1
|
Rodrigues CS, Gaifem J, Pereira MS, Alves MF, Silva M, Padrão N, Cavadas B, Moreira-Barbosa C, Alves I, Marcos-Pinto R, Torres J, Lavelle A, Colombel JF, Sokol H, Pinho SS. Alterations in mucosa branched N-glycans lead to dysbiosis and downregulation of ILC3: a key driver of intestinal inflammation. Gut Microbes 2025; 17:2461210. [PMID: 39918275 PMCID: PMC11810091 DOI: 10.1080/19490976.2025.2461210] [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: 08/29/2024] [Revised: 11/29/2024] [Accepted: 01/13/2025] [Indexed: 02/12/2025] Open
Abstract
The perturbation of the symbiotic relationship between microbes and intestinal immune system contributes to gut inflammation and Inflammatory Bowel Disease (IBD) development. The host mucosa glycans (glycocalyx) creates a major biological interface between gut microorganisms and host immunity that remains ill-defined. Glycans are essential players in IBD immunopathogenesis, even years before disease onset. However, how changes in mucosa glycosylation shape microbiome and how this impact gut immune response and inflammation remains to be clarified. Here, we revealed that alterations in the expression of complex branched N-glycans at gut mucosa surface, modeled in glycoengineered mice, resulted in dysbiosis, with a deficiency in Firmicutes bacteria. Concomitantly, this mucosa N-glycan switch was associated with a downregulation of type 3 innate lymphoid cells (ILC3)-mediated immune response, leading to the transition of ILC3 toward an ILC1 proinflammatory phenotype and increased TNFα production. In addition, we demonstrated that the mucosa glycosylation remodeling through prophylactic supplementation with glycans at steady state was able to restore microbial-derived short-chain fatty acids and microbial sensing (by NOD2 expression) alongside the rescue of the expression of ILC3 module, suppressing intestinal inflammation and controlling disease onset. In a complementary approach, we further showed that IBD patients, often displaying dysbiosis, exhibited a tendency of decreased MGAT5 expression at epithelial cells that was accompanied by reduced ILC3 expression in gut mucosa. Altogether, these results unlock the effects of alterations in mucosa glycome composition in the regulation of the bidirectional crosstalk between microbiota and gut immune response, revealing host branched N-glycans/microbiota/ILC3 axis as an essential pathway in gut homeostasis and in preventing health to intestinal inflammation transition.
Collapse
Affiliation(s)
- Cláudia S. Rodrigues
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Joana Gaifem
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
| | - Márcia S. Pereira
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Maria Francisca Alves
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
- Faculty of Sciences, University of Porto, Porto, Portugal
| | - Mariana Silva
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Nuno Padrão
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Bruno Cavadas
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
| | | | - Inês Alves
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
| | - Ricardo Marcos-Pinto
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
- Department of Gastroenterology, Centro Hospitalar do Porto, Porto, Portugal
- Centro de Investigação em Tecnologias e Serviços de Saúde, University of Porto, Porto, Portugal
| | - Joana Torres
- Division of Gastroenterology, Hospital Beatriz Ângelo, Loures, Portugal
- Faculty of Medicine, University of Lisbon, Lisbon, Portugal
- Division of Gastroenterology, Hospital da Luz, Lisbon, Portugal
| | - Aonghus Lavelle
- Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Gastroenterology Department, Sorbonne Université, INSERM, Paris, France
| | - Jean-Frederic Colombel
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Harry Sokol
- Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Gastroenterology Department, Sorbonne Université, INSERM, Paris, France
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Salomé S. Pinho
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
2
|
Zhao X, He MJ, Zhao M, Li HR, Zhuang ZM, Xing Y, Zhang XL, Zhao P. Crude Polygalae Radix after boiling with licorice decoction alleviates intestinal mucosal barrier injury of rats by regulating TLR4/NF-κB signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119661. [PMID: 40120702 DOI: 10.1016/j.jep.2025.119661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/11/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Polygala tenuifolia Willd. (pharmacologically termed Polygalae Radix, PR), a nootropic botanical in traditional Chinese medicine, demonstrates anxiolytic and cognitive-enhancing properties with two millennia of documented therapeutic applications. Long-term or large-dose use of crude Polygalae Radix (CPR) causes intestinal injury, which could be reduced by use of Glycyrrhiza uralensis Fisch. (licorice) decoction-boiled Polygalae Radix. However, the effects of boiling CPR with licorice decoction on reducing intestinal mucosal barrier injury have not been studied. AIM OF THE STUDY Our research mainly focused on the alleviating effects and underlying mechanism of CPR after boiling with licorice decoction on intestinal mucosal barrier injury in rats. METHODS AND MATERIALS SD rats were orally administered CPR and licorice decoction-boiled PR (LPR) extracts respectively for 15 consecutive days. Subsequently, levels of pro-inflammatory cytokines and immunoglobulins were measured, and histopathological changes in intestinal tissues were examined. The mRNA expression levels of pro-inflammatory cytokines were evaluated by qRT-PCR. The expression difference of TLR4/NF-κB signaling pathway key protein and tight junction (TJ) protein were evaluated using Western blotting and immunohistochemistry. RESULTS Processing PR with licorice decoction significantly ameliorated the downregulation of intestinal TJ proteins (occludin, claudin-1, and ZO-1) and elevated serum lipopolysaccharide levels induced by CPR. It alleviated the suppression of intestinal immunoglobulin A, serum immunoglobulin A and immunoglobulin G levels caused by CPR while mitigating intestinal mucosal injury and inflammatory responses. Additionally, processing PR with licorice decoction inhibited CPR-triggered upregulation of TLR4, NF-κB p65, p-NF-κB p65, and p-κBα proteins expression, while preventing IκBα downregulation in intestinal tissues. Furthermore, it significantly suppressed the upregulation of interleukin (IL)-6, IL-8, and tumor necrosis factor-α (TNF-α) mRNA expression while concurrently inhibiting the secretion levels of these pro-inflammatory cytokines in small intestine. CONCLUSION Our experimental data suggest that licorice decoction boiling effectively prevents CPR-induced reductions in TJ proteins and immunoglobulins expression, alleviates intestinal mucosal barrier injuries, and mediates these effects through suppression of TLR4/NF-κB signaling pathway activation and subsequent production of IL-6, IL-8, and TNF-α.
Collapse
Affiliation(s)
- Xin Zhao
- Medical School, Shandong Xiehe University, Jinan, 250109, PR China; Department of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Meng-Jiao He
- Department of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Meng Zhao
- Medical School, Shandong Xiehe University, Jinan, 250109, PR China
| | - Hao-Ran Li
- Department of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Zi-Ming Zhuang
- Department of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Yue Xing
- Department of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Xue-Lan Zhang
- Medical School, Shandong Xiehe University, Jinan, 250109, PR China; Department of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China.
| | - Pan Zhao
- Department of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China.
| |
Collapse
|
3
|
Wu XQ, Wan JW, Yang ZN, Liu HJ, Chang Y, Peng SB, Niu XT, Kong YD, Li M, Chen XM, Wang GQ. Protection of glutamine: The NF-κB/MLCK/MLC2 signaling pathway mediated by tight junction affects oxidative stress, inflammation and apoptosis in snakehead (Channa argus). FISH & SHELLFISH IMMUNOLOGY 2025; 158:110131. [PMID: 39826630 DOI: 10.1016/j.fsi.2025.110131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/27/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Lipopolysaccharide (LPS) destroys intestinal mechanical barrier and causes apoptosis by triggering oxidative stress and inflammatory responses. Glutamine (Gln) can maintain normal intestinal function under various stressed or pathological conditions. Thereby, this study aims to evaluate the protection of glutamine on intestinal health of snakehead (Channa argus), specifically regarding the NF-κB/MLCK/MLC2 signaling pathway mediated by tight junction affecting oxidative stress, inflammation and apoptosis. In this work, a model of intestinal tight junction injury in intestine of snakehead was constructed by injecting 4 mg/mL LPS into anus for 96 h. Before constructing the model, fish were treated with different levels of alanyl-glutamine (Ala-Gln) (0 %, 0.3 %, 0.6 %, 0.9 %, 1.2 % and 1.5 %) for 56 days. Microstructure and ultra microstructure showed that LPS-induced obvious intestinal damage and tight connection destruction, while Gln effectively alleviated these phenomena. In addition, results also showed that Gln can effectively inhibit LPS-induced damage to intestinal tight junction (zo-1, occludin, claudin5, claudin1, nf-κb p65, mlck and mlc2), alleviate oxidative stress (nrf2, sod, gsh, gpx and cat), ameliorate intestinal inflammation (tnf-α, il-1β, il-8, tlr5 and tlr2), thereby reduce apoptosis (p38mapk, caspase9, caspase8, caspase3 and bax). Crucially, the above results were related to NF-κB/MLCK/MLC2 signaling pathway mediated by tight junction. In conclusion, Gln has a good protective effect on LPS-induced intestinal injury in northern snakehead, providing a new perspective for regulating fish intestinal health.
Collapse
Affiliation(s)
- Xue-Qin Wu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Ji-Wu Wan
- Fisheries Technology Extension Station of Jilin Province, Changchun, 130012, China
| | - Zhi-Nan Yang
- Fisheries Technology Extension Station of Jilin Province, Changchun, 130012, China
| | - Hong-Jian Liu
- Fisheries Technology Extension Station of Jilin Province, Changchun, 130012, China
| | - Yue Chang
- Fisheries Technology Extension Station of Jilin Province, Changchun, 130012, China
| | - Si-Bo Peng
- Jilin Academy of Fishery Sciences, Changchun, 130033, China
| | - Xiao-Tian Niu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Yi-di Kong
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Min Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Xiu-Mei Chen
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China.
| | - Gui-Qin Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
4
|
Worakajit N, Satitsri S, Kitiyakara T, Muanprasat C. Myosin light chain kinase-mediated epithelial barrier dysfunction as a potential pathogenic mechanism of afatinib-induced diarrheas: A study in human colonoid model. Eur J Pharmacol 2025; 987:177174. [PMID: 39637932 DOI: 10.1016/j.ejphar.2024.177174] [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: 05/15/2024] [Revised: 08/22/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Diarrheas are an important adverse effect of afatinib, a tyrosine kinase inhibitor (TKI) anti-cancer drug, leading to mortality and morbidity in cancer patients with their pathophysiological mechanisms related to intestinal barrier dysfunctions being poorly understood. This study aimed to investigate the effect of afatinib on intestinal epithelial barrier integrity using a human colon-derived organoid model (colonoids). Afatinib (0.5 μM) significantly decreased the transepithelial electrical resistance (TEER) by ∼60% and increased apical-to-basolateral dextran flux by > 20 folds without causing apparent cytotoxicity in human colonoids. The delocalization of zonula occludens-1 (ZO-1) and a decrease in mRNA and protein expression of claudin-4 and ZO-1 were also observed in the afatinib-treated human colonoids. Afatinib induced nuclear translocation of nuclear factor kappa B (NF-κB) as well as mRNA and protein expression of NF-κB targets including tumor necrosis factor (TNF)-alpha, interleukin-8 (IL-8), and inducible nitric oxide synthase (iNOS) indicating the initiation of the NF-κB-mediated epithelial inflammatory responses. Interestingly, afatinib induced mRNA and protein expression of myosin light chain (MLC) kinase (MLCK) and MLC phosphorylation, a known inducer of intestinal epithelial barrier disruption. Treatment with iNOS inhibitor (1400W) or MLCK inhibitor (ML-7) reversed the effect of afatinib on mRNA expressions of ZO-1 and claudin-4, and TEER. Collectively, our results indicate that afatinib induces intestinal epithelial barrier dysfunction via mechanisms involving NF-κB-iNOS-MLCK pathways. This finding may pave the way for developing therapeutic strategies to reduce adverse effects and enhance efficacy of TKI in cancer patients.
Collapse
Affiliation(s)
- Nichakorn Worakajit
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Ratchathewi, Bangkok, 10400, Thailand; Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bang Phli, Samut Prakarn, 10540, Thailand
| | - Saravut Satitsri
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bang Phli, Samut Prakarn, 10540, Thailand
| | - Taya Kitiyakara
- Division of Gastroenterology and Hepatology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Chatchai Muanprasat
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bang Phli, Samut Prakarn, 10540, Thailand.
| |
Collapse
|
5
|
Tang S, Wu S, Zhang W, Ma L, Zuo L, Wang H. Immunology and treatments of fatty liver disease. Arch Toxicol 2025; 99:127-152. [PMID: 39692857 DOI: 10.1007/s00204-024-03920-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024]
Abstract
Alcoholic liver disease (ALD) and non-alcoholic fatty liver disease (NAFLD) are two major chronic liver diseases worldwide. The triggers for fatty liver can be derived from external sources such as adipose tissue, the gut, personal diet, and genetics, or internal sources, including immune cell responses, lipotoxicity, hepatocyte death, mitochondrial dysfunction, and extracellular vesicles. However, their pathogenesis varies to some extent. This review summarizes various immune mechanisms and therapeutic targets associated with these two types of fatty liver disease. It describes the gut-liver axis and adipose tissue-liver crosstalk, as well as the roles of different immune cells (both innate and adaptive immune cells) in fatty liver disease. Additionally, mitochondrial dysfunction, extracellular vesicles, microRNAs (miRNAs), and gastrointestinal hormones are also related to the pathogenesis of fatty liver. Understanding the pathogenesis of fatty liver and corresponding therapeutic strategies provides a new perspective for developing novel treatments for fatty liver disease.
