1β-induced apoptosis and inhibition of invasion and migration in UC model in vitro. In monolayer Caco-2 cells, TPM3 overexpression rescued the IL-1β-induced decrease in transepithelial electrical resistance and tight junction markers (ZO-1 and Occludin) and increase in permeability. In animal experiments, TPM3 overexpression increased body weight and colon length and decreased disease activity index in a DSS-induced UC model. In tissue staining, it alleviated pathological damage and upregulated Occuludin and TPM3 levels in the colon. CONCLUSION TPM3 levels correlated with UC disease course and TPM3 overexpression alleviated symptoms/phenotypes and barrier damage in UC models in vivo and in vitro. TPM3 may serve as a potential novel biomarker for UC diagnosis and prognosis.
Collapse
Affiliation(s)
- Xue-Qin Zhang
- The First People's Hospital of Qujing, No. 1, Yuanlin Road, Qujing, Yunnan 655000, China
| | - Jian-Mei Li
- The First People's Hospital of Qujing, No. 1, Yuanlin Road, Qujing, Yunnan 655000, China
| | - Feng-Qian Wang
- The First People's Hospital of Qujing, No. 1, Yuanlin Road, Qujing, Yunnan 655000, China
| | - Yan-Hui Ren
- The First People's Hospital of Qujing, No. 1, Yuanlin Road, Qujing, Yunnan 655000, China
| | - Shi-Xian Wu
- The First People's Hospital of Qujing, No. 1, Yuanlin Road, Qujing, Yunnan 655000, China
| | - Yao Wu
- The First People's Hospital of Qujing, No. 1, Yuanlin Road, Qujing, Yunnan 655000, China
| | - Yuan Tang
- The First People's Hospital of Qujing, No. 1, Yuanlin Road, Qujing, Yunnan 655000, China.
| |
Collapse
|
4
|
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
|
5
|
Gao C, Yang Z, Song R, Sheng H, Zhu L. Nanotechnology-based drug delivery system for targeted therapy of ulcerative colitis from traditional Chinese medicine: A review. Int J Pharm 2025; 673:125375. [PMID: 39965734 DOI: 10.1016/j.ijpharm.2025.125375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/27/2025] [Accepted: 02/15/2025] [Indexed: 02/20/2025]
Abstract
Ulcerative colitis (UC) is a chronic autoimmune disease and seriously affects the normal life of patients. Conventional therapeutic drugs are difficult to meet clinical needs. Traditional Chinese medicine (TCM) ingredients could effectively alleviate the symptoms of UC by anti-inflammatory, anti-oxidative, regulating the gut microbiota, and repairing the colonic epithelial barrier, but their low solubility and bioavailability severely limit their clinical application. Nano-drug delivery systems (NDDS) combined with TCM ingredients is a promising option for treating UC, and they could significantly enhance the stability, solubility, and bioavailability of TCM ingredients. The review describes the anti-UC mechanisms of TCM ingredients, systematically summarizes various kinds of NDDS for TCM ingredients according to different routes of administration, and highlights the advantages of NDDS for TCM ingredients in the treatmentof UC. In addition, we discuss the limitations of existing NDDS for TCM ingredients and the development direction in the future. This review will provide a basis for the future development of anti-UC NDDS for TCM ingredients.
Collapse
Affiliation(s)
- Chengcheng Gao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Zerun Yang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Ruirui Song
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Huagang Sheng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Liqiao Zhu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| |
Collapse
|
6
|
Jiang X, Ren J, Yu G, Wu W, Chen M, Zhao Y, He C. Targeting Bile-Acid Metabolism: Nutritional and Microbial Approaches to Alleviate Ulcerative Colitis. Nutrients 2025; 17:1174. [PMID: 40218932 PMCID: PMC11990178 DOI: 10.3390/nu17071174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/19/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory disease affecting the colorectum, posing a significant global health burden. Recent studies highlight the critical role of gut microbiota and its metabolites, particularly bile acids (BAs), in UC's pathogenesis. The relationship between BAs and gut microbiota is bidirectional: microbiota influence BA composition, while BAs regulate microbiota diversity and activity through receptors like Farnesoid X receptor (FXR) and Takeda G protein-coupled receptor 5 (TGR5). Targeting bile-acid metabolism to reshape gut microbiota presents a promising therapeutic strategy for UC. This review examines the classification and synthesis of BAs, their interactions with gut microbiota, and the potential of nutritional and microbial interventions. By focusing on these therapies, we aim to offer innovative approaches for effective UC management.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Canxia He
- School of Public Health, Health Science Center, Ningbo University, Ningbo 315211, China
| |
Collapse
|
7
|
Simon P, Török É, Szalontai K, Kari B, Neuperger P, Zavala N, Kanizsai I, Puskás LG, Török S, Szebeni GJ. Nutritional Support of Chronic Obstructive Pulmonary Disease. Nutrients 2025; 17:1149. [PMID: 40218907 PMCID: PMC11990120 DOI: 10.3390/nu17071149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/14/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Background: COPD is a heterogenous disease of the respiratory tract caused by diverse genetic factors along with environmental and lifestyle-related effects such as industrial dust inhalation and, most frequently, cigarette smoking. These factors lead to airflow obstruction and chronic respiratory symptoms. Additionally, the increased risk of infections exacerbates airway inflammation in COPD patients. As a consequence of the complex pathomechanisms and difficulty in treatment, COPD is among the leading causes of mortality both in the western countries and in the developing world. Results: The management of COPD is still a challenge for the clinicians; however, alternative interventions such as smoking cessation and lifestyle changes from a sedentary life to moderate physical activity with special attention to the diet may ameliorate patients' health. Here, we reviewed the effects of different dietary components and supplements on the conditions of COPD. Conclusions: COPD patients are continuously exposed to heavy metals, which are commonly present in cigarette smoke and polluted air. Meanwhile, they often experience significant nutrient deficiencies, which affect the detoxification of these toxic metals. This in turn can further disrupt nutritional balance by interfering with the absorption, metabolism, and utilization of essential micronutrients. Therefore, awareness and deliberate efforts should be made to check levels of micronutrients, with special attention to ensuring adequate levels of antioxidants, vitamin D, vitamin K2, magnesium, and iron, as these may be particularly important in reducing the risk of COPD development and limiting disease severity.
Collapse
Grants
- 2023-1.1.1-PIACI_FÓKUSZ-2024-00036 National Research, Development, and Innovation Office (NKFI), Hungary
- 2020-1.1.6-JÖVŐ-2021-00003 National Research, Development, and Innovation Office (NKFI), Hungary
- 2022-1.2.6-TÉT-IPARI-TR-2022-00023 National Research, Development, and Innovation Office (NKFI), Hungary
- 142877 FK22 National Research, Development, and Innovation Office (NKFI), Hungary.
- BO/00582/22/8 János Bolyai Research Scholarship of the Hungarian Academy of Sciences
Collapse
Affiliation(s)
- Péter Simon
- National Korányi Institute of Pulmonology, 1121 Budapest, Hungary;
| | - Éva Török
- Gastroenterology Center Buda, 1117 Budapest, Hungary;
| | - Klára Szalontai
- Department of Pulmonology, Szent-Györgyi Albert Medical Center, University of Szeged, 6772 Deszk, Hungary;
| | - Beáta Kari
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.K.); (P.N.); (N.Z.); (L.G.P.)
| | - Patrícia Neuperger
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.K.); (P.N.); (N.Z.); (L.G.P.)
| | - Norma Zavala
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.K.); (P.N.); (N.Z.); (L.G.P.)
| | | | - László G. Puskás
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.K.); (P.N.); (N.Z.); (L.G.P.)
- Anthelos Ltd., 6726 Szeged, Hungary
| | - Szilvia Török
- National Korányi Institute of Pulmonology, 1121 Budapest, Hungary;
| | - Gabor J. Szebeni
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.K.); (P.N.); (N.Z.); (L.G.P.)
- Department of Internal Medicine, Hematology Centre, Faculty of Medicine, University of Szeged, 6725 Szeged, Hungary
| |
Collapse
|
8
|
Li H, Han L, Zong Y, Feng R, Chen W, Geng J, Li J, Zhao Y, Wang Y, He Z, Du R. Deer oil improves ulcerative colitis induced by DSS in mice by regulating the intestinal microbiota and SCFAs metabolism and modulating NF-κB and Nrf2 signaling pathways. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:382-393. [PMID: 39189446 DOI: 10.1002/jsfa.13837] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 08/02/2024] [Accepted: 08/12/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Deer oil (DO), a byproduct of deer meat processing, possesses high nutritional value. This study aims to evaluate the protective effects of DO on dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) in mice and to explore its potential mechanisms of action. RESULTS DO was found to inhibit weight loss and colon shortening in colitis mice, significantly reduce disease activity index scores, and notably enhance the levels of tight junction proteins in colon tissues, thus improving intestinal barrier function. ELISA results indicated that DO markedly alleviated the mice's oxidative stress and inflammatory responses. Western blot analysis further demonstrated that DO significantly inhibited the phosphorylation of NF-κB while up-regulating the expression levels of Nrf2 and HO-1 proteins. Additionally, DO increased the abundance of beneficial bacteria such as Odoribacter, Blautia, and Muribaculum, reduced the abundance of harmful bacteria such as Bacteroides, Helicobacter, and Escherichia-Shigella, and promoted the production of short-chain fatty acids. CONCLUSION Our study provides the first evidence that DO can effectively improve DSS-induced UC in mice. The underlying mechanisms may involve maintaining intestinal barrier function, inhibiting inflammation, alleviating oxidative stress, and modulation of gut microbiota. These findings offer valuable insights for developing DO as an adjunct treatment for UC and as a functional food. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Hongyan Li
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun, China
| | - Lu Han
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun, China
| | - Ying Zong
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun, China
| | - Ruyi Feng
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun, China
| | - Weijia Chen
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun, China
| | - Jianan Geng
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun, China
| | - Jianming Li
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun, China
| | - Yan Zhao
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun, China
| | - Yuqi Wang
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun, China
| | - Zhongmei He
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun, China
| | - Rui Du
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun, China
- Engineering Research Center for High Efficiency Breeding and Product Development Technology of SikaDeer, Changchun, China
| |
Collapse
|
9
|
Arumugam P, Saha K, Nighot P. Intestinal Epithelial Tight Junction Barrier Regulation by Novel Pathways. Inflamm Bowel Dis 2025; 31:259-271. [PMID: 39321109 DOI: 10.1093/ibd/izae232] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Indexed: 09/27/2024]
Abstract
Intestinal epithelial tight junctions (TJs), a dynamically regulated barrier structure composed of occludin and claudin family of proteins, mediate the interaction between the host and the external environment by allowing selective paracellular permeability between the luminal and serosal compartments of the intestine. TJs are highly dynamic structures and can undergo constant architectural remodeling in response to various external stimuli. This is mediated by an array of intracellular signaling pathways that alters TJ protein expression and localization. Dysfunctional regulation of TJ components compromising the barrier homeostasis is an important pathogenic factor for pathological conditions including inflammatory bowel disease (IBD). Previous studies have elucidated the significance of TJ barrier integrity and key regulatory mechanisms through various in vitro and in vivo models. In recent years, considerable efforts have been made to understand the crosstalk between various signaling pathways that regulate formation and disassembly of TJs. This review provides a comprehensive view on the novel mechanisms that regulate the TJ barrier and permeability. We discuss the latest evidence on how ion transport, cytoskeleton and extracellular matrix proteins, signaling pathways, and cell survival mechanism of autophagy regulate intestinal TJ barrier function. We also provide a perspective on the context-specific outcomes of the TJ barrier modulation. The knowledge on the diverse TJ barrier regulatory mechanisms will provide further insights on the relevance of the TJ barrier defects and potential target molecules/pathways for IBD.
Collapse
Affiliation(s)
- Priya Arumugam
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA, USA
| | - Kushal Saha
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Prashant Nighot
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA, USA
| |
Collapse
|
10
|
Kumar A, Jayawardena D, Priyamvada S, Anbazhagan AN, Chatterjee I, Saksena S, Dudeja PK. SLC26A3 (DRA, the Congenital Chloride Diarrhea Gene): A Novel Therapeutic Target for Diarrheal Diseases. Cell Mol Gastroenterol Hepatol 2024; 19:101452. [PMID: 39736385 PMCID: PMC12003007 DOI: 10.1016/j.jcmgh.2024.101452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 12/22/2024] [Accepted: 12/22/2024] [Indexed: 01/01/2025]
Abstract
Diarrhea associated with enteric infections, gut inflammation, and genetic defects poses a major health burden and results in significant morbidity and mortality. Impaired fluid and electrolyte absorption or secretion in the intestine are the hallmark of diarrhea. Electroneutral NaCl absorption in the mammalian GI tract involves the coupling of Na+/H+ and Cl-/HCO3- exchangers. SLC26A3 (Down Regulated in Adenoma, DRA) is the major anion exchanger involved in luminal Cl- absorption and HCO3- secretion. Mutations in the SLC26A3 gene cause a severe disease called congenital chloride diarrhea (CLD). Multiple studies have shown that DRA function or expression is downregulated in infectious diarrheal disorders caused by EPEC, C rodentium, Salmonella, Clostridioides difficile and Cryptosporidium parvum infection. In addition, DRA levels are severely depleted in colonic mucosa of IBD patients and in mouse models of IBD (eg, DSS, TNBS, adoptive T-cell transfer, anti-CD-40, and IL-10 KO colitis). In addition, genetic defects exhibiting diarrhea including microvillus inclusion disease (MVID), keratin-8 depletion, knock-out mouse models of transcriptional factors (eg, CDX-2 and HNF1α/1β) also exhibit severe down regulation of DRA. Also, recent studies have shown that DRA is not only critical for chloride absorption but also plays a key role in maintaining gut epithelial barrier integrity, microbiome composition, and has now emerged as an IBD susceptibility gene. In this review, we provide strong evidence that DRA may serve as a novel therapeutic target with dual benefits in not only correcting diarrheal phenotype but also improving gut barrier integrity and inflammation in pathogen infection or IBD.
Collapse
Affiliation(s)
- Anoop Kumar
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, Illinois; Jesse Brown VA Medical Center, Chicago, Illinois
| | - Dulari Jayawardena
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, Illinois
| | - Shubha Priyamvada
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, Illinois
| | - Arivarasu N Anbazhagan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, Illinois
| | - Ishita Chatterjee
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, Illinois
| | - Seema Saksena
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, Illinois; Jesse Brown VA Medical Center, Chicago, Illinois
| | - Pradeep K Dudeja
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, Illinois; Jesse Brown VA Medical Center, Chicago, Illinois.
| |
Collapse
|
11
|
Kesaraju S, Li Y, Xing J, Tracy M, Wannemo K, Holder A, Zhao P, Khan MA, Kainov J, Rana N, Sidahmed M, Hyoju S, Smith L, Matthews J, Tay S, Khalili-Araghi F, Rana M, Oakes SA, Shen L, Weber CR. Inflammation-Induced Claudin-2 Upregulation Limits Pancreatitis Progression by Enhancing Tight Junction-Controlled Pancreatic Ductal Transport. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.01.555960. [PMID: 39605652 PMCID: PMC11601259 DOI: 10.1101/2023.09.01.555960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Pancreatitis is an inflammatory disease of the pancreas that can arise due to various factors, including environmental risks such as diet, alcohol, and smoking, as well as genetic predispositions. In some cases, pancreatitis may progress and become chronic, leading to irreversible damage and impaired pancreatic function. Genome-wide association studies (GWAS) have identified polymorphisms at the X-linked CLDN2 locus as risk factors for both sporadic and alcohol-related chronic pancreatitis. CLDN2 encodes claudin-2 (CLDN2), a paracellular cation-selective channel localized at tight junctions and expressed in the pancreas and other secretory organs. However, whether and how CLDN2 may modify pancreatitis susceptibility remains poorly understood. We aimed to clarify the potential role of CLDN2 in the onset and progression of pancreatitis. We employed multiple methodologies to examine the role of CLDN2 in human pancreatic tissue, caerulein-induced experimental pancreatitis mouse model, and pancreatic ductal epithelial organoids. In both human chronic pancreatitis tissues and caerulein-induced experimental pancreatitis, CLDN2 protein was significantly upregulated in pancreatic ductal epithelial cells. Our studies using pancreatic ductal epithelial organoids and mice demonstrated the inflammatory cytokine IFNγ upregulates claudin-2 expression at both RNA and protein levels. Following caerulein treatment, Ifng KO mice had diminished upregulation of CLDN2 relative to WT mice, indicating that caerulein-induced claudin-2 expression is partially driven by IFNγ. Functionally, Cldn2 knockout mice developed more severe caerulein-induced experimental pancreatitis, indicating CLDN2 plays a protective role in pancreatitis development. Pancreatic ductal epithelial organoid-based studies demonstrated that CLDN2 is critical for sodium-dependent water transport and necessary for cAMP-driven, CFTR-dependent fluid secretion. These findings suggest that functional crosstalk between CLDN2 and CFTR is essential for fluid transport in pancreatic ductal epithelium, which may protect against pancreatitis by adjusting pancreatic ductal secretion to prevent worsening autodigestion and inflammation. In conclusion, our studies suggest CLDN2 upregulation during pancreatitis may play a protective role in limiting disease development, and decreased CLDN2 function may increase pancreatitis severity. These results point to the possibility of modulating pancreatic ductal CLDN2 function as an approach for therapeutic intervention of pancreatitis.
Collapse
|
12
|
Zhang Q, Xiao W, Yu L, Tian F, Zhao J, Zhang H, Chen W, Zhai Q. Ameliorative effects of
Lactobacillus fermentum isolated from individuals following vegan, omnivorous and high-meat diets on ulcerative colitis in mice. FOOD SCIENCE AND HUMAN WELLNESS 2024; 13:3181-3192. [DOI: 10.26599/fshw.2023.9250005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
13
|
Kistler W, Villiger M, Villiger B, Yazici D, Pat Y, Mitamura Y, Ardicli S, Skolnick S, Dhir R, Akdis M, Nadeau K, Ogulur I, Akdis CA. Epithelial barrier theory in the context of nutrition and environmental exposure in athletes. Allergy 2024; 79:2912-2923. [PMID: 39011970 DOI: 10.1111/all.16221] [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: 03/27/2024] [Revised: 06/18/2024] [Accepted: 06/28/2024] [Indexed: 07/17/2024]
Abstract
Exposure to toxic substances, introduced into our daily lives during industrialization and modernization, can disrupt the epithelial barriers in the skin, respiratory, and gastrointestinal systems, leading to microbial dysbiosis and inflammation. Athletes and physically active individuals are at increased risk of exposure to agents that damage the epithelial barriers and microbiome, and their extreme physical exercise exerts stress on many organs, resulting in tissue damage and inflammation. Epithelial barrier-damaging substances include surfactants and enzymes in cleaning products, laundry and dishwasher detergents, chlorine in swimming pools, microplastics, air pollutants such as ozone, particulate matter, and diesel exhaust. Athletes' high-calorie diet often relies on processed foods that may contain food emulsifiers and other additives that may cause epithelial barrier dysfunction and microbial dysbiosis. The type of the material used in the sport equipment and clothing and their extensive exposure may increase the inflammatory effects. Excessive travel-related stress, sleep disturbances and different food and microbe exposure may represent additional factors. Here, we review the detrimental impact of toxic agents on epithelial barriers and microbiome; bring a new perspective on the factors affecting the health and performance of athletes and physically active individuals.
Collapse
Affiliation(s)
- Walter Kistler
- Medical Committee International Ice Hockey Federation, Zürich, Switzerland
- Swiss Research Institute for Sports Medicine (SRISM), Davos, Switzerland
- Department of Sports Medicine, Davos Hospital, Davos, Switzerland
| | - Michael Villiger
- Swiss Research Institute for Sports Medicine (SRISM), Davos, Switzerland
- Department of Sports Medicine, Davos Hospital, Davos, Switzerland
| | - Beat Villiger
- Swiss Research Institute for Sports Medicine (SRISM), Davos, Switzerland
- Department of Sports Medicine, Davos Hospital, Davos, Switzerland
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sena Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Stephen Skolnick
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Seed Health Inc., Los Angeles, California, USA
| | - Raja Dhir
- Seed Health Inc., Los Angeles, California, USA
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Kari Nadeau
- Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Research Institute for Sports Medicine (SRISM), Davos, Switzerland
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| |
Collapse
|
14
|
Al-Failakawi A, Al-Jarallah A, Rao M, Khan I. The Role of Claudins in the Pathogenesis of Dextran Sulfate Sodium-Induced Experimental Colitis: The Effects of Nobiletin. Biomolecules 2024; 14:1122. [PMID: 39334888 PMCID: PMC11430412 DOI: 10.3390/biom14091122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND The pathogenesis of inflammatory bowel diseases such as ulcerative colitis and Crohn's disease is not well understood. This study investigated the roles and regulation of the claudin-1, -2, -3, and -4 isoforms in the pathogenesis of ulcerative colitis, and the potential therapeutic effects of nobiletin. METHODS Colitis was induced in rats by administering dextran sulfate sodium [DSS] in drinking water for seven days. Animals were treated daily with nobiletin [oral, 60 mg/Kg body weight] and studied in four groups, C [non-colitis control], D [DSS-induced colitis], CN [nobiletin-treated non-colitis control], and DN [nobiletin-treated DSS-induced colitis]. On day seven, the animals were sacrificed, and colonic tissues were collected and analyzed. RESULTS Both macroscopic and microscopic findings suggest the progression of colitis. In the inflamed colon, claudin-1 and -4 proteins were decreased, claudin-2 increased, while the claudin-3 protein remained unchanged. Except for claudin-1, these changes were not paralleled by mRNA expression, indicating a complex regulatory mechanism. Uniform β-actin expression along with consistent quality and yield of total RNA indicated selectivity of these changes. Nobiletin treatment reversed these changes. CONCLUSIONS Altered expression of the claudin isoforms -1, -2, and -4 disrupts tight junctions, exposing the lamina propria to microflora, leading to electrolyte disturbance and the development of ulcerative colitis. Nobiletin with its anti-inflammatory properties may be useful in IBD.
Collapse
Affiliation(s)
- Asmaa Al-Failakawi
- Department of Biochemistry, College of Medicine, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait; (A.A.-F.); (A.A.-J.)
| | - Aishah Al-Jarallah
- Department of Biochemistry, College of Medicine, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait; (A.A.-F.); (A.A.-J.)
| | - Muddanna Rao
- Departments of Anatomy, College of Medicine, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait;
| | - Islam Khan
- Department of Biochemistry, College of Medicine, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait; (A.A.-F.); (A.A.-J.)
| |
Collapse
|
15
|
Peng L, Shi Y, Deng J, Chen J, Xiang P, Zhong X. DANCR maintained colon epithelial homeostasis by regulating the TNFα/NF-κB pathway. Biochem Biophys Res Commun 2024; 723:150176. [PMID: 38820627 DOI: 10.1016/j.bbrc.2024.150176] [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: 05/13/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/02/2024]
Abstract
Epithelial homeostasis is fundamental for the physiological functions of colon tissue. Dysregulation of colon epithelial structure leads to abnormal immune responses and diseases such as inflammatory bowel disease. In this work we found long non-coding RNA DANCR was a novel regulator to colon epithelial homeostasis. Silencing DANCR resulted in decreased expression of epithelial barrier proteins and enhanced susceptibility to TNFα stimulation, which was accompanied by hyperactivation of the NF-κB pathway. Mechanistical studies revealed DANCR modulated the expression of a protein methyltransferase SET7 to suppress responses to TNFα, as well as the activity of NF-κB pathway. In summary, DANCR regulated colon epithelial homeostasis through modulating the TNFα/NF-κB axis. These findings cast light on the discovery of novel regulators to colon epithelial homeostasis and added new evidence to the physiological functions of DANCR.
Collapse
Affiliation(s)
- Limei Peng
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Center for Stem Cell Biology and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-Sen University, 74 Zhongshan Road 2nd, Guangzhou, 510080, China
| | - Yingpeng Shi
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Center for Stem Cell Biology and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-Sen University, 74 Zhongshan Road 2nd, Guangzhou, 510080, China
| | - Jiacheng Deng
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Center for Stem Cell Biology and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-Sen University, 74 Zhongshan Road 2nd, Guangzhou, 510080, China
| | - Jieyi Chen
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Center for Stem Cell Biology and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-Sen University, 74 Zhongshan Road 2nd, Guangzhou, 510080, China
| | - Peng Xiang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Center for Stem Cell Biology and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-Sen University, 74 Zhongshan Road 2nd, Guangzhou, 510080, China
| | - Xiaomin Zhong
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Center for Stem Cell Biology and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-Sen University, 74 Zhongshan Road 2nd, Guangzhou, 510080, China.
| |
Collapse
|
16
|
Zhao Y, Si S, Ren Y, Wu X, Zhang Z, Tian Y, Li J, Li Y, Hou M, Yao X, Xu Z, Jiang R, Kang X, Gong Y, Li Q, Tian Y. Marine red yeast supplementation improves laying performance by regulating small intestinal homeostasis in aging chickens. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 18:177-190. [PMID: 39263442 PMCID: PMC11388669 DOI: 10.1016/j.aninu.2024.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/25/2024] [Accepted: 04/03/2024] [Indexed: 09/13/2024]
Abstract
Recent studies have shown that age-related aging evolution is accompanied by imbalances in intestinal homeostasis. Marine red yeast (MRY) is a functional probiotic that has been shown to have antioxidant, immune and other properties. Therefore, we chose 900 healthy Hy-Line Brown hens at 433 d old as the research subjects and evaluated the correlation between intestinal health, laying performance, and egg quality in aged hens through the supplementation of MRY. These laying hens were assigned into 5 groups and received diet supplementation with 0%, 0.5%, 1.0%, 1.5%, and 2% MRY for 12 weeks. The results showed that MRY supplementation increased egg production rate, average egg weight, and egg quality, and decreased feed conversion ratio and daily feed intake (P < 0.05). The MRY supplement improved antioxidant indicators such as superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), stimulated villus height, and increased the villus height to crypt depth ratio (V/C ratio) in the intestine (P < 0.05). It also regulated the expression of intestinal inflammatory factors (transforming growth factor-β [TGF-β], interleukin [IL]-1β, IL-8, tumor necrosis factor-α [TNF-α]) while increasing serum immunoglobulin G (IgG) levels (P < 0.05). Furthermore, MRY supplementation upregulated the mRNA expression of tight junction proteins (occludin and zonula occludens-1 [ZO-1]), anti-apoptotic gene (Bcl-2), and autophagy-related proteins (beclin-1 and light chain 3I [LC3I]) in the intestine (P < 0.05). The MRY supplement also led to an increase in the concentration of short-chain fatty acids in the cecum, and the relative abundance of the phylum Bacteroidetes, and genera Bacteroides and Rikenellaceae_RC9_gut_group. The LEfSe analysis revealed an enrichment of Sutterella and Akkermansia muciniphila. In conclusion, the results of this experiment indicated that the additional supplementation of MRY can improve the production performance of laying hens and may contribute to the restoration and balance of intestinal homeostasis, which supports the application potential of MRY as a green and efficient feed additive for improving the laying performance in chickens.
Collapse
Affiliation(s)
- Yudian Zhao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Sujin Si
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Yangguang Ren
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Xing Wu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Zihao Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Yixiang Tian
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Jing Li
- AB Vista, Marlborough SN8 4AN, UK
| | - Yijie Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Meng Hou
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Xueyang Yao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Zhaoheng Xu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Ruirui Jiang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Xiangtao Kang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Yujie Gong
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Qiang Li
- Henan College of Animal Husbandry and Economics, Zhengzhou 450046, China
| | - Yadong Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| |
Collapse
|
17
|
Rajasekaran JJ, Krishnamurthy HK, Bosco J, Jayaraman V, Krishna K, Wang T, Bei K. Oral Microbiome: A Review of Its Impact on Oral and Systemic Health. Microorganisms 2024; 12:1797. [PMID: 39338471 PMCID: PMC11434369 DOI: 10.3390/microorganisms12091797] [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: 07/01/2024] [Revised: 08/07/2024] [Accepted: 08/16/2024] [Indexed: 09/30/2024] Open
Abstract
PURPOSE OF REVIEW This review investigates the oral microbiome's composition, functions, influencing factors, connections to oral and systemic diseases, and personalized oral care strategies. RECENT FINDINGS The oral microbiome is a complex ecosystem consisting of bacteria, fungi, archaea, and viruses that contribute to oral health. Various factors, such as diet, smoking, alcohol consumption, lifestyle choices, and medical conditions, can affect the balance of the oral microbiome and lead to dysbiosis, which can result in oral health issues like dental caries, gingivitis, periodontitis, oral candidiasis, and halitosis. Importantly, our review explores novel associations between the oral microbiome and systemic diseases including gastrointestinal, cardiovascular, endocrinal, and neurological conditions, autoimmune diseases, and cancer. We comprehensively review the efficacy of interventions like dental probiotics, xylitol, oral rinses, fluoride, essential oils, oil pulling, and peptides in promoting oral health by modulating the oral microbiome. SUMMARY This review emphasizes the critical functions of the oral microbiota in dental and overall health, providing insights into the effects of microbial imbalances on various diseases. It underlines the significant connection between the oral microbiota and general health. Furthermore, it explores the advantages of probiotics and other dental care ingredients in promoting oral health and addressing common oral issues, offering a comprehensive strategy for personalized oral care.