Collapse
Affiliation(s)
- Sainan Tang
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Science, Anhui Medical University, Hefei, 230022, Anhui, China
| | - Shanshan Wu
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, Anhui, China
| | - Wenzhe Zhang
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
- The First College of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Lili Ma
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
- The First College of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Li Zuo
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China.
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Science, Anhui Medical University, Hefei, 230022, Anhui, China.
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
6
|
Shin HY, Jeong WB, Joung MY, Shin KS, Yu KW. Effects of Centella asiatica-isolated pectic polysaccharide on dextran sulfate sodium-induced colitis. Int J Biol Macromol 2024; 285:138237. [PMID: 39622374 DOI: 10.1016/j.ijbiomac.2024.138237] [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: 08/28/2024] [Revised: 11/15/2024] [Accepted: 11/29/2024] [Indexed: 12/08/2024]
Abstract
Dietary supplementation of polysaccharides demonstrates strong therapeutic actions on inflammatory bowel disease (IBD). Centella asiatica (CA) has traditionally been used in Ayurveda and Chinese medicine. However, the effects of CA-isolated pectic polysaccharides on IBD remain unknown. This study determined the effect of a CA-isolated pectic polysaccharide on IBD. The crude polysaccharide (CA-CP), isolated from a hot-water extract of CA, is a typical pectic polysaccharide composed mainly of galacturonic acid (40.6 %), galactose (27.6 %), arabinose (13.5 %), and rhamnose (8.5 %). CA-CP improved clinical symptoms in a DSS-induced colitis murine model, including weight change (9.9-12.0 %), disease activity index (31.6-51.9 %), colon length (13.2-21.5 %), and spleen weight (21.8-26.3 %). CA-CP effectively regulated the levels of inflammatory and junctional factors by mediating the MAPK and NF-κB pathways. CA-CP partially alleviated DSS-induced crypt destruction, submucosal edema, inflammatory infiltration, and mucin secretion. The content of total short-chain fatty acids increased substantially (cecum 29.8-53.7 %, feces 75.4-109.3 %) with oral CA-CP administration compared to the DSS group. Cecal microbial community analysis revealed that CA-CP administration regulated DSS-induced colitis by reducing the abundance of Escherichia (5.0-10.9 %) and Clostridium (0.3-0.4 %), while increasing the abundance of Bacteroidetes (6.8-8.5 %), Ligilactobacillus (2.7-6.0 %), and Bilophilia (0.3-0.6 %). The findings provide fundamental data for developing novel functional therapeutic agents for the prevention and treatment of colitis, using CA-isolated pectic polysaccharides. Furthermore, to the best of our knowledge, our study is the first to demonstrate the effects of pectic polysaccharides isolated from CA on IBD.
Collapse
Affiliation(s)
- Hyun Young Shin
- Transdisciplinary Major in Learning Health Systems, Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea.
| | - Won Bi Jeong
- Major in Food & Nutrition, Korea National University of Transportation, Chungbuk 27909, Republic of Korea.
| | - Mi Yeun Joung
- Corporation ChamSunJin Green Juice, Jincheon 27865, Republic of Korea.
| | - Kwang-Soon Shin
- Department of Food Science and Biotechnology, Kyonggi University, Suwon 16227, Republic of Korea.
| | - Kwang-Won Yu
- Major in Food & Nutrition, Korea National University of Transportation, Chungbuk 27909, Republic of Korea.
| |
Collapse
|
7
|
Wang K, Liu X, Huang H, Suo M, Wang J, Liu X, Zhang J, Chen X, Li Z. A new target for treating intervertebral disk degeneration: gut microbes. Front Microbiol 2024; 15:1452774. [PMID: 39678913 PMCID: PMC11638241 DOI: 10.3389/fmicb.2024.1452774] [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: 06/21/2024] [Accepted: 09/16/2024] [Indexed: 12/17/2024] Open
Abstract
Intervertebral disk degeneration (IDD) is a common clinical spinal disease and one of the main causes of low back pain (LBP). Generally speaking, IDD is considered a natural degenerative process with age. However, with the deepening of research, people have discovered that IDD is not only related to age, but also has many factors that can induce and accelerate its progression. In addition, the pathogenesis of IDD remains unclear, resulting in limited traditional treatment methods that cannot effectively prevent and treat IDD. Conservative treatment may lead to patients' dependence on drugs, and the pain relief effect is not obvious. Similarly, surgical treatment is highly invasive, with a longer recovery time and a higher recurrence rate. With the deepening of exploration, people have discovered that intestinal microorganisms are an important symbiotic microbial community in the human body and are closely related to the occurrence and development of various diseases. Changes in intestinal microorganisms and their metabolites may affect the body's inflammatory response, immune regulation, and metabolic processes, thereby affecting the health of the intervertebral disk. In this context, the gut microbiota has received considerable attention as a potential target for delaying or treating IDD. This article first introduces the impact of gut microbes on common distal organs, and then focuses on three potential mechanisms by which gut microbes and their metabolites influence IDD. Finally, we also summarized the methods of delaying or treating IDD by interfering with intestinal microorganisms and their metabolites. Further understanding of the potential mechanisms between intestinal microorganisms and IDD will help to formulate reasonable IDD treatment strategies to achieve ideal therapeutic effects.
Collapse
Affiliation(s)
- Kaizhong Wang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Dalian, Liaoning, China
| | - Xiangyan Liu
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Dalian, Liaoning, China
| | - Huagui Huang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Dalian, Liaoning, China
| | - Moran Suo
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Dalian, Liaoning, China
| | - Jinzuo Wang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Dalian, Liaoning, China
| | - Xin Liu
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Dalian, Liaoning, China
| | - Jing Zhang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Dalian, Liaoning, China
| | - Xin Chen
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhonghai Li
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Dalian, Liaoning, China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, Liaoning, China
| |
Collapse
|
8
|
Xu D, Wang X, Hou X, Wang X, Shi W, Hu Y. The effect of Lonicerae flos and Rhizoma curcumae longae extract on the intestinal development and function of broilers. Poult Sci 2024; 103:104225. [PMID: 39217666 PMCID: PMC11402626 DOI: 10.1016/j.psj.2024.104225] [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: 05/16/2024] [Revised: 07/26/2024] [Accepted: 08/11/2024] [Indexed: 09/04/2024] Open
Abstract
This study was conducted to explore effects of Lonicerae flos and Rhomoma curcumae longae extracts (LR) on intestinal function of broilers. Three hundred broiler chickens were randomly assigned to the following 5 groups. The control group were fed the basal diet; the antibiotic group were fed the basal diet supplemented with spectinomycin hydrochloride (50 million units/ton) + lincomycin hydrochloride (25 g/ton); the LRH, LRM and LRL groups were fed the basal diet supplemented with a high dose (750 g/ton of feed), normal dose (500 g/ton of feed), or low dose (250 g/ton of feed) of LR, respectively. The changes of intestinal structure, intestinal digestive enzyme activities, antioxidant enzyme activities, inflammatory cytokines, and bacterial abundances in the colon and cecum contents were determined. The results indicated that compared with the control group and the antibiotic group, LR significantly increased the villus length/crypt depth (VCR) of the intestine, and significantly inhibited oxidative stress and inflammatory responses in the broiler intestine. In addition, LR regulated intestinal function by increasing the abundance of the intestinal microorganisms in broilers. In conclusion, LR improved antioxidant capacity, intestinal morphology, and microorganisms, and inhibited inflammatory response. The effect of high and medium doses of LR was better than lower doses.
Collapse
Affiliation(s)
- Dahai Xu
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding, 071000, China; State Key Laboratory of Animal Nutrition and feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xiao Wang
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding, 071000, China
| | - Xiaojiao Hou
- Beijing Centre Biology Co., Ltd., Beijing 102600, China
| | - Xiumin Wang
- Beijing Centre Biology Co., Ltd., Beijing 102600, China
| | - Wanyu Shi
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding, 071000, China.
| | - Yongfei Hu
- State Key Laboratory of Animal Nutrition and feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
9
|
Budiailmiawan L, Aryati A, Kadir NA, Yusuf LI, Partakusuma LG, Markus L, Lismayanti L. Anaemia Profile and Inflammation Markers in Stunted Children Under Two Years in Indonesia. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1315. [PMID: 39594890 PMCID: PMC11592781 DOI: 10.3390/children11111315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024]
Abstract
Background: Stunting is a common issue affecting children who suffer from chronic malnutrition in Indonesia. The Indonesian government has introduced supplementary food programs for stunted children, but the results have been less satisfactory. This may be due to the presence of anaemia and comorbid diseases. Haematology tests and inflammation markers are necessary to identify these conditions. This study aimed to examine the anaemia profiles and inflammation markers in stunted children under two years old. Methods: A cross-sectional descriptive design with cluster samples and consecutive analysis was used. The study was conducted between December 2023 and March 2024 at the West Nusa Tenggara Hospital and Palabuhanratu Sukabumi Hospital laboratories. Samples were obtained from various Public Health Centres in Sukabumi, West Java, North Maluku, and West Nusa Tenggara. Data collection comprised interviews, measurements, and the assessment of haematology, biochemical, and inflammatory markers. Statistical analysis was conducted using SPSS version 20, which includes descriptive analysis, correlation, comparison, and chi-square tests. Results: Two hundred and ten stunted children were identified with various anaemias and comorbidities. These anaemias included suspected thalassemia (38.1%), iron deficiency (18.1%), and anaemia of chronic diseases (13.3%). Based on the inflammatory markers obtained, TB was suspected (21.4%), inflammatory bowel disease (18.1%) was suspected, and allergic proctocolitis was suspected (31.9%). Conclusions: Analysis of the anaemia profiles and inflammatory markers revealed various types of anaemia and suspected comorbidities in stunted children. It is recommended that anaemia profiles and inflammation markers be assessed at the primary healthcare level.
Collapse
Affiliation(s)
| | - Aryati Aryati
- Departement of Clinical Pathology, Faculty of Medicine, Universitas Airlangga, Surabaya 60286, East Java, Indonesia
- Institute of Tropical Diseases, Universitas Airlangga, Surabaya 60115, East Java, Indonesia
| | - Nursin Abd. Kadir
- Departement of Clinical Pathology, Faculty of Medicine, Universitas Hasanudin, Makassar 90245, South Sulawesi, Indonesia;
| | - Laily Indrayanti Yusuf
- General Hospital of West Nusa Tenggara Province, Mataram 84371, West Nusa Tenggara, Indonesia;
| | - Lia Gardenia Partakusuma
- Post Graduate Programme, Faculty of Medicine, YARSI University, Jakarta 10510, DKI Jakarta, Indonesia;
| | - Louisa Markus
- Regional General Hospital Cengkareng, Jakarta 11730, DKI Jakarta, Indonesia;
| | - Leni Lismayanti
- Departement of Clinical Pathology, Faculty of Medicine, Padjadjaran University, Dr. Hasan Sadikin Central General Hospital, Bandung 40161, West Java, Indonesia;
| |
Collapse
|
10
|
Balogun O, Brownmiller CR, Lee SO, Kang HW. Onion Peel Extract Prevents Intestinal Inflammation via AMK-Activated Protein Kinase Activation in Caco-2/HT-29 Cells. Nutrients 2024; 16:3609. [PMID: 39519442 PMCID: PMC11547908 DOI: 10.3390/nu16213609] [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: 09/12/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Obesogenic diets cause intestinal inflammation and dysfunction. Polyphenols have shown a positive impact on reducing inflammation in in vitro studies. However, their bioactivity may not be the same in the in vivo system due to structural alteration by the gastrointestinal digestive process. The purpose of this study was to investigate the anti-inflammatory effect of onion peel and its major bioactive compound, quercetin, in the intestine and further examine the impact of intestinal digestion on this effect. METHODS Onion peel extract (OPE) and quercetin (Q) were digested using gastrointestinal digestive enzymes in vitro and then treated into lipopolysaccharide (LPS)-stimulated Caco-2/HT-29 cells. Genes and proteins related to tight junction, inflammation, and epithelial integrity were measured. RESULTS OPE and digested OPE (DOPE) had a higher protective effect on LPS-induced tight junction and inflammatory genes and paracellular permeability than Q and digested Q (DQ). DOPE was more effective than OPE, while digestion did not change the activity of Q. The anti-inflammatory effect of OPE and Q with or without digestion was achieved by inhibiting nuclear factor kappa B through AMP-activated protein kinase-activated silent mating-type information regulation 2 homolog 1. CONCLUSIONS It was the first to find that a crude extract, after undergoing gastrointestinal digestion, demonstrated a notably superior anti-inflammatory effect in the cell study, suggesting the consumption of onion peels could potentially yield similar benefits in the human intestine. This discovery underscores the potential of onion peel polyphenols in combating intestinal inflammation, making them a compelling area of research for future therapeutic applications using food byproducts.