Collapse
Affiliation(s)
- John J. Rajasekaran
- Vibrant Sciences LLC, Santa Clara, CA 95054, USA; (H.K.K.); (V.J.); (K.K.); (T.W.); (K.B.)
| | | | - Jophi Bosco
- Vibrant America LLC, Santa Clara, CA 95054, USA;
| | - Vasanth Jayaraman
- Vibrant Sciences LLC, Santa Clara, CA 95054, USA; (H.K.K.); (V.J.); (K.K.); (T.W.); (K.B.)
| | - Karthik Krishna
- Vibrant Sciences LLC, Santa Clara, CA 95054, USA; (H.K.K.); (V.J.); (K.K.); (T.W.); (K.B.)
| | - Tianhao Wang
- Vibrant Sciences LLC, Santa Clara, CA 95054, USA; (H.K.K.); (V.J.); (K.K.); (T.W.); (K.B.)
| | - Kang Bei
- Vibrant Sciences LLC, Santa Clara, CA 95054, USA; (H.K.K.); (V.J.); (K.K.); (T.W.); (K.B.)
| |
Collapse
|
18
|
Jeong YJ, Lee HR, Park SA, Lee JW, Kim LK, Kim HJ, Seo JH, Heo TH. A derivative of 3-(1,3-diarylallylidene)oxindoles inhibits dextran sulfate sodium-induced colitis in mice. Pharmacol Rep 2024; 76:851-862. [PMID: 38916850 PMCID: PMC11294400 DOI: 10.1007/s43440-024-00616-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 06/15/2024] [Accepted: 06/17/2024] [Indexed: 06/26/2024]
Abstract
BACKGROUND IA-0130 is a derivative of 3-(1,3-diarylallylidene)oxindoles, which is a selective estrogen receptor modulator (SERM). A previous study demonstrated that SERM exhibits anti-inflammatory effects on colitis by promoting the anti-inflammatory phenotype of monocytes in murine colitis. However, the therapeutic effects of oxindole on colitis remain unknown. Therefore, we evaluated the efficacy of IA-0130 on dextran sulfate sodium (DSS)-induced mouse colitis. METHODS The DSS-induced colitis mouse model was established by administration of 2.5% DSS for 5 days. Mice were orally administered with IA-0130 (0.01 mg/kg or 0.1 mg/kg) or cyclosporin A (CsA; 30 mg/kg). Body weight, disease activity index score and colon length of mice were calculated and histological features of mouse colonic tissues were analyzed using hematoxylin and eosin staining. The expression of inflammatory cytokines and tight junction (TJ) proteins were analyzed using quantitative real-time PCR and enzyme-linked immunosorbent assay. The expression of interleukin-6 (IL-6) signaling molecules in colonic tissues were investigated using Western blotting and immunohistochemistry (IHC). RESULTS IA-0130 (0.1 mg/kg) and CsA (30 mg/kg) prevented colitis symptom, including weight loss, bleeding, colon shortening, and expression of pro-inflammatory cytokines in colon tissues. IA-0130 treatment regulated the mouse intestinal barrier permeability and inhibited abnormal TJ protein expression. IA-0130 down-regulated IL-6 expression and prevented the phosphorylation of signaling molecules in colonic tissues. CONCLUSIONS This study demonstrated that IA-0130 suppressed colitis progression by inhibiting the gp130 signaling pathway and expression of pro-inflammatory cytokines, and maintaining TJ integrity.
Collapse
Affiliation(s)
- Young-Jin Jeong
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences, BK21 FOUR Team for Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, NP512, Hall of Cardinal Jin-Suk Cheong, 43 Jibong-Ro, Bucheon-Si, Gyeonggi‑do, 14662, Republic of Korea
| | - Hae-Ri Lee
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences, BK21 FOUR Team for Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, NP512, Hall of Cardinal Jin-Suk Cheong, 43 Jibong-Ro, Bucheon-Si, Gyeonggi‑do, 14662, Republic of Korea
| | - Sun-Ae Park
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences, BK21 FOUR Team for Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, NP512, Hall of Cardinal Jin-Suk Cheong, 43 Jibong-Ro, Bucheon-Si, Gyeonggi‑do, 14662, Republic of Korea
| | - Joong-Woon Lee
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences, BK21 FOUR Team for Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, NP512, Hall of Cardinal Jin-Suk Cheong, 43 Jibong-Ro, Bucheon-Si, Gyeonggi‑do, 14662, Republic of Korea
| | - Lee Kyung Kim
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences, BK21 FOUR Team for Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, NP512, Hall of Cardinal Jin-Suk Cheong, 43 Jibong-Ro, Bucheon-Si, Gyeonggi‑do, 14662, Republic of Korea
| | - Hee Jung Kim
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences, BK21 FOUR Team for Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, NP512, Hall of Cardinal Jin-Suk Cheong, 43 Jibong-Ro, Bucheon-Si, Gyeonggi‑do, 14662, Republic of Korea
| | - Jae Hong Seo
- Laboratory of Pharmaceutical Manufacturing Chemistry, Integrated Research Institute of Pharmaceutical Sciences, College of Pharmacy, The Catholic University of Korea, 43 Jibong-Ro, Bucheon‑si, Gyeonggi‑do, 14662, Republic of Korea
| | - Tae-Hwe Heo
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences, BK21 FOUR Team for Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, NP512, Hall of Cardinal Jin-Suk Cheong, 43 Jibong-Ro, Bucheon-Si, Gyeonggi‑do, 14662, Republic of Korea.
| |
Collapse
|
19
|
Wang L, Nabi F, Zhang X, Zhou G, Shah QA, Li S, Lu Y, Mu S, Zhu X, Lin Z, Li J. Effects of Lactobacillus plantarum on Broiler Health: Integrated Microbial and Metabolomics Analysis. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10336-x. [PMID: 39090454 DOI: 10.1007/s12602-024-10336-x] [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] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
Given China's prohibition on the utilization of antibiotics as feed additives in 2020, we aim to investigate nutrition additives that are both efficient and safe. Lactobacillus, a well-recognized beneficial probiotic, has explicitly been investigated for its effects on health status of the host and overall impact on food industry. To evaluate effects of Lactobacillus plantarum (LW) supplementation on broiler chicken, we conducted comprehensive multi-omics analysis, growth performance evaluation, RT-qPCR analysis, and immunofluorescence. The findings revealed that LW supplementation resulted in a substantial progress in growth performance (approximately 205 g increase in final body weight in comparison to the control group (p < 0.01)). Additionally, LW exhibited promising potential for enhancing antioxidant properties of serum and promoting gut integrity and growth as evidenced by improved antioxidant indices (p < 0.01), intestinal villus morphology (p < 0.01), and enhanced gut barrier function (p < 0.01). Meanwhile, the multi-omics analysis, including 16S rRNA sequencing and liquid chromatography-tandem mass spectrometry, revealed an enrichment of beneficial microbes in the gut of broilers that were supplemented with LW, while simultaneously depleting harmful microorganisms. Moreover, a noteworthy modification was observed in gut metabolic profiling subsequent to the execution of the probiotic strategy. Specifically, variations were noticed in the levels of metabolites and metabolic pathways such as parathyroid hormone synthesis, inflammatory mediator regulation of TRP channels, oxidative phosphorylation, and mineral absorption. Taken together, our findings validate that LW administration produces valuable effects on the health and growth performance of broilers owing to its capability to boost the gut microbiota homeostasis and intestinal metabolism. Present findings signify the potential of LW as a dietary additive to promote growth and development in broiler chickens.
Collapse
Affiliation(s)
- Lei Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Fazul Nabi
- Faculty of Veterinary and Animal Sciences, Lasbela University of Agriculture, Water and Marine Sciences, Uthal, 90150, Pakistan
| | - Xiaohu Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Guangyu Zhou
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Qurban Ali Shah
- Faculty of Veterinary and Animal Sciences, Lasbela University of Agriculture, Water and Marine Sciences, Uthal, 90150, Pakistan
| | - Siyuan Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Yaozhong Lu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Siyang Mu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Xiaohui Zhu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Zhengrong Lin
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Jiakui Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
| |
Collapse
|
20
|
Compare D, Sgamato C, Rocco A, Coccoli P, Ambrosio C, Nardone G. The Leaky Gut and Human Diseases: "Can't Fill the Cup if You Don't Plug the Holes First". Dig Dis 2024; 42:548-566. [PMID: 39047703 DOI: 10.1159/000540379] [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: 03/25/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND The gut barrier is a sophisticated and dynamic system that forms the frontline defense between the external environment and the body's internal milieu and includes various structural and functional components engaged not only in digestion and nutrient absorption but also in immune regulation and overall health maintenance. SUMMARY When one or more components of the intestinal barrier lose their structure and escape their function, this may result in a leaky gut. Mounting evidence emphasizes the crucial role of the gut microbiome in preserving the integrity of the gut barrier and provides insights into the pathophysiological implications of conditions related to leaky gut in humans. Assessment of intestinal permeability has evolved from invasive techniques to noninvasive biomarkers, but challenges remain in achieving consensus about the best testing methods and their accuracy. Research on the modulation of gut permeability is just starting, and although no medical guidelines for the treatment of leaky gut syndrome are available, several treatment strategies are under investigation with promising results. KEY MESSAGES This review discusses the composition of the intestinal barrier, the pathophysiology of the leaky gut and its implications on human health, the measurement of intestinal permeability, and the therapeutic strategies to restore gut barrier integrity.
Collapse
Affiliation(s)
- Debora Compare
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| | - Costantino Sgamato
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| | - Alba Rocco
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| | - Pietro Coccoli
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| | - Carmen Ambrosio
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| | - Gerardo Nardone
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| |
Collapse
|
21
|
Rakotoarivelo V, Mayer TZ, Simard M, Flamand N, Di Marzo V. The Impact of the CB 2 Cannabinoid Receptor in Inflammatory Diseases: An Update. Molecules 2024; 29:3381. [PMID: 39064959 PMCID: PMC11279428 DOI: 10.3390/molecules29143381] [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: 06/18/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
The emergence of inflammatory diseases is a heavy burden on modern societies. Cannabis has been used for several millennia to treat inflammatory disorders such as rheumatism or gout. Since the characterization of cannabinoid receptors, CB1 and CB2, the potential of cannabinoid pharmacotherapy in inflammatory conditions has received great interest. Several studies have identified the importance of these receptors in immune cell migration and in the production of inflammatory mediators. As the presence of the CB2 receptor was documented to be more predominant in immune cells, several pharmacological agonists and antagonists have been designed to treat inflammation. To better define the potential of the CB2 receptor, three online databases, PubMed, Google Scholar and clinicaltrial.gov, were searched without language restriction. The full texts of articles presenting data on the endocannabinoid system, the CB2 receptor and its role in modulating inflammation in vitro, in animal models and in the context of clinical trials were reviewed. Finally, we discuss the clinical potential of the latest cannabinoid-based therapies in inflammatory diseases.
Collapse
Affiliation(s)
- Volatiana Rakotoarivelo
- Centre de Recherche de l’Institut Universitaire De Cardiologie Et De Pneumologie de Québec, Département of Médecine, Université Laval, Québec City, QC G1V 4G5, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Québec City, QC G1V 0V6, Canada
| | - Thomas Z. Mayer
- Centre de Recherche de l’Institut Universitaire De Cardiologie Et De Pneumologie de Québec, Département of Médecine, Université Laval, Québec City, QC G1V 4G5, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Québec City, QC G1V 0V6, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, and Centre NUTRISS, École de Nutrition, Université Laval, Québec City, QC G1V 0V6, Canada
| | - Mélissa Simard
- Centre de Recherche de l’Institut Universitaire De Cardiologie Et De Pneumologie de Québec, Département of Médecine, Université Laval, Québec City, QC G1V 4G5, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Québec City, QC G1V 0V6, Canada
| | - Nicolas Flamand
- Centre de Recherche de l’Institut Universitaire De Cardiologie Et De Pneumologie de Québec, Département of Médecine, Université Laval, Québec City, QC G1V 4G5, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Québec City, QC G1V 0V6, Canada
| | - Vincenzo Di Marzo
- Centre de Recherche de l’Institut Universitaire De Cardiologie Et De Pneumologie de Québec, Département of Médecine, Université Laval, Québec City, QC G1V 4G5, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Québec City, QC G1V 0V6, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, and Centre NUTRISS, École de Nutrition, Université Laval, Québec City, QC G1V 0V6, Canada
- Joint International Unit between the CNR of Italy and Université Laval on Chemical and Biomolecular Research on the Microbiome and Its Impact on Metabolic Health and Nutrition (UMI-MicroMeNu), Québec City, QC G1V 0V6, Canada
| |
Collapse
|
22
|
D’Ambrosio A, Altomare A, Boscarino T, Gori M, Balestrieri P, Putignani L, Del Chierico F, Carotti S, Cicala M, Guarino MPL, Piemonte V. Mathematical Modeling of Vedolizumab Treatment's Effect on Microbiota and Intestinal Permeability in Inflammatory Bowel Disease Patients. Bioengineering (Basel) 2024; 11:710. [PMID: 39061792 PMCID: PMC11274165 DOI: 10.3390/bioengineering11070710] [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: 06/01/2024] [Revised: 07/04/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Growing evidence suggests that impaired gut permeability and gut microbiota alterations are involved in the pathogenesis of Inflammatory Bowel Diseases (IBDs), which include Ulcerative Colitis (UC) and Crohn's Disease (CD). Vedolizumab is an anti-α4β7 antibody approved for IBD treatment, used as the first treatment or second-line therapy when the first line results in inadequate effectiveness. The aim of this study is to develop a mathematical model capable of describing the pathophysiological mechanisms of Vedolizumab treatment in IBD patients. In particular, the relationship between drug concentration in the blood, colonic mucosal permeability and fecal microbiota composition was investigated and modeled to detect and predict trends in order to support and tailor Vedolizumab therapies. To pursue this aim, clinical data from a pilot study on a cluster of 11 IBD patients were analyzed. Enrolled patients underwent colonoscopy in three phases (before (t0), after 24 weeks of (t1) and after 52 weeks of (t2 ) Vedolizumab treatment) to collect mucosal biopsies for transepithelial electrical resistance (TEER) evaluation (permeability to ions), intestinal permeability measurement and histological analysis. Moreover, fecal samples were collected for the intestinal microbiota analysis at the three time points. The collected data were compared to those of 11 healthy subjects at t0, who underwent colonoscopy for screening surveillance, and used to implement a three-compartmental mathematical model (comprising central blood, peripheral blood and the intestine). The latter extends previous evidence from the literature, based on the regression of experimental data, to link drug concentration in the peripheral blood compartment with Roseburia abundance and intestinal permeability. The clinical data showed that Vedolizumab treatment leads to an increase in TEER and a reduction in intestinal permeability to a paracellular probe, improving tissue inflammation status. Microbiota analysis showed increasing values of Roseburia, albeit not statistically significant. This trend was adequately reproduced by the mathematical model, which offers a useful tool to describe the pathophysiological effects of Vedolizumab therapy on colonic mucosal permeability and fecal microbiota composition. The model's satisfactory predictive capabilities and simplicity shed light on the relationship between the drug, the microbiota and permeability and allow for its straightforward extension to diverse therapeutic conditions.
Collapse
Affiliation(s)
- Antonio D’Ambrosio
- Unit of Chemical-Physics Fundamentals in Chemical Engineering, Department of Science and Technology for Sustainable Development and One Health, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy; (T.B.); (V.P.)
| | - Annamaria Altomare
- Department of Sciences and Technology of Sustainable Development and Human Health, Università Campus Biomedico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy;
- Gastroenterology Research Unit, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (M.G.); (M.C.); (M.P.L.G.)
| | - Tamara Boscarino
- Unit of Chemical-Physics Fundamentals in Chemical Engineering, Department of Science and Technology for Sustainable Development and One Health, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy; (T.B.); (V.P.)
| | - Manuele Gori
- Gastroenterology Research Unit, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (M.G.); (M.C.); (M.P.L.G.)
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), International Campus “A. Buzzati-Traverso”, Via E. Ramarini 32, Monterotondo Scalo, 00015 Rome, Italy
| | - Paola Balestrieri
- Gastroenterology Unit, Fondazione Policlinico Campus Bio-Medico di Roma, Via Alvaro del Portillo 200, 00128 Rome, Italy;
| | - Lorenza Putignani
- Units of Microbiomics and Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy;
| | - Federica Del Chierico
- Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy;
| | - Simone Carotti
- Microscopic and Ultrastructural Anatomy Research Unit, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy;
| | - Michele Cicala
- Gastroenterology Research Unit, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (M.G.); (M.C.); (M.P.L.G.)
- Microscopic and Ultrastructural Anatomy Research Unit, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy;
| | - Michele Pier Luca Guarino
- Gastroenterology Research Unit, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (M.G.); (M.C.); (M.P.L.G.)
- Gastroenterology Unit, Fondazione Policlinico Campus Bio-Medico di Roma, Via Alvaro del Portillo 200, 00128 Rome, Italy;
| | - Vincenzo Piemonte
- Unit of Chemical-Physics Fundamentals in Chemical Engineering, Department of Science and Technology for Sustainable Development and One Health, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy; (T.B.); (V.P.)
| |
Collapse
|
23
|
Wei Y, Zhao X, Xu T, Liu Z, Zuo Y, Zhang M, Zhang Y, Yin H. Soybean Bioactive Peptide Supplementation Affects the Intestinal Immune Antioxidant Function, Microbial Diversity, and Reproductive Organ Development in Roosters. Animals (Basel) 2024; 14:1954. [PMID: 38998068 PMCID: PMC11240439 DOI: 10.3390/ani14131954] [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: 06/06/2024] [Revised: 06/23/2024] [Accepted: 06/28/2024] [Indexed: 07/14/2024] Open
Abstract
Soybean is an important source of high-quality vegetable protein with various health-improving properties, and its main bioactive substances are small peptides produced by in vitro enzymatic hydrolytic processes. In traditional layer breeding, the nutritional health of roosters is frequently neglected, ultimately affecting the quality and quantity of offspring. This study investigated the effects of various quantities (0%, 0.15%, 0.30%, 0.45%, and 0.60%) of soybean bioactive peptide (SBP) feed additives on immunological and antioxidant functions, gut health, and reproductive performance of roosters. SBP supplementation significantly improved male growth and reproductive performance, including growth rate, feed conversion ratio, reproductive organ development, and semen quality. SBP also increased immune and antioxidant levels, boosted the integrity of the small intestinal physiological structure and barrier function, and diversity of cecal microbes, and decreased the apoptotic ratio of small intestinal epithelial cells. The effects of SBP on various functions of males showed a quadratic trend, with the optimal concentration determined to be 0.45%.
Collapse
Affiliation(s)
- Yimeng Wei
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiyu Zhao
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Tao Xu
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhenyan Liu
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yalan Zuo
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Mingxue Zhang
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yao Zhang
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Huadong Yin
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
24
|
Han J, Balasubramanian I, Flores JA, Bandyopadhyay S, Yang J, Liu Y, Singh R, Setty P, Kiela P, Ferraris R, Gao N. Intestinal lysozyme engagement of Salmonella Typhimurium stimulates the release of barrier-impairing InvE and Lpp1. J Biol Chem 2024; 300:107424. [PMID: 38823640 PMCID: PMC11255904 DOI: 10.1016/j.jbc.2024.107424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/06/2024] [Accepted: 05/17/2024] [Indexed: 06/03/2024] Open
Abstract
Lysozyme is a β-1,4-glycosidase that hydrolyzes the polysaccharide backbone of bacterial cell walls. With an additional bactericidal function mediated by a separate protein domain, lysozyme is considered a uniquely important antimicrobial molecule contributing to the host's innate immune response to infection. Elevated lysozyme production is found in various inflammatory conditions while patients with genetic risks for inflammatory bowel diseases demonstrate abnormal lysozyme expression, granule packaging, and secretion in Paneth cells. However, it remains unclear how a gain- or loss-of-function in host lysozyme may impact the host inflammatory responses to pathogenic infection. We challenged Lyz1-/- and ectopic Lyz1-expressing (Villin-Lyz1TG) mice with S. Typhimurium and then comprehensively assessed the inflammatory disease progression. We conducted proteomics analysis to identify molecules derived from human lysozyme-mediated processing of live Salmonella. We examined the barrier-impairing effects of these identified molecules in human intestinal epithelial cell monolayer and enteroids. Lyz1-/- mice are protected from infection in terms of morbidity, mortality, and barrier integrity, whereas Villin-Lyz1TG mice demonstrate exacerbated infection and inflammation. The growth and invasion of Salmonella in vitro are not affected by human or chicken lysozyme, whereas lysozyme encountering of live Salmonella stimulates the release of barrier-disrupting factors, InvE-sipC and Lpp1, which directly or indirectly impair the tight junctions. The direct engagement of host intestinal lysozyme with an enteric pathogen such as Salmonella promotes the release of virulence factors that are barrier-impairing and pro-inflammatory. Controlling lysozyme function may help alleviate the inflammatory progression.
Collapse
Affiliation(s)
- Jiangmeng Han
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | | | - Juan A Flores
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | | | - Jiaxing Yang
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Yue Liu
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Rajbir Singh
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Prashanth Setty
- Department of Pediatrics, Daniel Cracchiolo Institute for Pediatric Autoimmune Disease Research, Steele Children's Research Center, University of Arizona, Tucson, Arizona, USA
| | - Pawel Kiela
- Department of Pediatrics, Daniel Cracchiolo Institute for Pediatric Autoimmune Disease Research, Steele Children's Research Center, University of Arizona, Tucson, Arizona, USA
| | - Ronaldo Ferraris
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers Biomedical and Health Sciences, Newark, New Jersey, USA
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA; Department of Pharmacology, Physiology, and Neuroscience, Rutgers Biomedical and Health Sciences, Newark, New Jersey, USA.
| |
Collapse
|
25
|
Cicala M, Gori M, Balestrieri P, Altomare A, Tullio A, Di Cola S, Dejongh S, Graziani MG, Pagnini C, Carotti S, Perrone G, Ribolsi M, Fiorani M, Guarino MPL, Farré R. Colonic Epithelial Permeability to Ions Is Restored after Vedolizumab Treatment and May Predict Clinical Response in Inflammatory Bowel Disease Patients. Int J Mol Sci 2024; 25:5817. [PMID: 38892004 PMCID: PMC11172326 DOI: 10.3390/ijms25115817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Vedolizumab (VDZ) is used for treating inflammatory bowel disease (IBD) patients. A study investigating colonic epithelial barrier function ex vivo following VDZ is lacking. This work aims to evaluate ex vivo the colonic epithelial barrier function in IBD patients at baseline and during VDZ treatment, and to investigate the relationships between barrier function and clinical parameters. Colonic specimens were obtained from 23 IBD patients before, and at 24 and 52 weeks after VDZ treatment, and from 26 healthy volunteers (HV). Transepithelial electrical resistance (TEER, permeability to ions) and paracellular permeability were measured in Ussing chambers. IBD patients showed increased epithelial permeability to ions (TEER, 13.80 ± 1.04 Ω × cm2 vs. HV 20.70 ± 1.52 Ω × cm2, p < 0.001) without changes in paracellular permeability of a 4 kDa probe. VDZ increased TEER (18.09 ± 1.44 Ω × cm2, p < 0.001) after 52 weeks. A clinical response was observed in 58% and 25% of patients at week 24, and in 62% and 50% at week 52, in ulcerative colitis and Crohn's disease, respectively. Clinical and endoscopic scores were strongly associated with TEER. TEER < 14.65 Ω × cm2 predicted response to VDZ (OR 11; CI 2-59). VDZ reduces the increased permeability to ions observed in the colonic epithelium of IBD patients before treatment, in parallel to a clinical, histological (inflammatory infiltrate), and endoscopic improvement. A low TEER predicts clinical response to VDZ therapy.
Collapse
Affiliation(s)
- Michele Cicala
- Gastroenterology Unit, Fondazione Policlinico Universitario Campus Bio-Medico and Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (M.C.); (P.B.); (A.A.); (A.T.); (S.D.C.); (M.R.); (M.F.); (M.P.L.G.)
| | - Manuele Gori
- Gastroenterology Unit, Fondazione Policlinico Universitario Campus Bio-Medico and Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (M.C.); (P.B.); (A.A.); (A.T.); (S.D.C.); (M.R.); (M.F.); (M.P.L.G.)
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), International Campus “A. Buzzati-Traverso”, Via E. Ramarini 32, Monterotondo Scalo, 00015 Rome, Italy
| | - Paola Balestrieri
- Gastroenterology Unit, Fondazione Policlinico Universitario Campus Bio-Medico and Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (M.C.); (P.B.); (A.A.); (A.T.); (S.D.C.); (M.R.); (M.F.); (M.P.L.G.)
| | - Annamaria Altomare
- Gastroenterology Unit, Fondazione Policlinico Universitario Campus Bio-Medico and Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (M.C.); (P.B.); (A.A.); (A.T.); (S.D.C.); (M.R.); (M.F.); (M.P.L.G.)
| | - Alessandro Tullio
- Gastroenterology Unit, Fondazione Policlinico Universitario Campus Bio-Medico and Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (M.C.); (P.B.); (A.A.); (A.T.); (S.D.C.); (M.R.); (M.F.); (M.P.L.G.)
| | - Simone Di Cola
- Gastroenterology Unit, Fondazione Policlinico Universitario Campus Bio-Medico and Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (M.C.); (P.B.); (A.A.); (A.T.); (S.D.C.); (M.R.); (M.F.); (M.P.L.G.)
| | - Sander Dejongh
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, 3000 Leuven, Belgium; (S.D.); (R.F.)
- Laboratory of Nephrology and Renal Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Maria Giovanna Graziani
- Department of Gastroenterology and Digestive Endoscopy, S. Giovanni Addolorata Hospital, 00184 Rome, Italy; (M.G.G.); (C.P.)
| | - Cristiano Pagnini
- Department of Gastroenterology and Digestive Endoscopy, S. Giovanni Addolorata Hospital, 00184 Rome, Italy; (M.G.G.); (C.P.)
| | - Simone Carotti
- Microscopic and Ultrastructural Anatomy Research Unit Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy;
- Predictive Molecular Diagnostics, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Giuseppe Perrone
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy;
| | - Mentore Ribolsi
- Gastroenterology Unit, Fondazione Policlinico Universitario Campus Bio-Medico and Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (M.C.); (P.B.); (A.A.); (A.T.); (S.D.C.); (M.R.); (M.F.); (M.P.L.G.)
| | - Marcello Fiorani
- Gastroenterology Unit, Fondazione Policlinico Universitario Campus Bio-Medico and Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (M.C.); (P.B.); (A.A.); (A.T.); (S.D.C.); (M.R.); (M.F.); (M.P.L.G.)
| | - Michele P. L. Guarino
- Gastroenterology Unit, Fondazione Policlinico Universitario Campus Bio-Medico and Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (M.C.); (P.B.); (A.A.); (A.T.); (S.D.C.); (M.R.); (M.F.); (M.P.L.G.)
| | - Ricard Farré
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, 3000 Leuven, Belgium; (S.D.); (R.F.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28029 Madrid, Spain
| |
Collapse
|
26
|
Wang Y, Xiao J, Wei S, Su Y, Yang X, Su S, Lan L, Chen X, Huang T, Shan Q. Protective effect of zinc gluconate on intestinal mucosal barrier injury in antibiotics and LPS-induced mice. Front Microbiol 2024; 15:1407091. [PMID: 38855764 PMCID: PMC11157515 DOI: 10.3389/fmicb.2024.1407091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 05/07/2024] [Indexed: 06/11/2024] Open
Abstract
Objective The aim of the study is to investigate the function and mechanism of Zinc Gluconate (ZG) on intestinal mucosal barrier damage in antibiotics and Lipopolysaccharide (LPS)-induced mice. Methods We established a composite mouse model by inducing intestinal mucosal barrier damage using antibiotics and LPS. The animals were divided into five groups: Control (normal and model) and experimental (low, medium, and high-dose ZG treatments). We evaluated the intestinal mucosal barrier using various methods, including monitoring body weight and fecal changes, assessing pathological damage and ultrastructure of the mouse ileum, analyzing expression levels of tight junction (TJ)-related proteins and genes, confirming the TLR4/NF-κB signaling pathway, and examining the structure of the intestinal flora. Results In mice, the dual induction of antibiotics and LPS led to weight loss, fecal abnormalities, disruption of ileocecal mucosal structure, increased intestinal barrier permeability, and disorganization of the microbiota structure. ZG restored body weight, alleviated diarrheal symptoms and pathological damage, and maintained the structural integrity of intestinal epithelial cells (IECs). Additionally, ZG reduced intestinal mucosal permeability by upregulating TJ-associated proteins (ZO-1, Occludin, Claudin-1, and JAM-A) and downregulating MLCK, thereby repairing intestinal mucosal barrier damage induced by dual induction of antibiotics and LPS. Moreover, ZG suppressed the TLR4/NF-κB signaling pathway, demonstrating anti-inflammatory properties and preserving barrier integrity. Furthermore, ZG restored gut microbiota diversity and richness, evidenced by increased Shannon and Observed features indices, and decreased Simpson's index. ZG also modulated the relative abundance of beneficial human gut bacteria (Bacteroidetes, Firmicutes, Verrucomicrobia, Parabacteroides, Lactobacillus, and Akkermansia) and harmful bacteria (Proteobacteria and Enterobacter), repairing the damage induced by dual administration of antibiotics and LPS. Conclusion ZG attenuates the dual induction of antibiotics and LPS-induced intestinal barrier damage and also protects the intestinal barrier function in mice.