Collapse
Affiliation(s)
- Olugbenga Balogun
- Applied Science and Technology, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, USA;
| | - Cindi R. Brownmiller
- Department of Food Science, University of Arkansas, Fayetteville, AR 72704, USA; (C.R.B.); (S.-O.L.)
| | - Sun-Ok Lee
- Department of Food Science, University of Arkansas, Fayetteville, AR 72704, USA; (C.R.B.); (S.-O.L.)
| | - Hye Won Kang
- Food and Nutritional Sciences, Department of Family and Consumer Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, USA
| |
Collapse
|
11
|
Wu Q, Kan J, Fu C, Liu X, Cui Z, Wang S, Le Y, Li Z, Liu Q, Zhang Y, Du J. Insights into the unique roles of extracellular vesicles for gut health modulation: Mechanisms, challenges, and perspectives. CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 7:100301. [PMID: 39525958 PMCID: PMC11550031 DOI: 10.1016/j.crmicr.2024.100301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Extracellular vesicles (EVs), which play significant regulatory roles in maintaining homeostasis and influencing immune responses, significantly impact gut microbiota composition and function, affecting overall gut health. Despite considerable progress, there are still knowledge gaps regarding the mechanisms by which EVs, including plant-derived EVs (PDEVs), animal-derived EVs (ADEVs), and microbiota-derived EVs (MDEVs), modulate gut health. This review delves into the roles and mechanisms of EVs from diverse sources in regulating gut health, focusing on their contributions to maintaining epithelial barrier integrity, facilitating tissue healing, eliciting immune responses, controlling pathogens, and shaping microbiota. We emphasize open challenges and future perspectives for harnessing EVs in the modulation of gut health to gain a deeper understanding of their roles and impact. Importantly, a comprehensive research framework is presented to steer future investigations into the roles and implications of EVs on gut health, facilitating a more profound comprehension of this emerging field.
Collapse
Affiliation(s)
- Qiming Wu
- Nutrilite Health Institute, Shanghai 200031, China
| | - Juntao Kan
- Nutrilite Health Institute, Shanghai 200031, China
| | - Caili Fu
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
| | - Xin Liu
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
| | - Zhengying Cui
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
| | - Sixu Wang
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
| | - Yi Le
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
| | - Zhanming Li
- Department of Food Quality and Safety, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Qin Liu
- Centre for Chinese Medicine Drug Development Limited, Hong Kong Baptist University, 999077, Hong Kong Special Administrative Region of China
| | - Yuyu Zhang
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Jun Du
- Nutrilite Health Institute, Shanghai 200031, China
| |
Collapse
|
12
|
Yokoi F, Deguchi S, Watanabe Y, Takayama K. Establishment of an ulcerative colitis model using colon organoids derived from human induced pluripotent stem cells. iScience 2024; 27:111049. [PMID: 39435148 PMCID: PMC11492162 DOI: 10.1016/j.isci.2024.111049] [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: 02/28/2024] [Revised: 05/31/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024] Open
Abstract
The etiology of inflammatory bowel disease (IBD) is complex, with much room for a greater understanding and development of improved therapies. Therefore, establishing a reliable IBD model is crucial for future advancements. In this study, human induced pluripotent stem (iPS) cell-derived colon organoids (hiPSC-COs) were treated with a combination of tumor necrosis factor alpha (TNF-α), interferon-gamma (IFN-γ), and interleukin (IL)-1β (3 cytokines [3CK]), known to be elevated in the serum of IBD patients. Inflammatory responses in stromal cells and damage to intestinal epithelial cells were observed in the 3CK-treated hiPSC-COs. Comparison of molecular signatures of 3CK-treated hiPSC-COs with those of ulcerative colitis (UC) patient's colon revealed that 3CK-treated hiPSC-COs resemble UC patient's colon. Furthermore, the elevated production of inflammatory cytokines observed in 3CK-treated hiPSC-COs was attenuated by treatment with tofacitinib. Our UC model will be an essential tool to understand its pathologic mechanisms and identify effective therapeutic approaches.
Collapse
Affiliation(s)
- Fuki Yokoi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Sayaka Deguchi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Yukio Watanabe
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Kazuo Takayama
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
- AMED-CREST, Japan Agency for Medical Research and Development (AMED), Tokyo 100-0004, Japan
| |
Collapse
|
13
|
El-Saadony MT, Yang T, Saad AM, Alkafaas SS, Elkafas SS, Eldeeb GS, Mohammed DM, Salem HM, Korma SA, Loutfy SA, Alshahran MY, Ahmed AE, Mosa WFA, Abd El-Mageed TA, Ahmed AF, Fahmy MA, El-Tarabily MK, Mahmoud RM, AbuQamar SF, El-Tarabily KA, Lorenzo JM. Polyphenols: Chemistry, bioavailability, bioactivity, nutritional aspects and human health benefits: A review. Int J Biol Macromol 2024; 277:134223. [PMID: 39084416 DOI: 10.1016/j.ijbiomac.2024.134223] [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: 09/10/2022] [Revised: 06/17/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
Polyphenols, including phenolics, alkaloids, and terpenes, are secondary metabolites that are commonly found in fruits, vegetables, and beverages, such as tea, coffee, wine, chocolate, and beer. These compounds have gained considerable attention and market demand because of their potential health benefits. However, their application is limited due to their low absorption rates and reduced tissue distribution efficiency. Engineering polyphenol-protein complexes or conjugates can enhance the antioxidant properties, bioavailability, and stability of polyphenols and improve digestive enzyme hydrolysis, target-specific delivery, and overall biological functions. Complex polyphenols, such as melanin, tannins, and ellagitannins, can promote gut microbiota balance, bolster antioxidant defense, and improve overall human health. Despite these benefits, the safety of polyphenol complexes must be thoroughly evaluated before their use as functional food additives or supplements. This review provides a detailed overview of the types of macromolecular polyphenols, their chemical composition, and their role in food enrichment. The mechanisms by which complex polyphenols act as antioxidative, anti-inflammatory, and anticancer agents have also been discussed.
Collapse
Affiliation(s)
- Mohamed T El-Saadony
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | - Tao Yang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Pharmacy, Hainan Medical University, Haikou, 571199, China
| | - Ahmed M Saad
- Department of Biochemistry, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | - Samar Sami Alkafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Sara Samy Elkafas
- Production Engineering and Mechanical Design Department, Faculty of Engineering, Menofia University, Shebin El Kom, 32511, Egypt; Faculty of Control System and Robotics, Information Technologies, Mechanics and Optics (ITMO) University, Saint-Petersburg, Russia
| | - Gehad S Eldeeb
- Department of Food Technology, Faculty of Agriculture, Suez Canal University, Ismailia, 41522, Egypt
| | - Dina Mostafa Mohammed
- Nutrition and Food Sciences Department, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Heba M Salem
- Department of Poultry Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Sameh A Korma
- Department of Food Science, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | - Samah A Loutfy
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, 12211, Egypt
| | - Mohammad Y Alshahran
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, 9088, Saudi Arabia
| | - Ahmed Ezzat Ahmed
- Department of Biology, College of Science, King Khalid University, Abha, 61421, Saudi Arabia
| | - Walid F A Mosa
- Plant Production Department (Horticulture-Pomology), Faculty of Agriculture, Saba Basha, Alexandria University, Alexandria, 21531, Egypt
| | - Taia A Abd El-Mageed
- Soil and Water Department, Faculty of Agriculture, Fayoum University, Fayoum, 63514, Egypt
| | - Atef F Ahmed
- Department of Biology, College of Science, Taif University, Taif, 21944, Saudi Arabia
| | - Mohamed A Fahmy
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | | | - Reda M Mahmoud
- Dr Nutrition Pharmaceuticals (DNP), Dubai, 48685, United Arab Emirates
| | - Synan F AbuQamar
- Department of Biology, United Arab Emirates University, Al Ain, 15551, United Arab Emirates.
| | - Khaled A El-Tarabily
- Department of Biology, United Arab Emirates University, Al Ain, 15551, United Arab Emirates; Harry Butler Institute, Murdoch University, Murdoch, 6150, W.A., Australia
| | - José M Lorenzo
- Centro Tecnologico´ de La Carne de Galicia, Rúa Galicia No. 4, Parque Tecnologico de Galicia, San Cibrao das Vinas, Ourense, 32900, Spain; Universidad de Vigo, Area´ de Tecnología de Los Alimentos, Facultad de Ciencias de Ourense, Ourense, 32004, Spain
| |
Collapse
|
14
|
Zhu W, Cremonini E, Mastaloudis A, Oteiza PI. Glucoraphanin and sulforaphane mitigate TNFα-induced Caco-2 monolayers permeabilization and inflammation. Redox Biol 2024; 76:103359. [PMID: 39298837 PMCID: PMC11426148 DOI: 10.1016/j.redox.2024.103359] [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: 09/02/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024] Open
Abstract
Intestinal permeabilization is central to the pathophysiology of chronic gut inflammation. This study investigated the efficacy of glucoraphanin (GR), prevalent in cruciferous vegetables, particularly broccoli, and its derivative sulforaphane (SF), in inhibiting tumor necrosis factor alpha (TNFα)-induced Caco-2 cell monolayers inflammation and permeabilization through the regulation of redox-sensitive events. TNFα binding to its receptor led to a rapid increase in oxidant production and subsequent elevation in the mRNA levels of NOX1, NOX4, and Duox2. GR and SF dose-dependently mitigated both these short- and long-term alterations in redox homeostasis. Downstream, GR and SF inhibited the activation of the redox-sensitive signaling cascades NF-κB (p65 and IKK) and MAPK ERK1/2, which contribute to inflammation and barrier permeabilization. GR (1 μM) and SF (0.5-1 μM) prevented TNFα-induced monolayer permeabilization and the associated reduction in the levels of the tight junction (TJ) proteins occludin and ZO-1. Both GR and SF also mitigated TNFα-induced increased mRNA levels of the myosin light chain kinase, which promotes TJ opening. Molecular docking suggests that although GR is mostly not absorbed, it could interact with extracellular and membrane sites in NOX1. Inhibition of NOX1 activity by GR would mitigate TNFα receptor downstream signaling and associated events. These findings support the concept that not only SF, but also GR, could exert systemic health benefits by protecting the intestinal barrier against inflammation-induced permeabilization, in part by regulating redox-sensitive pathways. GR has heretofore not been viewed as a biologically active molecule, but rather, the benign precursor of highly active SF. The consumption of GR and/or SF-rich vegetables or supplements in the diet may offer a means to mitigate the detrimental consequences of intestinal permeabilization, not only in disease states but also in conditions characterized by chronic inflammation of dietary and lifestyle origin.
Collapse
Affiliation(s)
- Wei Zhu
- Department of Nutrition, University of California, Davis, CA, USA
| | | | | | - Patricia I Oteiza
- Department of Nutrition, University of California, Davis, CA, USA; Department of Environmental Toxicology, University of California, Davis, CA, USA.
| |
Collapse
|
15
|
Bahaa MM, Hegazy SK, Maher MM, Bahgat MM, El-Haggar SM. Pentoxifylline in patients with ulcerative colitis treated with mesalamine by modulation of IL-6/STAT3, ZO-1, and S1P pathways: a randomized controlled double-blinded study. Inflammopharmacology 2024; 32:3247-3258. [PMID: 39192162 DOI: 10.1007/s10787-024-01560-6] [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: 03/26/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Ulcerative colitis (UC) is an inflammatory bowel disease (IBD) that lasts a long time and has a variety of causes. AIM The primary aim of this study was to evaluate pentoxifylline's (PTX) essential function in patients with UC. METHODS Fifty-two mild to moderate UC patients who matched the eligibility requirements participated in this clinical study. One gram of mesalamine (t.i.d.) and a placebo were administered to the mesalamine group (n = 26) for a duration of 24 weeks. Mesalamine 1 g t.i.d. and PTX 400 mg two times daily were administered to the PTX group (n = 26) for 24 weeks. A gastroenterologist investigated patients at the start and 6 months after the medication was given to assess disease activity index (DAI) and numeric pain rating scale (NRS). Also, interleukin-6 (IL-6), sphingosine 1 phosphate (S1P), tumor necrosis factor-alpha (TNF-α), and fecal myeloperoxidase (MPO) were measured before and after therapy. Zonula occuldin-1 (ZO-1) and signal transducer and activator of transcription factor-3 (STAT-3) expression was assessed before and after therapy as well as histological assessment. Short Form 36 Health Survey (SF-36), was assessed for each patient before and after 6 months of treatment. RESULTS The PTX group showed statistically lower levels of serum SIP, TNF-α, IL-6, faecal MPO, gene expression of STAT-3, and a significant increase of ZO-1 in comparison with the mesalamine group. DAI and NRS significantly decreased whereas SF-36 significantly increased in the PTX group. CONCLUSION PTX could alleviate inflammation in patients with UC, so it might be promising adjunctive for patients with UC. TRIAL REGISTRATION IDENTIFIER NCT05558761.