Collapse
Affiliation(s)
- Yongcai Wang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Dazhou Central Hospital, Dazhou, China
| | - Juan Xiao
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Sumei Wei
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ying Su
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xia Yang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shiqi Su
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Liancheng Lan
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiuqi Chen
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ting Huang
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Qingwen Shan
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
27
|
Tharabenjasin P, Moonwiriyakit A, Sontikun J, Timpratueang K, Kuno S, Aiebchun T, Jongkon N, Mongkolrob R, Pabalan N, Choowongkomon K, Muanprasat C. The barrier-protective effect of β-eudesmol against type 2-inflammatory cytokine-induced tight junction disassembly in airway epithelial cells. PLoS One 2024; 19:e0302851. [PMID: 38687777 PMCID: PMC11060601 DOI: 10.1371/journal.pone.0302851] [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: 09/17/2023] [Accepted: 04/14/2024] [Indexed: 05/02/2024] Open
Abstract
Allergic inflammation, which is the pathogenesis of allergic rhinitis and asthma, is associated with disruption of the airway epithelial barrier due to the effects of type 2 inflammatory cytokines, i.e. interleukin-4 and interleukin-13 (IL-4/13). The anti-allergic inflammatory effect of β-eudesmol (BE) on the tight junction (TJ) of the airway epithelium has not previously been reported. Herein, the barrier protective effect of BE was determined by measurement of transepithelial electrical resistance and by paracellular permeability assay in an IL-4/13-treated 16HBE14o- monolayer. Pre-treatment of BE concentration- and time- dependently inhibited IL-4/13-induced TJ barrier disruption, with the most significant effect observed at 20 μM. Cytotoxicity analyses showed that BE, either alone or in combination with IL-4/13, had no effect on cell viability. Western blot and immunofluorescence analyses showed that BE inhibited IL-4/13-induced mislocalization of TJ components, including occludin and zonula occludens-1 (ZO-1), without affecting the expression of these two proteins. In addition, the mechanism of the TJ-protective effect of BE was mediated by inhibition of IL-4/13-induced STAT6 phosphorylation, in which BE might serve as an antagonist of cytokine receptors. In silico molecular docking analysis demonstrated that BE potentially interacted with the site I pocket of the type 2 IL-4 receptor, likely at Asn-126 and Tyr-127 amino acid residues. It can therefore be concluded that BE is able to prevent IL-4/13-induced TJ disassembly by interfering with cytokine-receptor interaction, leading to suppression of STAT6-induced mislocalization of occludin and ZO-1. BE is a promising candidate for a therapeutic intervention for inflammatory airway epithelial disorders driven by IL-4/13.
Collapse
Affiliation(s)
- Phuntila Tharabenjasin
- Chulabhorn International College of Medicine, Thammasat University (Rangsit Campus), Klongluang, Pathumthani, Thailand
| | - Aekkacha Moonwiriyakit
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bang Phli, Samut Prakan, Thailand
| | - Jenjira Sontikun
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bang Phli, Samut Prakan, Thailand
| | - Kanokphorn Timpratueang
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bang Phli, Samut Prakan, Thailand
| | - Suhaibee Kuno
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bang Phli, Samut Prakan, Thailand
| | - Thitinan Aiebchun
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Nathjanan Jongkon
- Department of Social and Applied Science, College of Industrial Technology, King Mongkut’s University of Technology North Bangkok, Bangkok, Thailand
| | - Rungrawee Mongkolrob
- Chulabhorn International College of Medicine, Thammasat University (Rangsit Campus), Klongluang, Pathumthani, Thailand
| | - Noel Pabalan
- Chulabhorn International College of Medicine, Thammasat University (Rangsit Campus), Klongluang, Pathumthani, Thailand
| | | | - Chatchai Muanprasat
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bang Phli, Samut Prakan, Thailand
| |
Collapse
|
28
|
Luu LDW, Pandey A, Paramsothy S, Ngo C, Castaño-Rodríguez N, Liu C, Kamm MA, Borody TJ, Man SM, Kaakoush NO. Profiling the colonic mucosal response to fecal microbiota transplantation identifies a role for GBP5 in colitis in humans and mice. Nat Commun 2024; 15:2645. [PMID: 38531874 DOI: 10.1038/s41467-024-46983-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 03/15/2024] [Indexed: 03/28/2024] Open
Abstract
Host molecular responses to fecal microbiota transplantation (FMT) in ulcerative colitis are not well understood. Here, we profile the human colonic mucosal transcriptome prior to and following FMT or placebo to identify molecules regulated during disease remission. FMT alters the transcriptome above the effect of placebo (n = 75 vs 3 genes, q < 0.05), including modulation of structural, metabolic and inflammatory pathways. This response is attributed to responders with no consistency observed in non-responders. Regulated pathways in responders include tight junctions, calcium signalling and xenobiotic metabolism. Genes significantly regulated longitudinally in responders post-FMT could discriminate them from responders and non-responders at baseline and non-responders post-FMT, with GBP5 and IRF4 downregulation being associated with remission. Female mice with a deletion of GBP5 are more resistant to developing colitis than their wild-type littermates, showing higher colonic IRF4 phosphorylation. The colonic mucosal response discriminates UC remission following FMT, with GBP5 playing a detrimental role in colitis.
Collapse
Affiliation(s)
- Laurence D W Luu
- School of Biomedical Sciences, UNSW, Sydney, NSW, 2052, Australia
- School of Biotechnology and Biomolecular Sciences, UNSW, Sydney, NSW, 2052, Australia
| | - Abhimanu Pandey
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, 2601, Australia
| | - Sudarshan Paramsothy
- Concord Clinical School, The University of Sydney, Sydney, NSW, 2139, Australia
- Department of Gastroenterology, Concord Repatriation General Hospital, Sydney, NSW, 2139, Australia
| | - Chinh Ngo
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, 2601, Australia
| | | | - Cheng Liu
- Conjoint Gastroenterology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, QLD, Australia
- School of Medicine, University of Queensland, Herston, QLD, 4006, Australia
- Mater Pathology, Mater Hospital Brisbane, South Brisbane, QLD, 4101, Australia
| | - Michael A Kamm
- Department of Gastroenterology, St Vincent's Hospital, Melbourne, VIC, 3065, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC, 3010, Australia
| | | | - Si Ming Man
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, 2601, Australia
| | | |
Collapse
|
29
|
Segui-Perez C, Stapels DAC, Ma Z, Su J, Passchier E, Westendorp B, Wubbolts RW, Wu W, van Putten JPM, Strijbis K. MUC13 negatively regulates tight junction proteins and intestinal epithelial barrier integrity via protein kinase C. J Cell Sci 2024; 137:jcs261468. [PMID: 38345099 PMCID: PMC10984281 DOI: 10.1242/jcs.261468] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/29/2024] [Indexed: 03/14/2024] Open
Abstract
Glycosylated mucin proteins contribute to the essential barrier function of the intestinal epithelium. The transmembrane mucin MUC13 is an abundant intestinal glycoprotein with important functions for mucosal maintenance that are not yet completely understood. We demonstrate that in human intestinal epithelial monolayers, MUC13 localized to both the apical surface and the tight junction (TJ) region on the lateral membrane. MUC13 deletion resulted in increased transepithelial resistance (TEER) and reduced translocation of small solutes. TEER buildup in ΔMUC13 cells could be prevented by addition of MLCK, ROCK or protein kinase C (PKC) inhibitors. The levels of TJ proteins including claudins and occludin were highly increased in membrane fractions of MUC13 knockout cells. Removal of the MUC13 cytoplasmic tail (CT) also altered TJ composition but did not affect TEER. The increased buildup of TJ complexes in ΔMUC13 and MUC13-ΔCT cells was dependent on PKC. The responsible PKC member might be PKCδ (or PRKCD) based on elevated protein levels in the absence of full-length MUC13. Our results demonstrate for the first time that a mucin protein can negatively regulate TJ function and stimulate intestinal barrier permeability.
Collapse
Affiliation(s)
- Celia Segui-Perez
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Daphne A. C. Stapels
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Ziliang Ma
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
- Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research (A*STAR), 138648 Singapore, Singapore
- Department of Pharmacy, National University of Singapore, 117543 Singapore, Singapore
| | - Jinyi Su
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Elsemieke Passchier
- UMAB, Department of Laboratory Pharmacy and Biomedical Genetics, Center for Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Bart Westendorp
- Department of Biomolecular Health Sciences, Division of Cell Biology, Metabolism and Cancer, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Richard W. Wubbolts
- Department of Biomolecular Health Sciences, Division of Cell Biology, Metabolism and Cancer, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Wei Wu
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
- Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research (A*STAR), 138648 Singapore, Singapore
- Department of Pharmacy, National University of Singapore, 117543 Singapore, Singapore
| | - Jos P. M. van Putten
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Karin Strijbis
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| |
Collapse
|
30
|
Stepanova M, Aherne CM. Adenosine in Intestinal Epithelial Barrier Function. Cells 2024; 13:381. [PMID: 38474346 PMCID: PMC10930693 DOI: 10.3390/cells13050381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/13/2024] [Accepted: 02/18/2024] [Indexed: 03/14/2024] Open
Abstract
At the intestinal front, several lines of defense are in place to resist infection and injury, the mucus layer, gut microbiome and strong epithelial junctions, to name a few. Their collaboration creates a resilient barrier. In intestinal disorders, such as inflammatory bowel disease (IBD), barrier function is compromised, which results in rampant inflammation and tissue injury. In response to the destruction, the intestinal epithelium releases adenosine, a small but powerful nucleoside that functions as an alarm signal. Amidst the chaos of inflammation, adenosine aims to restore order. Within the scope of its effects is the ability to regulate intestinal epithelial barrier integrity. This review aims to define the contributions of adenosine to mucus production, microbiome-dependent barrier protection, tight junction dynamics, chloride secretion and acid-base balance to reinforce its importance in the intestinal epithelial barrier.
Collapse
Affiliation(s)
- Mariya Stepanova
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland;
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Carol M. Aherne
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland;
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
31
|
Hartley VL, Qaqish AM, Wood MJ, Studnicka BT, Iwai K, Liu TC, MacDuff DA. HOIL1 Regulates Group 3 Innate Lymphoid Cells in the Colon and Protects against Systemic Dissemination, Colonic Ulceration, and Lethality from Citrobacter rodentium Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1823-1834. [PMID: 37902285 PMCID: PMC10841105 DOI: 10.4049/jimmunol.2300351] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/19/2023] [Indexed: 10/31/2023]
Abstract
Heme-oxidized IRP2 ubiquitin ligase-1 (HOIL1)-deficient patients experience chronic intestinal inflammation and diarrhea as well as increased susceptibility to bacterial infections. HOIL1 is a component of the linear ubiquitin chain assembly complex that regulates immune signaling pathways, including NF-κB-activating pathways. We have shown previously that HOIL1 is essential for survival following Citrobacter rodentium gastrointestinal infection of mice, but the mechanism of protection by HOIL1 was not examined. C. rodentium is an important murine model for human attaching and effacing pathogens, enteropathogenic and enterohemorrhagic Escherichia coli that cause diarrhea and foodborne illnesses and lead to severe disease in children and immunocompromised individuals. In this study, we found that C. rodentium infection resulted in severe colitis and dissemination of C. rodentium to systemic organs in HOIL1-deficient mice. HOIL1 was important in the innate immune response to limit early replication and dissemination of C. rodentium. Using bone marrow chimeras and cell type-specific knockout mice, we found that HOIL1 functioned in radiation-resistant cells and partly in radiation-sensitive cells and in myeloid cells to limit disease, but it was dispensable in intestinal epithelial cells. HOIL1 deficiency significantly impaired the expansion of group 3 innate lymphoid cells and their production of IL-22 during C. rodentium infection. Understanding the role HOIL1 plays in type 3 inflammation and in limiting the pathogenesis of attaching and effacing lesion-forming bacteria will provide further insight into the innate immune response to gastrointestinal pathogens and inflammatory disorders.