Collapse
Affiliation(s)
- Mostafa M Bahaa
- Pharmacy Practice Department, Faculty of Pharmacy, Horus University, New Damietta, Egypt.
| | - Sahar K Hegazy
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, El-Guiesh Street, El-Gharbia Government, Tanta, 31527, Egypt
| | - Maha M Maher
- Internal Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Internal Medicine Department, Faculty of Medicine, Horus University, New Damietta, Egypt
| | - Monir M Bahgat
- Internal Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Internal Medicine Department, Faculty of Medicine, Horus University, New Damietta, Egypt
| | - Sahar M El-Haggar
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, El-Guiesh Street, El-Gharbia Government, Tanta, 31527, Egypt
| |
Collapse
|
16
|
Zhu W, Xiong L, Oteiza PI. Structure-dependent capacity of procyanidin dimers to inhibit inflammation-induced barrier dysfunction in a cell model of intestinal epithelium. Redox Biol 2024; 75:103275. [PMID: 39059205 PMCID: PMC11327484 DOI: 10.1016/j.redox.2024.103275] [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: 06/25/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Diet is of major importance in modulating intestinal inflammation, as the gastrointestinal tract is directly exposed to high concentrations of dietary components. Procyanidins are flavan-3-ol oligomers abundant in fruits and vegetables. Although with limited or no intestinal absorption, they can have GI health benefits which can promote overall health. We previously observed that epicatechin gallate (ECG) and epigallocatechin gallate (EGCG) dimers inhibit in vitro colorectal cancer cell proliferation and invasiveness. Inflammation-mediated intestinal barrier permeabilization can result in a chronic inflammatory condition and promote colorectal cancer onset/progression. Thus, this study investigated the structure-dependent capacity of ECG, EGCG and (-)-epicatechin (EC) dimers to inhibit tumor necrosis factor alpha (TNFα)-induced inflammation, oxidative stress, and loss of barrier integrity in Caco-2 cells differentiated into an intestinal epithelial cell monolayer. Cells were incubated with TNFα (10 ng/ml), in the absence/presence of ECG, EGCG and EC dimers. The three dimers inhibited TNFα-mediated Caco-2 cell monolayer permeabilization, modulating events involved in the loss of barrier function and inflammation, i.e. decreased tight junction protein levels; increased matrix metalloproteinases expression and activity; increased NADPH oxidase expression and oxidant production; activation of the NF-κB and ERK1/2 pathways and downstream events leading to tight junction opening. For some of these mechanisms, the galloylated ECG and EGCG dimers had stronger protective potency than the non-galloylated EC dimer. These differences could be due to differential membrane interactions as pointed out by molecular dynamics simulation of procyanidin dimers-cell membrane interactions and/or by differential interactions with NOX1. Results show that dimeric procyanidins, although poorly absorbed, can promote health by alleviating intestinal inflammation, oxidative stress and barrier permeabilization.
Collapse
Affiliation(s)
- Wei Zhu
- Department of Nutrition, University of California, Davis, CA, 95618, USA
| | - Le Xiong
- Cleveland Clinic, Cleveland, OH, 44194, USA
| | - Patricia I Oteiza
- Department of Nutrition, University of California, Davis, CA, 95618, USA; Department of Environmental Toxicology, University of California, Davis, CA, 95618, USA.
| |
Collapse
|
17
|
Abdulqadir R, Al-Sadi R, Haque M, Gupta Y, Rawat M, Ma TY. Bifidobacterium bifidum Strain BB1 Inhibits Tumor Necrosis Factor-α-Induced Increase in Intestinal Epithelial Tight Junction Permeability via Toll-Like Receptor-2/Toll-Like Receptor-6 Receptor Complex-Dependent Stimulation of Peroxisome Proliferator-Activated Receptor γ and Suppression of NF-κB p65. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1664-1683. [PMID: 38885924 PMCID: PMC11372998 DOI: 10.1016/j.ajpath.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/16/2024] [Accepted: 05/16/2024] [Indexed: 06/20/2024]
Abstract
Bifidobacterium bifidum strain BB1 causes a strain-specific enhancement in intestinal epithelial tight junction (TJ) barrier. Tumor necrosis factor (TNF)-α induces an increase in intestinal epithelial TJ permeability and promotes intestinal inflammation. The major purpose of this study was to delineate the protective effect of BB1 against the TNF-α-induced increase in intestinal TJ permeability and to unravel the intracellular mechanisms involved. TNF-α produces an increase in intestinal epithelial TJ permeability in Caco-2 monolayers and in mice. Herein, the addition of BB1 inhibited the TNF-α increase in Caco-2 intestinal TJ permeability and mouse intestinal permeability in a strain-specific manner. BB1 inhibited the TNF-α-induced increase in intestinal TJ permeability by interfering with TNF-α-induced enterocyte NF-κB p50/p65 and myosin light chain kinase (MLCK) gene activation. The BB1 protective effect against the TNF-α-induced increase in intestinal permeability was mediated by toll-like receptor-2/toll-like receptor-6 heterodimer complex activation of peroxisome proliferator-activated receptor γ (PPAR-γ) and PPAR-γ pathway inhibition of TNF-α-induced inhibitory kappa B kinase α (IKK-α) activation, which, in turn, resulted in a step-wise inhibition of NF-κB p50/p65, MLCK gene, MLCK kinase activity, and MLCK-induced opening of the TJ barrier. In conclusion, these studies unraveled novel intracellular mechanisms of BB1 protection against the TNF-α-induced increase in intestinal TJ permeability. The current data show that BB1 protects against the TNF-α-induced increase in intestinal epithelial TJ permeability via a PPAR-γ-dependent inhibition of NF-κB p50/p65 and MLCK gene activation.
Collapse
Affiliation(s)
- Raz Abdulqadir
- Department of Medicine, Penn State College of Medicine, Hershey Medical Center, Hershey, Pennsylvania.
| | - Rana Al-Sadi
- Department of Medicine, Penn State College of Medicine, Hershey Medical Center, Hershey, Pennsylvania
| | - Mohammad Haque
- Department of Medicine, Penn State College of Medicine, Hershey Medical Center, Hershey, Pennsylvania
| | - Yash Gupta
- Department of Medicine, Penn State College of Medicine, Hershey Medical Center, Hershey, Pennsylvania
| | - Manmeet Rawat
- Department of Medicine, Penn State College of Medicine, Hershey Medical Center, Hershey, Pennsylvania
| | - Thomas Y Ma
- Department of Medicine, Penn State College of Medicine, Hershey Medical Center, Hershey, Pennsylvania.
| |
Collapse
|
18
|
Radler JB, McBride AR, Saha K, Nighot P, Holmes GM. Regional Heterogeneity in Intestinal Epithelial Barrier Permeability and Mesenteric Perfusion After Thoracic Spinal Cord Injury. Dig Dis Sci 2024; 69:3236-3248. [PMID: 39001959 DOI: 10.1007/s10620-024-08537-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/18/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Spinal cord injury (SCI) disrupts intestinal barrier function, thereby increasing antigen permeation and leading to poor outcomes. Despite the intestinal tract's anatomic and physiologic heterogeneity, studies following SCI have not comprehensively addressed intestinal pathophysiology with regional specificity. AIMS AND METHODS We used an experimental model of high thoracic SCI to investigate (1) regional mucosal oxidative stress using dihydroethidium labeling; (2) regional paracellular permeability to small- and large-molecular probes via Ussing chamber; (3) regional intestinal tight junction (TJ) protein expression; and (4) hindgut perfusion via the caudal mesenteric artery. RESULTS Dihydroethidium staining was significantly elevated within duodenal mucosa at 3-day post-SCI. Molar flux of [14C]-urea was significantly elevated in duodenum and proximal colon at 3-day post-SCI, while molar flux of [3H]-inulin was significantly elevated only in duodenum at 3-day post-SCI. Barrier permeability was mirrored by a significant increase in the expression of pore-forming TJ protein claudin-2 in duodenum and proximal colon at 3-day post-SCI. Claudin-2 expression remained significantly elevated in proximal colon at 3-week post-SCI. Expression of the barrier-forming TJ protein occludin was significantly reduced in duodenum at 3-day post-SCI. Caudal mesenteric artery flow was unchanged by SCI at 3 days or 3 weeks despite significant reductions in mean arterial pressure. CONCLUSION These data show that T3-SCI provokes elevated mucosal oxidative stress, altered expression of TJ proteins, and elevated intestinal barrier permeability in the proximal intestine. In contrast, mucosal oxidative stress and intestinal barrier permeability were unchanged in the hindgut after SCI. This regional heterogeneity may result from differential sensitivity to reduced mesenteric perfusion, though further studies are required to establish a causal link. Understanding regional differences in intestinal pathophysiology is essential for developing effective treatments and standards of care for individuals with SCI.
Collapse
Affiliation(s)
- Jackson B Radler
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, 500 University Dr., H109, Hershey, PA, 17033, USA
| | - Amanda R McBride
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, 500 University Dr., H109, Hershey, PA, 17033, USA
- Cooper Medical School of Rowan University, Camden, NJ, 08103, USA
| | - Kushal Saha
- Division of Gastroenterology and Hepatology, Department of Medicine, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Prashant Nighot
- Division of Gastroenterology and Hepatology, Department of Medicine, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Gregory M Holmes
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, 500 University Dr., H109, Hershey, PA, 17033, USA.
| |
Collapse
|
19
|
Adeniyi-Ipadeola GO, Hankins JD, Kambal A, Zeng XL, Patil K, Poplaski V, Bomidi C, Nguyen-Phuc H, Grimm SL, Coarfa C, Stossi F, Crawford SE, Blutt SE, Speer AL, Estes MK, Ramani S. Infant and adult human intestinal enteroids are morphologically and functionally distinct. mBio 2024; 15:e0131624. [PMID: 38953637 PMCID: PMC11323560 DOI: 10.1128/mbio.01316-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 05/29/2024] [Indexed: 07/04/2024] Open
Abstract
Human intestinal enteroids (HIEs) are gaining recognition as physiologically relevant models of the intestinal epithelium. While HIEs from adults are used extensively in biomedical research, few studies have used HIEs from infants. Considering the dramatic developmental changes that occur during infancy, it is important to establish models that represent infant intestinal characteristics and physiological responses. We established jejunal HIEs from infant surgical samples and performed comparisons to jejunal HIEs from adults using RNA sequencing (RNA-Seq) and morphologic analyses. We then validated differences in key pathways through functional studies and determined whether these cultures recapitulate known features of the infant intestinal epithelium. RNA-Seq analysis showed significant differences in the transcriptome of infant and adult HIEs, including differences in genes and pathways associated with cell differentiation and proliferation, tissue development, lipid metabolism, innate immunity, and biological adhesion. Validating these results, we observed a higher abundance of cells expressing specific enterocyte, goblet cell, and enteroendocrine cell markers in differentiated infant HIE monolayers, and greater numbers of proliferative cells in undifferentiated 3D cultures. Compared to adult HIEs, infant HIEs portray characteristics of an immature gastrointestinal epithelium including significantly shorter cell height, lower epithelial barrier integrity, and lower innate immune responses to infection with an oral poliovirus vaccine. HIEs established from infant intestinal tissues reflect characteristics of the infant gut and are distinct from adult cultures. Our data support the use of infant HIEs as an ex vivo model to advance studies of infant-specific diseases and drug discovery for this population. IMPORTANCE Tissue or biopsy stem cell-derived human intestinal enteroids are increasingly recognized as physiologically relevant models of the human gastrointestinal epithelium. While enteroids from adults and fetal tissues have been extensively used for studying many infectious and non-infectious diseases, there are few reports on enteroids from infants. We show that infant enteroids exhibit both transcriptomic and morphological differences compared to adult cultures. They also differ in functional responses to barrier disruption and innate immune responses to infection, suggesting that infant and adult enteroids are distinct model systems. Considering the dramatic changes in body composition and physiology that begin during infancy, tools that appropriately reflect intestinal development and diseases are critical. Infant enteroids exhibit key features of the infant gastrointestinal epithelium. This study is significant in establishing infant enteroids as age-appropriate models for infant intestinal physiology, infant-specific diseases, and responses to pathogens.
Collapse
Affiliation(s)
| | - Julia D. Hankins
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Amal Kambal
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Texas Medical Center Digestive Diseases Center Gastrointestinal Experimental Model Systems (GEMS) Core, Houston, Texas, USA
| | - Xi-Lei Zeng
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Texas Medical Center Digestive Diseases Center Gastrointestinal Experimental Model Systems (GEMS) Core, Houston, Texas, USA
| | - Ketki Patil
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Victoria Poplaski
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Carolyn Bomidi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Hoa Nguyen-Phuc
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Sandra L. Grimm
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Center for Precision and Environmental Health, Baylor College of Medicine, Houston, Texas, USA
| | - Cristian Coarfa
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Center for Precision and Environmental Health, Baylor College of Medicine, Houston, Texas, USA
| | - Fabio Stossi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Gulf Coast Consortium Center for Advanced Microscopy and Image Informatics, Houston, Texas, USA
| | - Sue E. Crawford
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Sarah E. Blutt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Texas Medical Center Digestive Diseases Center Gastrointestinal Experimental Model Systems (GEMS) Core, Houston, Texas, USA
| | - Allison L. Speer
- Department of Pediatric Surgery, The University of Texas Health Science Center, Houston, Texas, USA
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Texas Medical Center Digestive Diseases Center Gastrointestinal Experimental Model Systems (GEMS) Core, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
20
|
Li J, Li Q, Ma W, Zhang Y, Li X. Expression of MAF bZIP transcription factor B protects against ulcerative colitis through the inhibition of the NF-κB pathway. Immun Inflamm Dis 2024; 12:e1372. [PMID: 39172054 PMCID: PMC11340633 DOI: 10.1002/iid3.1372] [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: 12/18/2023] [Revised: 07/22/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024] Open
Abstract
PURPOSE The aim of this study was to explore whether MAF bZIP transcription factor B (MAFB) might alleviate ulcerative colitis (UC) in dextran sulfate sodium (DSS)-induced mice and LPS-induced IEC-6 cells. METHODS UC in vivo and in vitro model was established by using DSS and LPS, respectively. The mice body weight and disease activity index (DAI) score were recorded daily, and colon length was measured. Moreover, the permeability was evaluated utilizing a fluorescein isothiocyanate dextran (FITC-Dextran) probe. Histopathological changes of DSS-induced colitis mice was assessed utilizing H&E staining. Next, qRT-PCR was performed to detect IL-1β, IL-6, TNF-α, and IL-10 level in in vivo and in vitro. Furthermore, the level of MDA, SOD, CAT, and GSH were evaluated in colon tissues. Besides, the expressions of tight junction proteins and NF-κB pathway relative proteins were examined in colitis mice and IEC-6 cells using western blot, immunohistochemistry and immunofluorescence. RESULTS MAFB level was downregulated in DSS-induced colitis mice. Moreover, the upregulation of MAFB protected mice from DSS-induced colitis by suppressing DSS-induced inflammation, oxidative stress, and intestinal barrier impairment. We also demonstrated that the upregulation of MAFB inactivated NF-κB pathway in DSS-caused colitis mice. Subsequently, we observed that MAFB upregulation could inhibit LPS-caused epithelial barrier impairment and inflammation in IEC-6 cells. Additionally, MAFB overexpression could suppress the activation of NF-κB pathway in IEC-6 cells. CONCLUSION The upregulation of MAFB could protect against UC via the suppression of inflammation and the intestinal barrier impairment through inhibiting the NF-κB pathway.
Collapse
Affiliation(s)
- Jingwen Li
- Department of GastroenterologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
| | - Qingmin Li
- Department of General PracticeShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Department of MedicineZhangqiu District Gaoguanzhai Community Health Service CenterJinanShandongChina
| | - Wei Ma
- Department of General PracticeShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
| | - Yongsheng Zhang
- Department of General PracticeShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
| | - Xiaonan Li
- Department of General PracticeShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
| |
Collapse
|
21
|
Haque M, Kaminsky L, Abdulqadir R, Engers J, Kovtunov E, Rawat M, Al-Sadi R, Ma TY. Lactobacillus acidophilus inhibits the TNF-α-induced increase in intestinal epithelial tight junction permeability via a TLR-2 and PI3K-dependent inhibition of NF-κB activation. Front Immunol 2024; 15:1348010. [PMID: 39081324 PMCID: PMC11286488 DOI: 10.3389/fimmu.2024.1348010] [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: 12/01/2023] [Accepted: 06/25/2024] [Indexed: 08/02/2024] Open
Abstract
Background Defective intestinal epithelial tight junction (TJ), characterized by an increase in intestinal TJ permeability, has been shown to play a critical role in the pathogenesis of inflammatory bowel disease (IBD). Tumor necrosis factor-α (TNF-α) is a key pro-inflammatory cytokine involved in the immunopathology of IBD and has been shown to cause an increase in intestinal epithelial TJ permeability. Although TNF-α antibodies and other biologics have been advanced for use in IBD treatment, these therapies are associated with severe side effects and have limited efficacy, and there is an urgent need for therapies with benign profiles and high therapeutic efficacy. Probiotic bacteria have beneficial effects and are generally safe and represent an important class of potential therapeutic agents in IBD. Lactobacillus acidophilus (LA) is one of the most used probiotics for wide-ranging health benefits, including in gastrointestinal, metabolic, and inflammatory disorders. A specific strain of LA, LA1, was recently demonstrated to have protective and therapeutic effects on the intestinal epithelial TJ barrier. However, the mechanisms of actions of LA1 remain largely unknown. Methods The primary aim of this study was to investigate microbial-epithelial interactions and novel signaling pathways that regulate the effect of LA1 on TNF-α-induced increase in intestinal epithelial TJ permeability, using cell culture and animal model systems. Results and Conclusion Pre-treatment of filter-grown Caco-2 monolayers with LA1 prevented the TNF-α-induced increase in intestinal epithelial TJ permeability by inhibiting TNF-α-induced activation of NF-κB p50/p65 and myosin light chain kinase (MLCK) gene and kinase activity in a TLR-2-dependent manner. LA1 produced a TLR-2- and MyD88-dependent activation of NF-κB p50/p65 in immune cells; however, LA1, in intestinal cells, inhibited the NF-κB p50/p65 activation in a TLR-2-dependent but MyD88-independent manner. In addition, LA1 inhibition of NF-κB p50/p65 and MLCK gene was mediated by TLR-2 pathway activation of phosphatidylinositol 3-kinase (PI3K) and IKK-α phosphorylation. Our results demonstrated novel intracellular signaling pathways by which LA1/TLR-2 suppresses the TNF-α pathway activation of NF-κB p50/p65 in intestinal epithelial cells and protects against the TNF-α-induced increase in intestinal epithelial TJ permeability.
Collapse
Affiliation(s)
- Mohammad Haque
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Lauren Kaminsky
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Raz Abdulqadir
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Jessica Engers
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Evgeny Kovtunov
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Manmeet Rawat
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Rana Al-Sadi
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Thomas Y. Ma
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| |
Collapse
|
22
|
Hsu MF, Koike S, Chen CS, Najjar SM, Meng TC, Haj FG. Pharmacological inhibition of the Src homology phosphatase 2 confers partial protection in a mouse model of alcohol-associated liver disease. Biomed Pharmacother 2024; 175:116590. [PMID: 38653109 DOI: 10.1016/j.biopha.2024.116590] [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: 09/26/2023] [Revised: 04/06/2024] [Accepted: 04/10/2024] [Indexed: 04/25/2024] Open
Abstract
Alcohol-associated liver disease (ALD) is a leading factor of liver-related death worldwide. ALD has various manifestations that include steatosis, hepatitis, and cirrhosis and is currently without approved pharmacotherapies. The Src homology phosphatase 2 (Shp2) is a drug target in some cancers due to its positive regulation of Ras-mitogen-activated protein kinase signaling and cell proliferation. Shp2 pharmacological inhibition yields beneficial outcomes in animal disease models, but its impact on ALD remains unexplored. This study aims to investigate the effects of Shp2 inhibition and its validity using a preclinical mouse model of ALD. We report that the administration of SHP099, a potent and selective allosteric inhibitor of Shp2, partially ameliorated ethanol-induced hepatic injury, inflammation, and steatosis in mice. Additionally, Shp2 inhibition was associated with reduced ethanol-evoked activation of extracellular signal-regulated kinase (ERK), oxidative, and endoplasmic reticulum (ER) stress in the liver. Besides the liver, excessive alcohol consumption induces multi-organ injury and dysfunction, including the intestine. Notably, Shp2 inhibition diminished ethanol-induced intestinal inflammation and permeability, abrogated the reduction in tight junction protein expression, and the activation of ERK and stress signaling in the ileum. Collectively, Shp2 pharmacological inhibition mitigates the deleterious effects of ethanol in the liver and intestine in a mouse model of ALD. Given the multifactorial aspects underlying ALD pathogenesis, additional studies are needed to decipher the utility of Shp2 inhibition alone or as a component in a multitherapeutic regimen to combat this deadly malady.
Collapse
Affiliation(s)
- Ming-Fo Hsu
- Department of Nutrition, University of California Davis, One Shields Ave, Davis, CA 95616, USA.
| | - Shinichiro Koike
- Department of Nutrition, University of California Davis, One Shields Ave, Davis, CA 95616, USA
| | - Chang-Shan Chen
- Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei, Taiwan
| | - Sonia M Najjar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA; Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Tzu-Ching Meng
- Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei, Taiwan
| | - Fawaz G Haj
- Department of Nutrition, University of California Davis, One Shields Ave, Davis, CA 95616, USA; Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA; Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, University of California Davis, Sacramento, CA 95817, USA.
| |
Collapse
|
23
|
Tang H, Zhou H, Zhang L, Tang T, Li N. Molecular mechanism of MLCK1 inducing 5-Fu resistance in colorectal cancer cells through activation of TNFR2/NF-κB pathway. Discov Oncol 2024; 15:159. [PMID: 38735014 PMCID: PMC11089027 DOI: 10.1007/s12672-024-01019-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 05/07/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND AND AIMS Chemotherapy resistance in colorectal cancer have been faced with significant challenges in recent years. Particular interest is directed to tumor microenvironment function. Recent work has, identified a small molecule named Divertin that prevents myosin light chain kinase 1(MLCK1) recruitment to the perijunctional actomyosin ring(PAMR), restores barrier function after tumor necrosis factor(TNF)-induced barrier loss and prevents disease progression in experimental inflammatory bowel disease. Studies have shown that MLCK is a potential target for affecting intestinal barrier function, as well as for tumor therapy. However, the relative contributions of MLCK expression and chemotherapy resistance in colorectal cancers have not been defined. METHODS Statistical analysis of MYLK gene expression differences in colorectal cancer patients and normal population and prognosis results from The Cancer Genome Atlas(TCGA) data. Cell activity was detected by Cell counting Kit-8. Cell proliferation was detected by monoclonal plate. The apoptosis was detected by flow cytometry and western blot. Determine the role of MLCK1 in inducing 5-Fluorouracil(5-Fu) resistance in colorectal cancer cells was detected by overexpression of MLCK1 and knock-down expression of MLCK1. RESULTS MLCK1 is expressed at different levels in different colorectal cancer cells, high MLCK1 expressing cell lines are less sensitive to 5-Fu, and low MLCK1 expressing cell lines are more sensitive to 5-Fu. MLCK1 high expression enhances resistance to 5-Fu in colorectal cancer cells and the sensitivity to 5-Fu was increased after knocking down the expression of MLCK1, that might be closely correlated to TNFR2/NF-κB pathway. CONCLUSIONS MLCK1 high expression can enhance resistance to 5-Fu in colorectal cancer cells and the sensitivity to 5-Fu was increased after knocking down the expression of MLCK1, that might be closely correlated to TNFR2/NF-κB pathway, which will provide a new method for the treatment of colorectal cancer patients who are resistant to 5-Fu chemotherapy.
Collapse
Affiliation(s)
- Huifen Tang
- Department of Hematology, The Affiliated Hospital, Hangzhou Normal University, 126# Wenzhou Road, Hangzhou, 310015, Zhejiang, People's Republic of China
| | - Hui Zhou
- Department of Hematology, The Affiliated Hospital, Hangzhou Normal University, 126# Wenzhou Road, Hangzhou, 310015, Zhejiang, People's Republic of China
| | - Liang Zhang
- Department of Hematology, The Affiliated Hospital, Hangzhou Normal University, 126# Wenzhou Road, Hangzhou, 310015, Zhejiang, People's Republic of China
| | - Tingting Tang
- Department of Hematology, The Affiliated Hospital, Hangzhou Normal University, 126# Wenzhou Road, Hangzhou, 310015, Zhejiang, People's Republic of China
| | - Ning Li
- Department of Hematology, The Affiliated Hospital, Hangzhou Normal University, 126# Wenzhou Road, Hangzhou, 310015, Zhejiang, People's Republic of China.
| |
Collapse
|
24
|
Xu W, Hua Z, Wang Y, Tang W, Ou W, Liu F, Yang Y, Ding W, Wang Z, Cui L, Ge W, Gu Y, Wang X, Chen Y, Liu CY, Du P. AMBRA1 promotes intestinal inflammation by antagonizing PP4R1/PP4c mediated IKK dephosphorylation in an autophagy-independent manner. Cell Death Differ 2024; 31:618-634. [PMID: 38424148 PMCID: PMC11094188 DOI: 10.1038/s41418-024-01275-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/02/2024] Open
Abstract
IκB kinase (IKK) complex is central regulators of the NF-κB pathway, and dysregulation of IKK phosphorylation leads to hyperactivation of proinflammatory response in various chronic inflammatory diseases, including inflammatory bowel disease (IBD). However, the dynamic modulation of IKK phosphorylation and dephosphorylation in intestinal inflammation remains uncharacterized. Here, we found that autophagy/beclin-1 regulator 1 (AMBRA1) was highly expressed in inflamed colons in a colitis mouse model and in clinical IBD samples. Importantly, AMBRA1 deletion significantly decreased proinflammatory cytokine expression and enhanced the therapeutic effect of infliximab on intestinal inflammation. Mechanistically, the N-term F1 domain of AMBRA1 was required for AMBRA1 to competitively interact with protein phosphatase 4 regulatory subunit 1 (PP4R1) and catalytic protein phosphatase 4 (PP4c) to suppress their interactions with IKK, promote the dissociation of the PP4R1/PP4c complex, and antagonize the dephosphorylation activity of this complex towards the IKK complex. In response to TNF-α stimulation, IKKα phosphorylates AMBRA1 at S1043 to stabilize AMBRA1 expression by impairing its binding to Cullin4A (CUL4A) to decrease its CUL4A-mediated K48-linked ubiquitination. Overall, our study identifies an autophagy-independent function of AMBRA1 as a positive modulator of IKK phosphorylation to promote intestinal inflammation, thus providing a new targeted therapeutic strategy for patients with refractory IBD.
Collapse
Affiliation(s)
- Weimin Xu
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200092, China
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China
| | - Zhebin Hua
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200092, China
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China
| | - Yaosheng Wang
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200092, China
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China
| | - Wenbo Tang
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200092, China
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China
| | - Weijun Ou
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200092, China
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China
| | - Fangyuan Liu
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200092, China
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China
| | - Yiqing Yang
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200092, China
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China
| | - Wenjun Ding
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200092, China
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China
| | - Zhongchuan Wang
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200092, China
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China
| | - Long Cui
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200092, China
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China
| | - Wensong Ge
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200092, China
| | - Yubei Gu
- Department of Gastroenterology, Rui Jin Hospital, affiliate to Shanghai Jiao Tong University, school of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Xiaolei Wang
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - YingWei Chen
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200092, China.
| | - Chen-Ying Liu
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200092, China.