Collapse
Affiliation(s)
- Victoria L. Hartley
- Department of Microbiology and Immunology, University of Illinois Chicago College of Medicine, Chicago, Illinois, USA
| | - Arwa M. Qaqish
- Department of Microbiology and Immunology, University of Illinois Chicago College of Medicine, Chicago, Illinois, USA
| | - Matthew J. Wood
- Department of Microbiology and Immunology, University of Illinois Chicago College of Medicine, Chicago, Illinois, USA
| | - Brian T. Studnicka
- Department of Microbiology and Immunology, University of Illinois Chicago College of Medicine, Chicago, Illinois, USA
| | - Kazuhiro Iwai
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ta-Chiang Liu
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Donna A. MacDuff
- Department of Microbiology and Immunology, University of Illinois Chicago College of Medicine, Chicago, Illinois, USA
| |
Collapse
|
32
|
Chernyavskij DA, Galkin II, Pavlyuchenkova AN, Fedorov AV, Chelombitko MA. Role of Mitochondria in Intestinal Epithelial Barrier Dysfunction in Inflammatory Bowel Disease. Mol Biol 2023; 57:1024-1037. [DOI: 10.1134/s0026893323060043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/26/2023] [Accepted: 05/29/2023] [Indexed: 01/05/2025]
Abstract
Abstract
Inflammatory bowel disease (IBD) is widespread in industrial countries with every 20th citizen being affected. Dysregulation of the epithelial barrier function is considered to play a key role in IBD. Permeability of the intestinal epithelium depends mostly on its self-renewal potential and the condition of intercellular junctions. Mitochondria are involved in regulating various intracellular processes in addition to their energy function. Recent data implicate mitochondria in intestinal epithelial barrier regulation and IBD. Mitochondrial dysfunction is possibly one of the factors that underlie the structural abnormalities of tight junctions and the cytoskeleton in intestinal epithelial cells and decrease the self-renewal capacity of the epithelium. The barrier function of the intestinal epithelium is consequently distorted, and IBD develops. The mechanisms of these processes are still unclear and require further research.
Collapse
|
33
|
Dunleavy KA, Raffals LE, Camilleri M. Intestinal Barrier Dysfunction in Inflammatory Bowel Disease: Underpinning Pathogenesis and Therapeutics. Dig Dis Sci 2023; 68:4306-4320. [PMID: 37773554 PMCID: PMC10798146 DOI: 10.1007/s10620-023-08122-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 09/19/2023] [Indexed: 10/01/2023]
Abstract
The intestinal barrier is composed of several essential elements including luminal enzymes, bile acids, water layer, epithelial layer, and enterocyte layer. It acts as a dynamic interface between the luminal contents of food, commensal and pathogenic bacteria, and the gastrointestinal tract. The role of barrier dysfunction is of significant research interest in the development and targeted treatment of chronic inflammatory gastrointestinal conditions, such as inflammatory bowel disease. This review aims to examine the role of intestinal barrier dysfunction in the development of inflammatory bowel disease, the pathophysiology of increased barrier permeability in inflammatory bowel disease, and to explore potential treatment targets and clinical applications.
Collapse
Affiliation(s)
- Katie A Dunleavy
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First St. S.W., Rochester, MN, 55905, USA
| | - Laura E Raffals
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First St. S.W., Rochester, MN, 55905, USA.
| | - Michael Camilleri
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First St. S.W., Rochester, MN, 55905, USA
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER), Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
34
|
Liu RD, Meng XY, Li CL, Lin XZ, Xu QY, Xu H, Long SR, Cui J, Wang ZQ. Trichinella spiralis cathepsin L damages the tight junctions of intestinal epithelial cells and mediates larval invasion. PLoS Negl Trop Dis 2023; 17:e0011816. [PMID: 38048314 PMCID: PMC10721182 DOI: 10.1371/journal.pntd.0011816] [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: 06/05/2023] [Revised: 12/14/2023] [Accepted: 11/22/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND Cathepsin L, a lysosomal enzyme, participates in diverse physiological processes. Recombinant Trichinella spiralis cathepsin L domains (rTsCatL2) exhibited natural cysteine protease activity and hydrolyzed host immunoglobulin and extracellular matrix proteins in vitro, but its functions in larval invasion are unknown. The aim of this study was to explore its functions in T. spiralis invasion of the host's intestinal epithelial cells. METHODOLOGY/PRINCIPAL FINDINGS RNAi significantly suppressed the expression of TsCatL mRNA and protein with TsCatL specific siRNA-302. T. spiralis larval invasion of Caco-2 cells was reduced by 39.87% and 38.36%, respectively, when anti-TsCatL2 serum and siRNA-302 were used. Mice challenged with siRNA-302-treated muscle larvae (ML) exhibited a substantial reduction in intestinal infective larvae, adult worm, and ML burden compared to the PBS group, with reductions of 44.37%, 47.57%, and 57.06%, respectively. The development and fecundity of the females from the mice infected with siRNA-302-treated ML was significantly inhibited. After incubation of rTsCatL2 with Caco-2 cells, immunofluorescence test showed that the rTsCatL2 gradually entered into the cells, altered the localization of cellular tight junction proteins (claudin 1, occludin and zo-1), adhesion junction protein (e-cadherin) and extracellular matrix protein (laminin), and intercellular junctions were lost. Western blot showed a 58.65% reduction in claudin 1 expression in Caco-2 cells treated with rTsCatL2. Co-IP showed that rTsCatL2 interacted with laminin and collagen I but not with claudin 1, e-cadherin, occludin and fibronectin in Caco-2 cells. Moreover, rTsCatL2 disrupted the intestinal epithelial barrier by inducing cellular autophagy. CONCLUSIONS rTsCatL2 disrupts the intestinal epithelial barrier and facilitates T. spiralis larval invasion.
Collapse
Affiliation(s)
- Ruo Dan Liu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, PR China
| | - Xiang Yu Meng
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, PR China
| | - Chen Le Li
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, PR China
| | - Xin Zhi Lin
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, PR China
| | - Qiu Yi Xu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, PR China
| | - Han Xu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, PR China
| | - Shao Rong Long
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, PR China
| | - Jing Cui
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, PR China
| | - Zhong Quan Wang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, PR China
| |
Collapse
|
35
|
Schneemann M, Heils L, Moos V, Weiß F, Krug SM, Weiner J, Beule D, Gerhard R, Schulzke JD, Bücker R. A Colonic Organoid Model Challenged with the Large Toxins of Clostridioides difficile TcdA and TcdB Exhibit Deregulated Tight Junction Proteins. Toxins (Basel) 2023; 15:643. [PMID: 37999506 PMCID: PMC10674794 DOI: 10.3390/toxins15110643] [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: 10/05/2023] [Revised: 10/25/2023] [Accepted: 11/01/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Clostridioides difficile toxins TcdA and TcdB are responsible for diarrhea and colitis. Lack of functional studies in organoid models of the gut prompted us to elucidate the toxin's effects on epithelial barrier function and the molecular mechanisms for diarrhea and inflammation. METHODS Human adult colon organoids were cultured on membrane inserts. Tight junction (TJ) proteins and actin cytoskeleton were analyzed for expression via Western blotting and via confocal laser-scanning microscopy for subcellular localization. RESULTS Polarized intestinal organoid monolayers were established from stem cell-containing colon organoids to apply toxins from the apical side and to perform functional measurements in the organoid model. The toxins caused a reduction in transepithelial electrical resistance in human colonic organoid monolayers with sublethal concentrations. Concomitantly, we detected increased paracellular permeability fluorescein and FITC-dextran-4000. Human colonic organoid monolayers exposed to the toxins exhibited redistribution of barrier-forming TJ proteins claudin-1, -4 and tricellulin, whereas channel-forming claudin-2 expression was increased. Perijunctional F-actin cytoskeleton organization was affected. CONCLUSIONS Adult stem cell-derived human colonic organoid monolayers were applicable as a colon infection model for electrophysiological measurements. The TJ changes noted can explain the epithelial barrier dysfunction and diarrhea in patients, as well as increased entry of luminal antigens triggering inflammation.
Collapse
Affiliation(s)
- Martina Schneemann
- Clinical Physiology, Nutritional Medicine, Charité—Universitätsmedizin Berlin, Campus Benjamin Franklin, 12203 Berlin, Germany
| | - Lucas Heils
- Clinical Physiology, Nutritional Medicine, Charité—Universitätsmedizin Berlin, Campus Benjamin Franklin, 12203 Berlin, Germany
| | - Verena Moos
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité—Universitätsmedizin Berlin, Campus Benjamin Franklin, 12203 Berlin, Germany
| | - Franziska Weiß
- Clinical Physiology, Nutritional Medicine, Charité—Universitätsmedizin Berlin, Campus Benjamin Franklin, 12203 Berlin, Germany
| | - Susanne M. Krug
- Clinical Physiology, Nutritional Medicine, Charité—Universitätsmedizin Berlin, Campus Benjamin Franklin, 12203 Berlin, Germany
| | - January Weiner
- Core Unit Bioinformatics (CUBI), Berlin Institute of Health at Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Dieter Beule
- Core Unit Bioinformatics (CUBI), Berlin Institute of Health at Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Ralf Gerhard
- Institute of Toxicology, Hannover Medical School, 30625 Hannover, Germany
| | - Jörg-Dieter Schulzke
- Clinical Physiology, Nutritional Medicine, Charité—Universitätsmedizin Berlin, Campus Benjamin Franklin, 12203 Berlin, Germany
| | - Roland Bücker
- Clinical Physiology, Nutritional Medicine, Charité—Universitätsmedizin Berlin, Campus Benjamin Franklin, 12203 Berlin, Germany
| |
Collapse
|
36
|
Sienkiewicz M, Sroka K, Binienda A, Jurk D, Fichna J. A new face of old cells: An overview about the role of senescence and telomeres in inflammatory bowel diseases. Ageing Res Rev 2023; 91:102083. [PMID: 37802318 DOI: 10.1016/j.arr.2023.102083] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/01/2023] [Accepted: 10/02/2023] [Indexed: 10/08/2023]
Abstract
Cellular senescence is a pivotal factor contributing to aging and the pathophysiology of age-related diseases. Despite the presence of inflammation and abnormal immune system function in both inflammatory bowel diseases (IBD) and senescence, the relationship between the two remains largely unexplored. Therefore, our study aimed to investigate the intricate connection between cellular senescence, telomeres, and IBD. The review highlights the presence of senescence markers, particularly p16 and p21, in IBD patients, suggesting their potential association with disease progression and mucosal inflammation. We emphasize the critical role of macrophages in eliminating senescent cells and how disturbance in effective clearance may contribute to persistent senescence and inflammation in IBD. Additionally, we shed light on the involvement of telomeres in IBD, as their dysfunction impairs enterocyte function and disrupts colonic barrier integrity, potentially exacerbating the pathogenesis of the disease. Targeting senescence and telomere dysfunctions holds promise for the development of innovative therapeutic approaches to mitigate intestinal inflammation and alleviate symptoms in IBD patients. By unraveling the precise role of senescence in IBD, we can pave the way for the discovery of novel therapeutic interventions that effectively address the underlying mechanisms of intestinal inflammation, offering hope for improved management and treatment of IBD patients.
Collapse
Affiliation(s)
- Michał Sienkiewicz
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Kamila Sroka
- Department of Family Medicine and Public Health, University of Opole, Opole, Poland
| | - Agata Binienda
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Diana Jurk
- Robert and Arlene Kogod Center On Aging, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
37
|
Rath T, Atreya R, Bodenschatz J, Uter W, Geppert CI, Vitali F, Zundler S, Waldner MJ, Hartmann A, Neurath MF. Healing of the epithelial barrier in the ileum is superior to endoscopic and histologic remission for predicting major adverse outcomes in ulcerative colitis. Front Med (Lausanne) 2023; 10:1221449. [PMID: 37881628 PMCID: PMC10595008 DOI: 10.3389/fmed.2023.1221449] [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: 05/12/2023] [Accepted: 08/09/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND Achieving endoscopic remission is a key therapeutic goal in patients with ulcerative colitis (UC) that is associated with favorable long-term disease outcomes. Here, we prospectively compared the predictive value of endoscopic and/or histologic remission against ileal barrier healing for predicting long-term disease behavior in a large cohort of UC patients in clinical remission. METHODS At baseline, UC patients in clinical remission underwent ileocolonoscopy with assessment of ileal barrier function by confocal endomicroscopy. Endoscopic and histologic disease activity and ileal barrier healing were scored using validated scores. During subsequent follow-up (FU), patients were closely monitored for clinical disease activity and occurrence of major adverse outcomes (MAO) defined as the following: disease relapse; UC-related hospitalization; UC-related surgery; necessity for initiation or dose escalation of systemic steroids, immunosuppressants, small molecules or biological therapy. RESULTS Of the 73 UC patients included, 67% experienced MAO during a mean FU of 25 months. The probability of MAO-free survival was significantly higher in UC patients with endoscopic and/or histologic remission compared to patients with endoscopically and/or histologically active disease. Ileal barrier healing on endomicroscopy was highly accurate for predicting the further course of UC and outcompeted endoscopic and histologic remission for predicting MAO-free survival. CONCLUSION Ileal barrier healing in clinically remittent UC patients can accurately predict future MAO development and is superior in its predictive capabilities than endoscopic and histologic remission. Ileal barrier healing therefore represents a novel and superior surrogate parameter for stratification of UC patients according to their risk for development of complicated disease behavior. CLINICAL TRIAL REGISTRATION https://classic.clinicaltrials.gov/ct2/show/NCT05157750, identifier NCT05157750.
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
| | - Julia Bodenschatz
- Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | - Wolfgang Uter
- Institute for Medical Informatics, Biometry and Epidemiology, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | - Carol I. Geppert
- Institute for Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | - Francesco Vitali
- Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | - Sebastian Zundler
- Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | - Maximilian J. Waldner
- Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | - Arndt Hartmann
- Institute for Pathology, 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
|
38
|
Kröhn L, Azabdaftari A, Heuberger J, Hudert C, Zilbauer M, Breiderhoff T, Bufler P. Modulation of intestinal IL-37 expression and its impact on the epithelial innate immune response and barrier integrity. Front Immunol 2023; 14:1261666. [PMID: 37799712 PMCID: PMC10548260 DOI: 10.3389/fimmu.2023.1261666] [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: 07/19/2023] [Accepted: 08/31/2023] [Indexed: 10/07/2023] Open
Abstract
Background and Aims Intestinal epithelial cells separate the luminal flora from lamina propria immune cells and regulate innate immune responses in the gut. An imbalance of the mucosal immune response and disrupted intestinal barrier integrity contribute to the evolution of inflammatory bowel diseases. Interleukin (IL)-37 has broad anti- inflammatory activity and is expressed by the human intestinal epithelium. Mice ectopically expressing human IL-37 show reduced epithelial damage and inflammation after DSS-induced colitis. Here, we investigated the impact of IL-37 on the innate immune response and tight junction protein expression of mouse intestinal organoids and the modulation of IL37 expression in human intestinal organoids. Methods Murine intestinal organoids were generated from IL-37tg and wildtype mice. Human ileal organoids were generated from healthy young donors. Results Expression of transgene IL-37 or recombinant IL-37 protein did not significantly reduce overall proinflammatory cytokine mRNA expression in murine intestinal organoids. However, higher IL37 expression correlated with a reduced proinflammatory cytokine response in murine colonic organoids. IL37 mRNA expression in human ileal organoids was modulated by proinflammatory cytokines showing an increased expression upon TNF-α-stimulation and decreased expression upon IFN-gamma stimulation. Transgene IL-37 expression did not rescue TNF-α-induced changes in morphology as well as ZO-1, occludin, claudin-2, and E-cadherin expression patterns of murine jejunal organoids. Conclusions We speculate that the anti-inflammatory activity of IL-37 in the intestine is mainly mediated by lamina propria immune cells protecting intestinal epithelial integrity.