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China.
| | - Peng Du
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200092, China.
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China.
| |
Collapse
|
25
|
Belaid M, Javorovic J, Pastorin G, Vllasaliu D. Development of an in vitro co-culture model using Caco-2 and J774A.1 cells to mimic intestinal inflammation. Eur J Pharm Biopharm 2024; 197:114243. [PMID: 38432601 DOI: 10.1016/j.ejpb.2024.114243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 03/05/2024]
Abstract
In vitro models that mimic the pathophysiology in vivo are important tools to study mechanisms of disease and assess the pharmacology and toxicity of drugs. In this work, we report the development of a novel model of intestinal inflammation. This model is based on the co-culture of intestinal epithelial Caco-2 cells and murine J774A.1 macrophages. The model is shown to mimic the intestinal barrier in both healthy and inflamed state. In the healthy state, without external stimulation, Caco-2 and J774A.1 cells were co-cultured in one system without affecting the barrier integrity of intestinal epithelial cells and without inducing release of cytokines from macrophages. To mimic the inflamed intestine, Caco-2 cells were primed with an optimised cytokine cocktail (TNF-⍺, IFN-γ and IL-1β) and J774A.1 cells were pre-exposed to lipopolysaccharide (LPS) and IFN-γ for 24 h before combining the two cell lines into co-culture. In these conditions, a significant disruption of the epithelial barrier and an increase in pro-inflammatory cytokine (TNF-⍺ and IL-6) levels released from macrophages were detected. The data also show that inflammation in the co-culture model was temporary and reversible upon the removal of the inflammatory stimulus. This new in vitro model could be a valuable tool for investigating the safety and efficacy of drugs in the context of intestinal inflammation and provides advantages over other reported co-culture models of intestinal inflammation in terms of cost and simplicity.
Collapse
Affiliation(s)
- Mona Belaid
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 9NH, United Kingdom; Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Jana Javorovic
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 9NH, United Kingdom
| | - Giorgia Pastorin
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Driton Vllasaliu
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 9NH, United Kingdom.
| |
Collapse
|
26
|
Bravo Iniguez A, Du M, Zhu MJ. α-Ketoglutarate for Preventing and Managing Intestinal Epithelial Dysfunction. Adv Nutr 2024; 15:100200. [PMID: 38438107 PMCID: PMC11016550 DOI: 10.1016/j.advnut.2024.100200] [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: 11/22/2023] [Revised: 02/16/2024] [Accepted: 02/29/2024] [Indexed: 03/06/2024] Open
Abstract
The epithelium lining the intestinal tract serves a multifaceted role. It plays a crucial role in nutrient absorption and immune regulation and also acts as a protective barrier, separating underlying tissues from the gut lumen content. Disruptions in the delicate balance of the gut epithelium trigger inflammatory responses, aggravate conditions such as inflammatory bowel disease, and potentially lead to more severe complications such as colorectal cancer. Maintaining intestinal epithelial homeostasis is vital for overall health, and there is growing interest in identifying nutraceuticals that can strengthen the intestinal epithelium. α-Ketoglutarate, a metabolite of the tricarboxylic acid cycle, displays a variety of bioactive effects, including functioning as an antioxidant, a necessary cofactor for epigenetic modification, and exerting anti-inflammatory effects. This article presents a comprehensive overview of studies investigating the potential of α-ketoglutarate supplementation in preventing dysfunction of the intestinal epithelium.
Collapse
Affiliation(s)
| | - Min Du
- Department of Animal Sciences, Washington State University, Pullman, WA, United States
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA, United States.
| |
Collapse
|
27
|
Li Z, He H, Chen M, Ni M, Guo C, Wan Z, Zhou J, Wang Z, Wang Y, Cai H, Li M, Sun H, Xu H. Novel mechanism of Clostridium butyricum alleviated coprophagy prevention-induced intestinal inflammation in rabbit. Int Immunopharmacol 2024; 130:111773. [PMID: 38430808 DOI: 10.1016/j.intimp.2024.111773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
As bacteria synthesize nutrients primarily in the cecum, coprophagy is indispensable for supplying rabbits with essential nutrients. Recent research has demonstrated its pivotal role in maintaining intestinal microbiota homeostasis and immune regulation in rabbits, although the specific mechanism remains unknown. Here, we used coprophagy prevention (CP) to investigate the effects of coprophagy on the cecum homeostasis and microbiota in New Zealand white rabbits. Furthermore, whether supplementation of Clostridium butyricum (C. butyricum) may alleviate the cecum inflammation and apoptosis caused by CP was also explored. Four groups were randomly assigned: control (Con), sham-coprophagy prevention (SCP), coprophagy prevention (CP), and CP and C. butyricum addition (CPCB). Compared to Con and SCP, CP augmented cecum inflammation and apoptosis, as well as bacterial adhesion to the cecal epithelial mucosa, while decreasing the expression of tight junction proteins (ZO-1, occluding, and claudin-1). The relative abundance of short-chain fatty acids (SCFAs)-producing bacteria was significantly decreased in the CP group. Inversely, there was an increase in the Firmicutes/Bacteroidetes ratio and the relative abundance of Christensenellaceae_R-7_group. Additionally, CP increased the levels of Flagellin, IFN-γ, TNF-a, and IL-1β in cecum contents and promoted the expression of TLR5/MyD88/NF-κB pathway in cecum tissues. However, the CPCB group showed significant improvements in all parameters compared to the CP group. Dietary C. butyricum supplementation significantly increased the production of SCFAs, particularly butyric acid, triggering anti-inflammatory, tissue repairing, and barrier-protective responses. Notably, CPCB effectively mitigated CP-induced apoptosis and inflammation. In summary, CP disrupts the cecum epithelial barrier and induces inflammation in New Zealand white rabbits, but these effects can be alleviated by C. butyricum supplementation. This process appears to be largely associated with the TLR5/MyD88/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Zhichao Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, PR China
| | - Hui He
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, PR China
| | - Mengjuan Chen
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, PR China
| | - Mengke Ni
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, PR China
| | - Chaohui Guo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, PR China
| | - Zhiyi Wan
- College of Biological Sciences, China Agricultural University, No.2 Yuan Ming Yuan West Road, Beijing 100193, PR China
| | - Jianshe Zhou
- Institute of Fisheries Science, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa 850032, PR China
| | - Zhitong Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, PR China
| | - Yaling Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, PR China
| | - Hanfang Cai
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, PR China
| | - Ming Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, PR China.
| | - HuiZeng Sun
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, PR China.
| | - Huifen Xu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, PR China.
| |
Collapse
|
28
|
López-Posadas R, Bagley DC, Pardo-Pastor C, Ortiz-Zapater E. The epithelium takes the stage in asthma and inflammatory bowel diseases. Front Cell Dev Biol 2024; 12:1258859. [PMID: 38529406 PMCID: PMC10961468 DOI: 10.3389/fcell.2024.1258859] [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: 07/14/2023] [Accepted: 02/22/2024] [Indexed: 03/27/2024] Open
Abstract
The epithelium is a dynamic barrier and the damage to this epithelial layer governs a variety of complex mechanisms involving not only epithelial cells but all resident tissue constituents, including immune and stroma cells. Traditionally, diseases characterized by a damaged epithelium have been considered "immunological diseases," and research efforts aimed at preventing and treating these diseases have primarily focused on immuno-centric therapeutic strategies, that often fail to halt or reverse the natural progression of the disease. In this review, we intend to focus on specific mechanisms driven by the epithelium that ensure barrier function. We will bring asthma and Inflammatory Bowel Diseases into the spotlight, as we believe that these two diseases serve as pertinent examples of epithelium derived pathologies. Finally, we will argue how targeting the epithelium is emerging as a novel therapeutic strategy that holds promise for addressing these chronic diseases.
Collapse
Affiliation(s)
- Rocío López-Posadas
- Department of Medicine 1, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-Universtiy Eralngen-Nürnberg, Erlangen, Germany
| | - Dustin C. Bagley
- Randall Centre for Cell and Molecular Biophysics, New Hunt’s House, School of Basic and Medical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Carlos Pardo-Pastor
- Randall Centre for Cell and Molecular Biophysics, New Hunt’s House, School of Basic and Medical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Elena Ortiz-Zapater
- Department of Biochemistry and Molecular Biology, Universitat de Valencia, Valencia, Spain
- Instituto Investigación Hospital Clínico-INCLIVA, Valencia, Spain
| |
Collapse
|
29
|
Wang L, Li M, Gu Y, Shi J, Yan J, Wang X, Li B, Wang B, Zhong W, Cao H. Dietary flavonoids-microbiota crosstalk in intestinal inflammation and carcinogenesis. J Nutr Biochem 2024; 125:109494. [PMID: 37866426 DOI: 10.1016/j.jnutbio.2023.109494] [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: 09/27/2022] [Revised: 02/20/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
Colorectal cancer (CRC) is currently the third leading cancer and commonly develops from chronic intestinal inflammation. A strong association was found between gut microbiota and intestinal inflammation and carcinogenic risk. Flavonoids, which are abundant in vegetables and fruits, can inhibit inflammation, regulate gut microbiota, protect gut barrier integrity, and modulate immune cell function, thereby attenuating colitis and preventing carcinogenesis. Upon digestion, about 90% of flavonoids are transported to the colon without being absorbed in the small intestine. This phenomenon increases the abundance of beneficial bacteria and enhances the production of short-chain fatty acids. The gut microbe further metabolizes these flavonoids. Interestingly, some metabolites of flavonoids play crucial roles in anti-inflammation and anti-tumor effects. This review summarizes the modulatory effect of flavonoids on gut microbiota and their metabolism by intestinal microbe under disease conditions, including inflammatory bowel disease, colitis-associated cancer (CAC), and CRC. We focus on dietary flavonoids and microbial interactions in intestinal mucosal barriers as well as intestinal immune cells. Results provide novel insights to better understand the crosstalk between dietary flavonoids and gut microbiota and support the standpoint that dietary flavonoids prevent intestinal inflammation and carcinogenesis.
Collapse
Affiliation(s)
- Lei Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China; Department of Gastroenterology and Hepatology, The Affiliated Hospital of Chengde Medical College, Hebei, China
| | - Mengfan Li
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Yu Gu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Junli Shi
- Department of Gastroenterology and Hepatology, The Affiliated Hospital of Chengde Medical College, Hebei, China
| | - Jing Yan
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China; Department of Nutrition, the Second Affiliated Hospital, Air Force Medical University, Xi'an, China
| | - Xin Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Bingqing Li
- Department of Gastroenterology and Hepatology, The Affiliated Hospital of Chengde Medical College, Hebei, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Weilong Zhong
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| |
Collapse
|
30
|
Gudi R, Johnson BM, Gaudreau MC, Sun W, Ball L, Vasu C. Intestinal permeability and inflammatory features of juvenile age correlate with the eventual systemic autoimmunity in lupus-prone female SWR × NZB F1 (SNF1) mice. Immunology 2024; 171:235-249. [PMID: 37947218 PMCID: PMC10842200 DOI: 10.1111/imm.13713] [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: 04/24/2023] [Accepted: 10/29/2023] [Indexed: 11/12/2023] Open
Abstract
The incidence of systemic lupus erythematosus (SLE) is about nine times higher in women than in men, and the underlying mechanisms that contribute to this gender bias are not fully understood. Previously, using lupus-prone (SWR × NZB)F1 (SNF1) mice, we have shown that the intestinal immune system could play a role in the initiation and progression of disease in SLE, and depletion of gut microbiota produces more pronounced disease protection in females than in males. Here, we show that the gut permeability features of lupus-prone female SNF1 mice at juvenile ages directly correlate with the expression levels of pro-inflammatory factors, faecal IgA abundance and nAg reactivity and the eventual systemic autoantibody levels and proteinuria onset. Furthermore, we observed that the disease protection achieved in female SNF1 mice upon depletion of gut microbiota correlates with the diminished gut inflammatory protein levels, intestinal permeability and circulating microbial DNA levels. However, faecal microbiota transplant from juvenile male and females did not result in modulation of gut inflammatory features or permeability. Overall, these observations suggest that the early onset of intestinal inflammation, systemic autoantibody production and clinical stage disease in lupus-prone females is linked to higher gut permeability in them starting at as early as juvenile age. While the higher gut permeability in juvenile lupus-prone females is dependent on the presence of gut microbes, it appears to be independent of the composition of gut microbiota.
Collapse
Affiliation(s)
- Radhika Gudi
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, SC-29425
| | - Benjamin M. Johnson
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, SC-29425
| | - Marie-Claude Gaudreau
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, SC-29425
| | - Wei Sun
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, SC-29425
| | - Lauren Ball
- Department of Pharmacology, College of Medicine, Medical University of South Carolina, Charleston, SC-29425
| | - Chenthamarakshan Vasu
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, SC-29425
| |
Collapse
|
31
|
Saha MR, Dey P. Pharmacological benefits of Acacia against metabolic diseases: intestinal-level bioactivities and favorable modulation of gut microbiota. Arch Physiol Biochem 2024; 130:70-86. [PMID: 34411504 DOI: 10.1080/13813455.2021.1966475] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 08/05/2021] [Indexed: 10/20/2022]
Abstract
CONTEXT Obesity-associated chronic metabolic disease is a leading contributor to mortality globally. Plants belonging to the genera Acacia are routinely used for the treatment of diverse metabolic diseases under different ethnomedicinal practices around the globe. OBJECTIVE The current review centres around the pharmacological evidence of intestinal-level mechanisms for metabolic health benefits by Acacia spp. RESULTS Acacia spp. increase the proportions of gut commensals (Bifidobacterium and Lactobacillus) and reduces the population of opportunistic pathobionts (Escherichia coli and Clostridium). Acacia gum that is rich in fibre, can also be a source of prebiotics to improve gut health. The intestinal-level anti-inflammatory activities of Acacia are likely to contribute to improvements in gut barrier function that would prevent gut-to-systemic endotoxin translocation and limit "low-grade" inflammation associated with metabolic diseases. CONCLUSION This comprehensive review for the first time has emphasised the intestinal-level benefits of Acacia spp. which could be instrumental in limiting the burden of metabolic disease.
Collapse
Affiliation(s)
- Manas Ranjan Saha
- Department of Life Science, Vidyasagar Primary Teachers Training Institute (B.Ed.), Malda, India
| | - Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| |
Collapse
|
32
|
Mukherjee T, Kumar N, Chawla M, Philpott DJ, Basak S. The NF-κB signaling system in the immunopathogenesis of inflammatory bowel disease. Sci Signal 2024; 17:eadh1641. [PMID: 38194476 DOI: 10.1126/scisignal.adh1641] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 12/11/2023] [Indexed: 01/11/2024]
Abstract
Inflammatory bowel disease (IBD) is an idiopathic, chronic condition characterized by episodes of inflammation in the gastrointestinal tract. The nuclear factor κB (NF-κB) system describes a family of dimeric transcription factors. Canonical NF-κB signaling is stimulated by and enhances inflammation, whereas noncanonical NF-κB signaling contributes to immune organogenesis. Dysregulation of NF-κB factors drives various inflammatory pathologies, including IBD. Signals from many immune sensors activate NF-κB subunits in the intestine, which maintain an equilibrium between local microbiota and host responses. Genetic association studies of patients with IBD and preclinical mouse models confirm the importance of the NF-κB system in host defense in the gut. Other studies have investigated the roles of these factors in intestinal barrier function and in inflammatory gut pathologies associated with IBD. NF-κB signaling modulates innate and adaptive immune responses and the production of immunoregulatory proteins, anti-inflammatory cytokines, antimicrobial peptides, and other tolerogenic factors in the intestine. Furthermore, genetic studies have revealed critical cell type-specific roles for NF-κB proteins in intestinal immune homeostasis, inflammation, and restitution that contribute to the etiopathology of IBD-associated manifestations. Here, we summarize our knowledge of the roles of these NF-κB pathways, which are activated in different intestinal cell types by specific ligands, and their cross-talk, in fueling aberrant intestinal inflammation. We argue that an in-depth understanding of aberrant immune signaling mechanisms may hold the key to identifying predictive or prognostic biomarkers and developing better therapeutics against inflammatory gut pathologies.
Collapse
Affiliation(s)
- Tapas Mukherjee
- Systems Immunology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Naveen Kumar
- Systems Immunology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Meenakshi Chawla
- Systems Immunology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Soumen Basak
- Systems Immunology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| |
Collapse
|
33
|
Wang X, Wang J, Okyere SK, Huang R, Shao C, Yousif M, Deng J, Hu Y. Ageratina adenophora damages the rumen epithelium via inducing the expression of inflammatory factors in goats. J Anim Sci 2024; 102:skad418. [PMID: 38142130 PMCID: PMC10781442 DOI: 10.1093/jas/skad418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 12/21/2023] [Indexed: 12/25/2023] Open
Abstract
The aim of this experiment was to investigate the effects of Ageratina adenophora on the expression of epithelium tight junction proteins and inflammatory factors in the rumen of goats. Twelve goats were randomly divided into three groups. The first group was the blank control group (n = 3, C) which was fed normal diet. The second group was fistulas control group (n = 3, RFC), which was fitted with rumen fistulas, and fed normal diet. The third group was the A. adenophora test group (n = 6, AA), which was fitted with rumen fistulas and fed a mixture of 60% of normal diet and 40% of A. adenophora grass powder. The feeding experiment lasted for 90 d, after which all goats were sacrificed and samples were collected from the rumen dorsal sac and ventral sac. The relative expression of mRNA of inflammatory factors in the rumen epithelium (tumor necrosis factor alpha [TNF-α], interferon gamma [IFN-γ], interleukin 1 beta [IL-1β], IL-2, IL-4, IL-6, and IL-10) and tight junction protein genes (occludin, claudin-1, and ZO-1) was measured by quantitative real-time fluorescence PCR. Expression of tight junction proteins in the rumen epithelium was measured by Western blot. A correlation was established between the expression of inflammatory factors and tight junction protein genes using Graph Pad Prism. The results showed that A. adenophora caused a significant increase in the mRNA expression levels of TNF-α, IFN-γ, IL-1β, IL-2, IL-6, and IL-10 in the rumen epithelial (P < 0.05 or P < 0.01). The expression of tight junction proteins at both gene and protein levels was significantly decreased (P < 0.05 or P < 0.01). Furthermore, the correlation analysis revealed that the changes in tight junction protein expression in the test group were closely related to the upregulation of the expression of inflammatory factors TNF-α and IFN-γ in rumen epithelial cells. In conclusion, the expression of inflammatory factors was increased and the expression of tight junction proteins was decreased in goats after feeding on A. adenophora, which caused some damage to the rumen epithelium.
Collapse
Affiliation(s)
- Xiaoxuan Wang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, People’s Republic of China
| | - Jianchen Wang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, People’s Republic of China
| | - Samuel Kumi Okyere
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, People’s Republic of China
- Department of Pharmaceutical Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Ruya Huang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, People’s Republic of China
| | - Chenyang Shao
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, People’s Republic of China
| | - Muhammad Yousif
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, People’s Republic of China
| | - Junliang Deng
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, People’s Republic of China
| | - Yanchun Hu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, People’s Republic of China
| |
Collapse
|
34
|
Kobayashi K, Mochizuki J, Yamazaki F, Sashihara T. Yogurt starter strains ameliorate intestinal barrier dysfunction via activating AMPK in Caco-2 cells. Tissue Barriers 2024; 12:2184157. [PMID: 36852963 PMCID: PMC10832913 DOI: 10.1080/21688370.2023.2184157] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 02/19/2023] [Indexed: 03/01/2023] Open
Abstract
Lactic acid bacteria (LAB) are commonly used probiotics that improve human health in various aspects. We previously reported that yogurt starter strains, Lactobacillus delbrueckii subsp. bulgaricus 2038 and Streptococcus thermophilus 1131, potentially enhance the intestinal epithelial barrier function by inducing the expression of antimicrobial peptides in the small intestine. However, their effects on physical barrier functions remain unknown. In this study, we found that both strains ameliorated the decreased trans-epithelial resistance and the increased permeability of fluorescein isothiocyanate-dextran induced by tumor necrosis factor (TNF)-α and interferon (IFN)-γ in Caco-2 cells. We also demonstrated that LAB prevented a decrease in the expression and disassembly of tight junctions (TJs) induced by TNF-α and IFN-γ. To assess the repair activity of TJs, a calcium switch assay was performed. Both strains were found to promote the reassembly of TJs, and their activity was canceled by the inhibitor of AMP-activated protein kinase (AMPK). Moreover, these strains showed increased AMPK phosphorylation. These observations suggest that the strains ameliorated physical barrier dysfunction via the activation of AMPK. The activities preventing barrier destruction induced by TNF-α and IFN-γ were strain-dependent. Several strains containing L. bulgaricus 2038 and S. thermophilus 1131 significantly suppressed the barrier impairment, and L. bulgaricus 2038 showed the strongest activity among them. Our findings suggest that the intake of L. bulgaricus 2038 and S. thermophilus 1131 is a potential strategy for the prevention and repair of leaky gut.
Collapse
Affiliation(s)
- Kyosuke Kobayashi
- Food Microbiology and Function Research Laboratories, R&D Division, Meiji Co., Ltd, Tokyo, Japan
| | - Junko Mochizuki
- Food Microbiology and Function Research Laboratories, R&D Division, Meiji Co., Ltd, Tokyo, Japan
| | - Fuka Yamazaki
- Food Microbiology and Function Research Laboratories, R&D Division, Meiji Co., Ltd, Tokyo, Japan
| | - Toshihiro Sashihara
- Food Microbiology and Function Research Laboratories, R&D Division, Meiji Co., Ltd, Tokyo, Japan
| |
Collapse
|
35
|
Alake SE, Ice J, Robinson K, Price P, Hatter B, Wozniak K, Lin D, Chowanadisai W, Smith BJ, Lucas EA. Reduced estrogen signaling contributes to bone loss and cardiac dysfunction in interleukin-10 knockout mice. Physiol Rep 2024; 12:e15914. [PMID: 38217044 PMCID: PMC10787104 DOI: 10.14814/phy2.15914] [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: 08/28/2023] [Revised: 12/02/2023] [Accepted: 12/07/2023] [Indexed: 01/14/2024] Open
Abstract
Characterization of the interleukin (IL)-10 knockout (KO) mouse with chronic gut inflammation, cardiovascular dysfunction, and bone loss suggests a critical role for this cytokine in interorgan communication within the gut, bone, and cardiovascular axis. We sought to understand the role of IL-10 in the cross-talk between these systems. Six-week-old IL-10 KO mice and their wild type (WT) counterparts were maintained on a standard rodent diet for 3 or 6 months. Gene expression of proinflammatory markers and Fgf23, serum 17β-estradiol (E2), and cardiac protein expression were assessed. Ileal Il17a and Tnf mRNA increased while Il6 mRNA increased in the bone and heart by at least 2-fold in IL-10 KO mice. Bone Dmp1 and Phex mRNA were repressed at 6 months in IL-10 KO mice, resulting in increased Fgf23 mRNA (~4-fold) that contributed to increased fibrosis. In the IL-10 KO mice, gut bacterial β-glucuronidase activity and ovarian Cyp19a1 mRNA were lower (p < 0.05), consistent with reduced serum E2 and reduced cardiac pNOS3 (Ser1119 ) in these mice. Treatment of ileal lymphocytes with E2 reduced gut inflammation in WT but not IL-10 KO mice. In conclusion, our data suggest that diminished estrogen and defective bone mineralization increased FGF23 which contributed to cardiac fibrosis in the IL-10 KO mouse.
Collapse
Affiliation(s)
- Sanmi E. Alake
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - John Ice
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Kara Robinson
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Payton Price
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Bethany Hatter
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Karen Wozniak
- Department of Microbiology and Molecular GeneticsOklahoma State UniversityStillwaterOklahomaUSA
| | - Dingbo Lin
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Winyoo Chowanadisai
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Brenda J. Smith
- Department of Obstetrics and GynecologyIndiana School of MedicineIndianapolisIndianaUSA
- Indiana Center for Musculoskeletal HealthIndiana School of MedicineIndianapolisIndianaUSA
| | - Edralin A. Lucas
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| |
Collapse
|
36
|
Melo-Marques I, Cardoso SM, Empadinhas N. Bacterial extracellular vesicles at the interface of gut microbiota and immunity. Gut Microbes 2024; 16:2396494. [PMID: 39340209 PMCID: PMC11444517 DOI: 10.1080/19490976.2024.2396494] [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: 02/08/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
Bacterial extracellular vesicles (BEVs) are nano-sized lipid-shielded structures released by bacteria and that play an important role in intercellular communication. Their broad taxonomic origins and varying cargo compositions suggest their active participation in significant biological mechanisms. Specifically, they are involved in directly modulating microbial ecosystems, competing with other organisms, contributing to pathogenicity, and influencing the immunity of their hosts. This review examines the mechanisms that underlie the modulatory effects of BEVs on gut dynamics and immunity. Understanding how BEVs modulate microbiota composition and functional imbalances is crucial, as gut dysbiosis is implicated not only in the pathogenesis of various gastrointestinal, metabolic, and neurological diseases, but also in reducing resistance to colonization by enteric pathogens, which is particularly concerning given the current antimicrobial resistance crisis. This review summarizes recent advancements in the field of BEVs to encourage further research into these enigmatic entities. This will facilitate a better understanding of intra- and interkingdom communication phenomena and reveal promising therapeutic approaches.
Collapse
Affiliation(s)
- Inês Melo-Marques
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Sandra Morais Cardoso
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Nuno Empadinhas
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
37
|
Li Y, Li W, Zeng Z, Han Y, Chen Q, Dong X, Wang Z, Feng S, Cao W. Lasso peptide MccY alleviates non-typhoidal salmonellae-induced mouse gut inflammation via regulation of intestinal barrier function and gut microbiota. Microbiol Spectr 2023; 11:e0178423. [PMID: 37819128 PMCID: PMC10714986 DOI: 10.1128/spectrum.01784-23] [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: 05/05/2023] [Accepted: 08/28/2023] [Indexed: 10/13/2023] Open
Abstract
IMPORTANCE Diseases caused by Enterobacteriaceae multidrug-resistant strains have become increasingly difficult to manage. It is necessary to verify the new antibacterial drug MccY effect on non-typhoid Salmonella infection in mice since it is regarded as a promising microcin. The results demonstrated that MccY has a potential therapeutic application value in the protection against Salmonella-induced intestinal damage and alleviating related intestinal dysbiosis and metabolic disorders. MccY could be a promising candidate as an antimicrobial or anti-inflammatory agent for treating infectious diseases.