Collapse
Affiliation(s)
- Laura Kröhn
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Aline Azabdaftari
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Julian Heuberger
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Hudert
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Matthias Zilbauer
- Wellcome Trust–Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Tilman Breiderhoff
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Philip Bufler
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
39
|
Wang YM, Abdullah S, Luebbering N, Langenberg L, Duell A, Lake K, Lane A, Hils B, Vazquez Silva O, Trapp M, Nalapareddy K, Koo J, Denson LA, Jodele S, Haslam DB, Faubion WA, Davies SM, Khandelwal P. Intestinal permeability in patients undergoing stem cell transplantation correlates with systemic acute phase responses and dysbiosis. Blood Adv 2023; 7:5137-5151. [PMID: 37083597 PMCID: PMC10480541 DOI: 10.1182/bloodadvances.2023009960] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/05/2023] [Accepted: 04/05/2023] [Indexed: 04/22/2023] Open
Abstract
Intestinal permeability may correlate with adverse outcomes during hematopoietic stem cell transplantation (HSCT), but longitudinal quantification with traditional oral mannitol and lactulose is not feasible in HSCT recipients because of mucositis and diarrhea. A modified lactulose:rhamnose (LR) assay is validated in children with environmental enteritis. Our study objective was to quantify peri-HSCT intestinal permeability changes using the modified LR assay. The LR assay was administered before transplant, at day +7 and +30 to 80 pediatric and young adult patients who received allogeneic HSCT. Lactulose and rhamnose were detected using urine mass spectrometry and expressed as an L:R ratio. Metagenomic shotgun sequencing of stool for microbiome analyses and enzyme-linked immunosorbent assay analyses of plasma lipopolysaccharide binding protein (LBP), ST2, REG3α, claudin1, occludin, and intestinal alkaline phosphatase were performed at the same timepoints. L:R ratios were increased at day +7 but returned to baseline at day +30 in most patients (P = .014). Conditioning regimen intensity did not affect the trajectory of L:R (P = .39). Baseline L:R ratios did not vary with diagnosis. L:R correlated with LBP levels (r2 = 0.208; P = .0014). High L:R ratios were associated with lower microbiome diversity (P = .035), loss of anaerobic organisms (P = .020), and higher plasma LBP (P = .0014). No adverse gastrointestinal effects occurred because of LR. Intestinal permeability as measured through L:R ratios after allogeneic HSCT correlates with intestinal dysbiosis and elevated plasma LBP. The LR assay is well-tolerated and may identify transplant recipients who are more likely to experience adverse outcomes.
Collapse
Affiliation(s)
- YunZu Michele Wang
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine, Cincinnati, OH
| | - Sheyar Abdullah
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Nathan Luebbering
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Lucille Langenberg
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Alexandra Duell
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Kelly Lake
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Adam Lane
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine, Cincinnati, OH
| | - Brian Hils
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Ormarie Vazquez Silva
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Monica Trapp
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Kodandaramireddy Nalapareddy
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Jane Koo
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine, Cincinnati, OH
| | - Lee A. Denson
- University of Cincinnati College of Medicine, Cincinnati, OH
- Department of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Sonata Jodele
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine, Cincinnati, OH
| | - David B. Haslam
- University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | | | - Stella M. Davies
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine, Cincinnati, OH
| | - Pooja Khandelwal
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
40
|
Fan YM, Wei YY, Wang HR, Yu-Ga, Zhang YN, Hao Z. Inhibitory effect of Portulaca oleracea L. aqueous extract and juice on NLRP3 inflammasome activation in an ulcerative colitis mouse model. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:86380-86394. [PMID: 37402916 DOI: 10.1007/s11356-023-28365-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 06/17/2023] [Indexed: 07/06/2023]
Abstract
Portulaca oleracea L. (PO) is an edible and medicinal plant used for treating gastrointestinal diseases. However, the effects of PO on ulcerative colitis (UC) and underlying mechanisms remain unclear. This study investigated the effects of PO aqueous extract (POE) and PO juice (PJ) on dextran sulfate sodium (DSS)-induced UC in a mouse model and attempted to unravel their underlying mechanisms. The results revealed that PJ contains more bioactive compounds and has more overlapping targets with UC than POE. Both POE and PJ effectively reduced Disease Activity Index scores and inflammatory cell infiltration in the UC mouse model, but PJ had a better effect than POE. Furthermore, PJ inhibited pyroptosis by decreasing the expression of the NLRP3 inflammasome, while also repairing the dysfunction of the intestinal barrier by upregulating the expression of tight junction proteins. Therefore, based on the study findings, we concluded that PJ can improve DSS-induced UC and may suppress pyroptosis by interfering with the activation of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Yi-Meng Fan
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultura University, Beijing, 100193, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing, 100193, People's Republic of China
- National Center of Technology Innovation for Medicinal function of Food, National Food and Strategic Reserves Administration, Beijing, 100193, China
| | - Yuan-Yuan Wei
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultura University, Beijing, 100193, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing, 100193, People's Republic of China
- National Center of Technology Innovation for Medicinal function of Food, National Food and Strategic Reserves Administration, Beijing, 100193, China
| | - Hui-Ru Wang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultura University, Beijing, 100193, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing, 100193, People's Republic of China
- National Center of Technology Innovation for Medicinal function of Food, National Food and Strategic Reserves Administration, Beijing, 100193, China
| | - Yu-Ga
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultura University, Beijing, 100193, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing, 100193, People's Republic of China
- National Center of Technology Innovation for Medicinal function of Food, National Food and Strategic Reserves Administration, Beijing, 100193, China
| | - Yan-Nan Zhang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultura University, Beijing, 100193, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing, 100193, People's Republic of China
- National Center of Technology Innovation for Medicinal function of Food, National Food and Strategic Reserves Administration, Beijing, 100193, China
| | - Zhihui Hao
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultura University, Beijing, 100193, China.
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing, 100193, People's Republic of China.
- National Center of Technology Innovation for Medicinal function of Food, National Food and Strategic Reserves Administration, Beijing, 100193, China.
| |
Collapse
|
41
|
Ganapathy AS, Saha K, Wang A, Arumugam P, Dharmaprakash V, Yochum G, Koltun W, Nighot M, Perdew G, Thompson TA, Ma T, Nighot P. Alpha-tocopherylquinone differentially modulates claudins to enhance intestinal epithelial tight junction barrier via AhR and Nrf2 pathways. Cell Rep 2023; 42:112705. [PMID: 37393618 PMCID: PMC10528852 DOI: 10.1016/j.celrep.2023.112705] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/25/2023] [Accepted: 06/12/2023] [Indexed: 07/04/2023] Open
Abstract
Defects in intestinal epithelial tight junctions (TJs) allow paracellular permeation of noxious luminal antigens and are important pathogenic factors in inflammatory bowel disease (IBD). We show that alpha-tocopherylquinone (TQ), a quinone-structured oxidation product of vitamin E, consistently enhances the intestinal TJ barrier by increasing barrier-forming claudin-3 (CLDN3) and reducing channel-forming CLDN2 in Caco-2 cell monolayers (in vitro), mouse models (in vivo), and surgically resected human colons (ex vivo). TQ reduces colonic permeability and ameliorates colitis symptoms in multiple colitis models. TQ, bifunctionally, activates both aryl hydrocarbon receptor (AhR) and nuclear factor erythroid 2-related factor 2 (Nrf2) pathways. Genetic deletion studies reveal that TQ-induced AhR activation transcriptionally increases CLDN3 via xenobiotic response element (XRE) in the CLDN3 promoter. Conversely, TQ suppresses CLDN2 expression via Nrf2-mediated STAT3 inhibition. TQ offers a naturally occurring, non-toxic intervention for enhancement of the intestinal TJ barrier and adjunct therapeutics to treat intestinal inflammation.
Collapse
Affiliation(s)
| | - Kushal Saha
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Alexandra Wang
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Priya Arumugam
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Viszwapriya Dharmaprakash
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Gregory Yochum
- Division of Colon and Rectal Surgery, Department of Surgery, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Walter Koltun
- Division of Colon and Rectal Surgery, Department of Surgery, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Meghali Nighot
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Gary Perdew
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA
| | - Todd A Thompson
- University of New Mexico College of Pharmacy, Albuquerque, NM 87131, USA
| | - Thomas Ma
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Prashant Nighot
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
42
|
Varani J, McClintock SD, Nadeem DM, Harber I, Zeidan D, Aslam MN. A multi-mineral intervention to counter pro-inflammatory activity and to improve the barrier in human colon organoids. Front Cell Dev Biol 2023; 11:1132905. [PMID: 37476158 PMCID: PMC10354648 DOI: 10.3389/fcell.2023.1132905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 06/21/2023] [Indexed: 07/22/2023] Open
Abstract
Introduction: Ulcerative colitis is a chronic inflammatory condition, and continuous inflammatory stimulus may lead to barrier dysfunction. The goal of this study was to assess barrier proteomic expression by a red algae-derived multi-mineral intervention in the absence or presence of pro-inflammatory insult. Methods: Human colon organoids were maintained in a control culture medium alone or exposed to lipopolysaccharide with a combination of three pro-inflammatory cytokines [tumor necrosis factor-α, interleukin-1β and interferon-γ (LPS-cytokines)] to mimic the environment in the inflamed colon. Untreated organoids and those exposed to LPS-cytokines were concomitantly treated for 14 days with a multi-mineral product (Aquamin®) that has previously been shown to improve barrier structure/function. The colon organoids were subjected to proteomic analysis to obtain a broad view of the protein changes induced by the two interventions alone and in combination. In parallel, confocal fluorescence microscopy, tissue cohesion and transepithelial electrical resistance (TEER) measurements were used to assess barrier structure/function. Results: The LPS-cytokine mix altered the expression of multiple proteins that influence innate immunity and promote inflammation. Several of these were significantly decreased with Aquamin® alone but only a modest decrease in a subset of these proteins was detected by Aquamin® in the presence of LPS-cytokines. Among these, a subset of inflammation-related proteins including fibrinogen-β and -γ chains (FGB and FGG), phospholipase A2 (PLA2G2A) and SPARC was significantly downregulated in the presence of Aquamin® (alone and in combination with LPS-cytokines); another subset of proteins with anti-inflammatory, antioxidant or anti-microbial activity was upregulated by Aquamin® treatment. When provided alone, Aquamin® strongly upregulated proteins that contribute to barrier formation and tissue strength. Concomitant treatment with LPS-cytokines did not inhibit barrier formation in response to Aquamin®. Confocal microscopy also displayed increased expression of desmoglein-2 (DSG2) and cadherin-17 (CDH17) with Aquamin®, either alone or in the presence of the pro-inflammatory stimulus. Increased cohesion and TEER with Aquamin® (alone or in the presence of LPS-cytokines) indicates improved barrier function. Conclusion: Taken together, these findings suggest that multi-mineral intervention (Aquamin®) may provide a novel approach to combating inflammation in the colon by improving barrier structure/function as well as by directly altering the expression of certain pro-inflammatory proteins.
Collapse
Affiliation(s)
| | | | | | | | | | - Muhammad N. Aslam
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
43
|
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
|
44
|
Capaldo CT. Claudin Barriers on the Brink: How Conflicting Tissue and Cellular Priorities Drive IBD Pathogenesis. Int J Mol Sci 2023; 24:8562. [PMID: 37239907 PMCID: PMC10218714 DOI: 10.3390/ijms24108562] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Inflammatory bowel diseases (IBDs) are characterized by acute or chronic recurring inflammation of the intestinal mucosa, often with increasing severity over time. Life-long morbidities and diminishing quality of life for IBD patients compel a search for a better understanding of the molecular contributors to disease progression. One unifying feature of IBDs is the failure of the gut to form an effective barrier, a core role for intercellular complexes called tight junctions. In this review, the claudin family of tight junction proteins are discussed as they are a fundamental component of intestinal barriers. Importantly, claudin expression and/or protein localization is altered in IBD, leading to the supposition that intestinal barrier dysfunction exacerbates immune hyperactivity and disease. Claudins are a large family of transmembrane structural proteins that constrain the passage of ions, water, or substances between cells. However, growing evidence suggests non-canonical claudin functions during mucosal homeostasis and healing after injury. Therefore, whether claudins participate in adaptive or pathological IBD responses remains an open question. By reviewing current studies, the possibility is assessed that with claudins, a jack-of-all-trades is master of none. Potentially, a robust claudin barrier and wound restitution involve conflicting biophysical phenomena, exposing barrier vulnerabilities and a tissue-wide frailty during healing in IBD.
Collapse
Affiliation(s)
- Christopher T Capaldo
- College of Natural and Computer Sciences, Hawai'i Pacific University, Honolulu, HI 96813, USA
| |
Collapse
|
45
|
Zhang Y, Wang L, Wang ZD, Zhou Q, Zhou X, Zhou T, Guan YX, Liu X. Surface-anchored microbial enzyme-responsive solid lipid nanoparticles enabling colonic budesonide release for ulcerative colitis treatment. J Nanobiotechnology 2023; 21:145. [PMID: 37127609 PMCID: PMC10152766 DOI: 10.1186/s12951-023-01889-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 04/10/2023] [Indexed: 05/03/2023] Open
Abstract
Colon-targeted oral drug delivery systems (CDDSs) are desirable for the treatment of ulcerative colitis (UC), which is a disease with high relapse and remission rates associated with immune system inflammation and dysregulation localized within the lining of the large bowel. However, the success of current available approaches used for colon-targeted therapy is limited. Budesonide (BUD) is a corticosteroid drug, and its rectal and oral formulations are used to treat UC, but the inconvenience of rectal administration and the systemic toxicity of oral administration restrict its long-term use. In this study, we designed and prepared colon-targeted solid lipid nanoparticles (SLNs) encapsulating BUD to treat UC by oral administration. A negatively charged surfactant (NaCS-C12) was synthesized to anchor cellulase-responsive layers consisting of polyelectrolyte complexes (PECs) formed by negatively charged NaCS and cationic chitosan onto the SLNs. The release rate and colon-specific release behavior of BUD could be easily modified by regulating the number of coated layers. We found that the two-layer BUD-loaded SLNs (SLN-BUD-2L) with a nanoscale particle size and negative zeta potential showed the designed colon-specific drug release profile in response to localized high cellulase activity. In addition, SLN-BUD-2L exhibited excellent anti-inflammatory activity in a dextran sulfate sodium (DSS)-induced colitis mouse model, suggesting its potential anti-UC applications.
Collapse
Affiliation(s)
- Yipeng Zhang
- Department of Pharmacology and Department of Radiology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314100, China
| | - Liying Wang
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Zi-Dan Wang
- Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Quan Zhou
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, 310058, China
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Xuefei Zhou
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Tianhua Zhou
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Yi-Xin Guan
- Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xiangrui Liu
- Department of Pharmacology and Department of Radiology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
- Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China.
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314100, China.
- Cancer Center, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
46
|
Thangaleela S, Sivamaruthi BS, Radha A, Kesika P, Chaiyasut C. Neuromyelitis Optica Spectrum Disorders: Clinical Perspectives, Molecular Mechanisms, and Treatments. APPLIED SCIENCES 2023; 13:5029. [DOI: 10.3390/app13085029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Neuromyelitis optica (NMO) is a rare autoimmune inflammatory disorder affecting the central nervous system (CNS), specifically the optic nerve and the spinal cord, with severe clinical manifestations, including optic neuritis (ON) and transverse myelitis. Initially, NMO was wrongly understood as a condition related to multiple sclerosis (MS), due to a few similar clinical and radiological features, until the discovery of the AQP4 antibody (NMO-IgG/AQP4-ab). Various etiological factors, such as genetic-environmental factors, medication, low levels of vitamins, and others, contribute to the initiation of NMO pathogenesis. The autoantibodies against AQP4 target the AQP4 channel at the blood–brain barrier (BBB) of the astrocyte end feet, which leads to high permeability or leakage of the BBB that causes more influx of AQP4-antibodies into the cerebrospinal fluid (CSF) of NMO patients. The binding of AQP4-IgG onto the AQP4 extracellular epitopes initiates astrocyte damage through complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC). Thus, a membrane attack complex is formed due to complement cascade activation; the membrane attack complex targets the AQP4 channels in the astrocytes, leading to astrocyte cell damage, demyelination of neurons and oligodendrocytes, and neuroinflammation. The treatment of NMOSD could improve relapse symptoms, restore neurological functions, and alleviate immunosuppression. Corticosteroids, apheresis therapies, immunosuppressive drugs, and B cell inactivating and complement cascade blocking agents have been used to treat NMOSD. This review intends to provide all possible recent studies related to molecular mechanisms, clinical perspectives, and treatment methodologies of the disease, particularly focusing on recent developments in clinical criteria and therapeutic formulations.
Collapse
Affiliation(s)
- Subramanian Thangaleela
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Arumugam Radha
- Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India
| | - Periyanaina Kesika
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chaiyavat Chaiyasut
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
47
|
Saha K, Subramenium Ganapathy A, Wang A, Michael Morris N, Suchanec E, Ding W, Yochum G, Koltun W, Nighot M, Ma T, Nighot P. Autophagy Reduces the Degradation and Promotes Membrane Localization of Occludin to Enhance the Intestinal Epithelial Tight Junction Barrier against Paracellular Macromolecule Flux. J Crohns Colitis 2023; 17:433-449. [PMID: 36219473 PMCID: PMC10069622 DOI: 10.1093/ecco-jcc/jjac148] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Indexed: 01/18/2023]
Abstract
BACKGROUND AND AIMS Functional loss of the gut epithelium's paracellular tight junction [TJ] barrier and defective autophagy are factors potentiating inflammatory bowel disease [IBD]. Previously, we showed the role of autophagy in enhancing the intestinal TJ barrier via pore-forming claudin-2 degradation. How autophagy regulates the TJ barrier-forming proteins remains unknown. Here, we investigated the role of autophagy in the regulation of occludin, a principal TJ component involved in TJ barrier enhancement. RESULTS Autophagy induction using pharmacological activators and nutrient starvation increased total occludin levels in intestinal epithelial cells, mouse colonocytes and human colonoids. Autophagy induction enriched membrane occludin levels and reduced paracellular permeability of macromolecules. Autophagy-mediated TJ barrier enhancement was contingent on the presence of occludin as OCLN-/- nullified its TJ barrier-enhancing effect against macromolecular flux. Autophagy inhibited the constitutive degradation of occludin by preventing its caveolar endocytosis from the membrane and protected against inflammation-induced TJ barrier loss. Autophagy enhanced the phosphorylation of ERK-1/2 and inhibition of these kinases in Caco-2 cells and human colonic mucosa prevented the macromolecular barrier-enhancing effects of autophagy. In vivo, autophagy induction by rapamycin enhanced occludin levels in wild-type mouse intestines and protected against lipopolysaccharide- and tumour necrosis factor-α-induced TJ barrier loss. Disruption of autophagy with acute Atg7 knockout in adult mice decreased intestinal occludin levels, increasing baseline colonic TJ permeability and exacerbating the effect of experimental colitis. CONCLUSION Our data suggest a novel role of autophagy in promoting the intestinal TJ barrier by increasing occludin levels in an ERK1/2 mitogen-activated protein kinase-dependent mechanism.
Collapse
Affiliation(s)
- Kushal Saha
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Ashwinkumar Subramenium Ganapathy
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Alexandra Wang
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Nathan Michael Morris
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Eric Suchanec
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Wei Ding
- Division of Colon and Rectal Surgery, Department of Surgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Gregory Yochum
- Division of Colon and Rectal Surgery, Department of Surgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Walter Koltun
- Division of Colon and Rectal Surgery, Department of Surgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Meghali Nighot
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Thomas Ma
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Prashant Nighot
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
48
|
Livanos AE, Dunn A, Fischer J, Ungaro RC, Turpin W, Lee SH, Rui S, Del Valle DM, Jougon JJ, Martinez-Delgado G, Riddle MS, Murray JA, Laird RM, Torres J, Agrawal M, Magee JS, Dervieux T, Gnjatic S, Sheppard D, Sands BE, Porter CK, Croitoru K, Petralia F, Colombel JF, Mehandru S. Anti-Integrin αvβ6 Autoantibodies Are a Novel Biomarker That Antedate Ulcerative Colitis. Gastroenterology 2023; 164:619-629. [PMID: 36634824 PMCID: PMC10284061 DOI: 10.1053/j.gastro.2022.12.042] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/14/2023]
Abstract
BACKGROUND & AIMS Better biomarkers for prediction of ulcerative colitis (UC) development and prognostication are needed. Anti-integrin αvβ6 (anti-αvβ6) autoantibodies have been described in patients with UC. We tested for the presence of anti-αvβ6 antibodies in the preclinical phase of UC and studied their association with disease-related outcomes after diagnosis. METHODS Anti-αvβ6 autoantibodies were measured in 4 longitudinal serum samples collected from 82 subjects who later developed UC and 82 matched controls from a Department of Defense preclinical cohort (PREDICTS [Proteomic Evaluation and Discovery in an IBD Cohort of Tri-service Subjects]). In a distinct, external validation cohort (Crohn's and Colitis Canada Genetic Environmental Microbial project cohort), we tested 12 pre-UC subjects and 49 matched controls. Furthermore, anti-αvβ6 autoantibodies were measured in 2 incident UC cohorts (COMPASS [Comprehensive Care for the Recently Diagnosed IBD Patients], n = 55 and OSCCAR [Ocean State Crohn's and Colitis Area Registry], n = 104) and associations between anti-αvβ6 autoantibodies and UC-related outcomes were defined using Cox proportional hazards model. RESULTS Anti-αvβ6 autoantibodies were significantly higher among individuals who developed UC compared with controls up to 10 years before diagnosis in PREDICTS. The anti-αvβ6 autoantibody seropositivity was 12.2% 10 years before diagnosis and increased to 52.4% at the time of diagnosis in subjects who developed UC compared with 2.7% in controls across the 4 time points. Anti-αvβ6 autoantibodies predicted UC development with an area under the curve of at least 0.8 up to 10 years before diagnosis. The presence of anti-αvβ6 autoantibodies in preclinical UC samples was validated in the GEM cohort. Finally, high anti-αvβ6 autoantibodies was associated with a composite of adverse UC outcomes, including hospitalization, disease extension, colectomy, systemic steroid use, and/or escalation to biologic therapy in recently diagnosed UC. CONCLUSIONS Anti-integrin αvβ6 autoantibodies precede the clinical diagnosis of UC by up to 10 years and are associated with adverse UC-related outcomes.
Collapse
Affiliation(s)
- Alexandra E Livanos
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alexandra Dunn
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jeremy Fischer
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ryan C Ungaro
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Williams Turpin
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology and Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Sun-Ho Lee
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology and Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shumin Rui
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Diane Marie Del Valle
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Human Immune Monitoring Center, Precision Institute of Immunology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Julia J Jougon
- Hepato-Gastroenterology Department, Claude Huriez Hospital, University of Lille, Lille, France
| | | | - Mark S Riddle
- University of Nevada, Reno School of Medicine, Reno, Nevada; Veterans Affairs Sierra Nevada Health Care System, Reno, Nevada
| | - Joseph A Murray
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Renee M Laird
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, Maryland; Henry M. Jackson Foundation for Military Medicine, Bethesda, Maryland
| | - Joana Torres
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; Gastroenterology Division, Hospital Beatriz Ângelo, Loures, Portugal; Gastroenterology Division, Hospital da Luz, Lisbon, Portugal; Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Manasi Agrawal
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jared S Magee
- Gastroenterology, Walter Reed National Military Medical Center, Bethesda, Maryland
| | | | - Sacha Gnjatic
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Human Immune Monitoring Center, Precision Institute of Immunology, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Dean Sheppard
- Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Bruce E Sands
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Chad K Porter
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, Maryland
| | - Kenneth Croitoru
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology and Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Francesca Petralia
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jean-Frederic Colombel
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Saurabh Mehandru
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
49
|
Rath T, Atreya R, Bodenschatz J, Uter W, Geppert CE, Vitali F, Fischer S, Waldner MJ, Colombel JF, Hartmann A, Neurath MF. Intestinal Barrier Healing Is Superior to Endoscopic and Histologic Remission for Predicting Major Adverse Outcomes in Inflammatory Bowel Disease: The Prospective ERIca Trial. Gastroenterology 2023; 164:241-255. [PMID: 36279923 DOI: 10.1053/j.gastro.2022.10.014] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND & AIMS Endoscopic and histologic remission have emerged as key therapeutic goals in the management of inflammatory bowel diseases (IBD) that are associated with favorable long-term disease outcomes. Here, we prospectively compared the predictive value of barrier healing with endoscopic and histologic remission for predicting long-term disease behavior in a large cohort of patients with IBD in clinical remission. METHODS At baseline, patients with IBD in clinical remission underwent ileocolonoscopy with assessment of intestinal barrier function by confocal endomicroscopy. Endoscopic and histologic disease activity, as well as barrier healing, was prospectively assessed along established scores. During subsequent follow-up, patients were closely monitored for clinical disease activity and the occurrence of major adverse outcomes (MAOs): disease flares, IBD-related hospitalization or surgery, and initiation or dose escalation of systemic steroids, immunosuppressants, small molecules, or biological therapy. RESULTS The final analysis included 181 patients, 100 with Crohn's disease [CD] and 81 with ulcerative colitis (UC). During a mean follow-up of 35 (CD) and 25 (UC) months, 73% of patients with CD and 69% of patients with UC experienced at least 1 MAO. The probability of MAO-free survival was significantly higher in patients with IBD with endoscopic remission compared with endoscopically active disease. In addition, histologic remission predicted MAO-free survival in patients with UC but not CD. Barrier healing on endomicroscopy was superior to endoscopic and histologic remission for predicting MAO-free survival in both UC and CD. CONCLUSIONS Barrier healing is associated with decreased risk of disease progression in patients with clinically remittent IBD, with superior predictive performance compared with endoscopic and histologic remission. Analysis of barrier function might be considered as a future treatment target in clinical trials. CLINICALTRIALS gov number, NCT05157750.
Collapse
Affiliation(s)
- Timo Rath
- Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Medical Clinic 1, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | - Raja Atreya
- Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Medical Clinic 1, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | - Julia Bodenschatz
- Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Medical Clinic 1, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | - Wolfgang Uter
- Department of Medical Informatics, Biometry and Epidemiology, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | - Carol E Geppert
- Institute for Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | - Francesco Vitali
- Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Medical Clinic 1, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | - Sarah Fischer
- Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Medical Clinic 1, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | - Maximilian J Waldner
- Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Medical Clinic 1, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | - Jean-Frédéric Colombel
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Arndt Hartmann
- Institute for Pathology, 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, Medical Clinic 1, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany.
| |
Collapse
|
50
|
Novel Horizons in Postbiotics: Lactobacillaceae Extracellular Vesicles and Their Applications in Health and Disease. Nutrients 2022; 14:nu14245296. [PMID: 36558455 PMCID: PMC9782203 DOI: 10.3390/nu14245296] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Lactobacillus probiotics contained in dietary supplements or functional foods are well-known for their beneficial properties exerted on host health and diverse pathological situations. Their capacity to improve inflammatory bowel disease (IBD) and regulate the immune system is especially remarkable. Although bacteria-host interactions have been thought to occur directly, the key role that extracellular vesicles (EVs) derived from probiotics play on this point is being unveiled. EVs are lipid bilayer-enclosed particles that carry a wide range of cargo compounds and act in different signalling pathways. Notably, these EVs have been recently proposed as a safe alternative to the utilisation of live bacteria since they can avoid the possible risks that probiotics may entail in vulnerable cases such as immunocompromised patients. Therefore, this review aims to give an updated overview of the existing knowledge about EVs from different Lactobacillus strains, their mechanisms and effects in host health and different pathological conditions. All of the information collected suggests that EVs could be considered as potential tools for the development of future novel therapeutic approaches.
Collapse
|