Collapse
Affiliation(s)
- Yu Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Wenjing Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zhiwei Zeng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yu Han
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Qinxi Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xinyi Dong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zepeng Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Saixiang Feng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangdong, China
- Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, Guangzhou, China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China
| | - Weisheng Cao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangdong, China
- Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, Guangzhou, China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China
| |
Collapse
|
38
|
Jiang XS, Fu BL, Yang XX, Qin HY. TNF-α Mediated the Disruption of Hepatic Tight Junction Expression in Blood-Biliary Barrier of Colitis via Downregulating PI3K/AKT Signaling Pathway. Biol Pharm Bull 2023; 46:1769-1777. [PMID: 37899248 DOI: 10.1248/bpb.b23-00503] [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] [Indexed: 10/31/2023]
Abstract
Hepatocyte tight junctions (TJ) constituted blood-biliary barrier is the most important hepatic barrier for separating bile from the bloodstream, disruption or dysfunction of TJ barrier is involved in hepatobiliary manifestations of colitis, but the underlying mechanism is still not clear. This study aims to investigate the effect and underlying mechanism of tumor necrosis factor alpha (TNF-α) on hepatic TJ protein expression in blood-biliary barrier and identify its role in the pathogenesis of acute colitis-related cholestasis. Acute colitis rat model was induced by trinitrobenzene sulfonic acid (TNBS) intra-colonic administration. TJs expression of blood-biliary barrier was tested in colitis rats, the serum TNF-α level was also determined in order to elucidate the correlation of TNF-α and TJs. HepaRG cells were used to investigate the effect of TNF-α on TJs, and the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) signaling pathway were also evaluated in rats and TNF-α treated HepaRG cells. Acute colitis was induced in rats at 5 d post TNBS, which is accompanied with cholestasis-like alteration. Serum TNF-α level was increased in colitis rats and positively correlated with the alteration of total bile acids and bilirubin, marked decrease in TJs was found in TNF-α treated HepaRG cells and the rats, down-regulated PI3K/AKT signaling pathway were also identified in TNF-α treated HepaRG cells and the rats. The study concluded that serum TNF-α mediated the down-regulation of PI3K/AKT signaling pathway, which contributed to the reduction of TJ protein expression in acute colitis-related intrahepatic cholestasis. These findings suggest that TNF-α plays an important role in the pathogenesis of intrahepatic cholestasis of colitis.
Collapse
Affiliation(s)
| | - Bi-le Fu
- The First Clinical Medical College, Lanzhou University
- College of Pharmacy, Lanzhou University
| | - Xin-Xin Yang
- The First Clinical Medical College, Lanzhou University
| | - Hong-Yan Qin
- Department of Pharmacy, First Hospital of Lanzhou University
| |
Collapse
|
39
|
Güney Z, Kurgan Ş, Önder C, Serdar MA, Günhan Ö, Günhan M. Expression of tight junction proteins in smokers and non-smokers with generalized Stage III periodontitis. J Periodontal Res 2023; 58:1281-1289. [PMID: 37697913 DOI: 10.1111/jre.13184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 08/22/2023] [Accepted: 09/01/2023] [Indexed: 09/13/2023]
Abstract
OBJECTIVE This study aims to evaluate the gingival crevicular fluid (GCF) levels of tumor necrosis factor-α (TNF-α), zonula occludens-1 (ZO-1), occludin (Occ), and tricellulin (Tric) in periodontitis, as well as their alterations due to smoking. BACKGROUND Tight junctions (TJ), which consist of transmembrane and cytoplasmic scaffolding proteins, connect the epithelial cells of the periodontium. Occ, claudins, junctional adhesion molecules, and Tric are transmembrane TJ proteins found at the cell membrane. The transmembrane TJ proteins and the intracellular cytoskeleton are directly linked by cytoplasmic scaffolding proteins such as ZO-1. Although the functions and locations of these molecules have been defined, their behavior in periodontal inflammation is unknown. METHODS The study included four groups: individuals with periodontal health without smoking (C; n = 31), individuals with generalized Stage III periodontitis without smoking (P; n = 28), individuals with periodontal health while smoking (CS; n = 22), and individuals with generalized Stage III periodontitis while smoking (PS; n = 18). Clinical periodontal parameters were recorded, and enzyme-linked immunosorbent assay (ELISA) was used to examine ZO-1, Occ, Tric, and TNF-α levels in GCF. RESULTS In the periodontitis groups, clinical parameters were significantly higher (p < .001). The site-specific levels of TNF-α, ZO-1, Tric, and Occ in the P group were statistically higher than those in the other groups (p < .05). TNF-α, probing pocket depth (PPD), and bleeding on probing (BOP) exhibited positive correlations with all TJ proteins (p < .005). CONCLUSIONS Smoking could potentially affect the levels of epithelial TJ proteins in the GCF, thereby potentially playing a significant role in the pathogenesis of the periodontal disease.
Collapse
Affiliation(s)
- Zeliha Güney
- Department of Periodontology, Faculty of Dentistry, Ankara University, Ankara, Turkey
- Department of Periodontology, Faculty of Dentistry, Ankara Medipol University, Ankara, Turkey
| | - Şivge Kurgan
- Department of Periodontology, Faculty of Dentistry, Ankara University, Ankara, Turkey
| | - Canan Önder
- Department of Periodontology, Faculty of Dentistry, Ankara University, Ankara, Turkey
| | - Muhittin A Serdar
- Department of Medical Biochemistry, School of Medicine, Acibadem University, İstanbul, Turkey
| | - Ömer Günhan
- Department of Pathology, School of Medicine, TOBB ETÜ University, Ankara, Turkey
| | - Meral Günhan
- Department of Periodontology, Faculty of Dentistry, Ankara University, Ankara, Turkey
| |
Collapse
|
40
|
Bai Y, Miyanaga K, Yamamoto N. Enhanced tight junction in Caco-2 cells by the pretreatment with Lactobacillus johnsonii strain MG. Biosci Biotechnol Biochem 2023; 87:1532-1536. [PMID: 37704400 DOI: 10.1093/bbb/zbad131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/21/2023] [Indexed: 09/15/2023]
Abstract
We investigated roles of Lactobacillus johnsonii MG (MG) isolated from mice with interaction with tight junction on gut barrier function with Caco-2 cell model. Pretreatment with MG enhanced barrier function and showed protective effect against Enterococcus faecium provided damage. MG treatment increased the gene expressions of transcriptional regulator NFKB and major tight junction protein, ZO-1.
Collapse
Affiliation(s)
- Yuying Bai
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Kazuhiko Miyanaga
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
- Department of Infection and Immunity, School of Medicine, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke-Shi, Tochigi, Japan
| | - Naoyuki Yamamoto
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| |
Collapse
|
41
|
Xia W, Li S, Li L, Zhang S, Wang X, Ding W, Ding L, Zhang X, Wang Z. Role of anthraquinones in combating insulin resistance. Front Pharmacol 2023; 14:1275430. [PMID: 38053837 PMCID: PMC10694622 DOI: 10.3389/fphar.2023.1275430] [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: 08/10/2023] [Accepted: 11/06/2023] [Indexed: 12/07/2023] Open
Abstract
Insulin resistance presents a formidable public health challenge that is intricately linked to the onset and progression of various chronic ailments, including diabetes, cardiovascular disease, hypertension, metabolic syndrome, nonalcoholic fatty liver disease, and cancer. Effectively addressing insulin resistance is paramount in preventing and managing these metabolic disorders. Natural herbal remedies show promise in combating insulin resistance, with anthraquinone extracts garnering attention for their role in enhancing insulin sensitivity and treating diabetes. Anthraquinones are believed to ameliorate insulin resistance through diverse pathways, encompassing activation of the AMP-activated protein kinase (AMPK) signaling pathway, restoration of insulin signal transduction, attenuation of inflammatory pathways, and modulation of gut microbiota. This comprehensive review aims to consolidate the potential anthraquinone compounds that exert beneficial effects on insulin resistance, elucidating the underlying mechanisms responsible for their therapeutic impact. The evidence discussed in this review points toward the potential utilization of anthraquinones as a promising therapeutic strategy to combat insulin resistance and its associated metabolic diseases.
Collapse
Affiliation(s)
- Wanru Xia
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shuqian Li
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - LinZehao Li
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shibo Zhang
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaolei Wang
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Wenyu Ding
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Lina Ding
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiandang Zhang
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Zhibin Wang
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
42
|
Zheng W, Guan Y, Wu B. Effects of Yupingfeng Polysaccharides as Feed Supplement on Immune Function and Intestinal Microbiome in Chickens. Microorganisms 2023; 11:2774. [PMID: 38004785 PMCID: PMC10672924 DOI: 10.3390/microorganisms11112774] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/01/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
The health of chicks is closely related to their productivity. Yupingfeng polysaccharide (YPF-P) is a kind of water-soluble polysaccharide extracted from Yupingfeng powder; it has high pharmacological activity and can be used as a potential substitute for antibiotics to improve the health of chicks. This study aimed to investigate the effects of YPF-P on immune performance, the duodenum, and the cecal microflora of chicks. All chickens (4224) were randomly distributed into four groups (eight replicas/group, 132 hens/replica). The control group was fed a basal diet (0 g/kg YPF-P), while the experimental groups were fed basal diets supplemented with 1, 2, or 4 g/kg YPF-P. The results showed that YPF-P significantly increased the thymus index (p < 0.05). The content of total antioxidant capacity (T-AOC), superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), immunoglobulin A (IgA), and IgG and immunoglobulin M (IgM) was upregulated in the serum by YPF-P (p < 0.05). YPF-P decreased the content of malondialdehyde (MDA) (p < 0.05). Further, 16S rRNA sequencing showed that 2 g/kg YPF-P modulated the predominant duodenum and cecal microbial community structure, which increased the number of Faecalibacterium, Megamonas, Bacteroides, Alistipes, NK4A214_group, and Enterococcus. In conclusion, YPF-P ameliorated the growth performance of chicks by regulating serum immune and antioxidant balance, as well as the intestinal microbiota.
Collapse
Affiliation(s)
| | | | - Bo Wu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan 528000, China
| |
Collapse
|
43
|
Li Z, Han C, Wang Z, Li Z, Ruan L, Lin H, Zhou C. Black soldier fly pulp in the diet of golden pompano: Effect on growth performance, liver antioxidant and intestinal health. FISH & SHELLFISH IMMUNOLOGY 2023; 142:109156. [PMID: 37827247 DOI: 10.1016/j.fsi.2023.109156] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/01/2023] [Accepted: 10/10/2023] [Indexed: 10/14/2023]
Abstract
Black soldier fly (Hermetia illucens) has been widely researched as a protein source for fish meal replacement in aquaculture, but few studies have focused on its potential as a feed additive for growth and immune enhancement. We conducted a 56-day culture experiment to determine the impact of feed addition of black soldier fly pulp (BSFP, with 86.2% small peptides in dry basis) on growth performance, plasma biochemistry, liver antioxidant levels, intestinal immunity, digestion and microbiota of juvenile golden pompano (Trachinotus ovatus, 5.63 ± 0.02 g). BSFP was added to the basal diet at 0%, 1%, 3%, 5%, 7% and 9% (named Control, BSFP-1, BSFP-3, BSFP-5, BSFP-7, BSFP-9), respectively. BSFP increased the weight gain rate, specific growth rate, protein efficiency ratio and reduced the feed conversion rate of juvenile T. ovatus, the optimal growth performance was reached at BSFP-1, after which a negative feedback phenomenon was observed. Low levels of BSFP upregulated the expression of hepatic antioxidant, intestinal tight junctions, anti-inflammatory related genes and enhanced antioxidant, immune and intestinal digestive enzyme activities, which simultaneously reduced hepatic malondialdehyde and plasma aspartate transaminase and alanine aminotransferase concentrations. However, at BSFP-7, catalase activity was significantly reduced, while NF-κB p65 and pro-inflammatory cytokines transcription was significantly enhanced (P < 0.05). The results suggest that high doses of BSFP addition may damage fish health by inhibiting small peptide uptake, decreasing the activity of antioxidant enzyme and activating the canonical NF-κB pathway. Conversely, low doses of BSFP enhanced intestinal tight junction protein transcription, digestive enzyme activity and immune performance, inhibited pathogenic microbiota, while enhancing liver antioxidant capacity, which was associated with activated Nrf2-Keap1 pathway and suppressed NF-κB pathway, showing its potential as a feed additive to aquafeeds.
Collapse
Affiliation(s)
- Zuzhe Li
- Key Laboratory of Aquatic Product Processing, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, China; College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Chengzong Han
- Key Laboratory of Aquatic Product Processing, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, China; College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Zhanzhan Wang
- Key Laboratory of Aquatic Product Processing, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, China
| | - Zhenyu Li
- Guangdong Green Coral Bio-Technology Co., Ltd, Dongguan, 523000, China
| | - Leshan Ruan
- Key Laboratory of Aquatic Product Processing, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, China; College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Heizhao Lin
- Shenzhen Base of South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shenzhen, 518121, China; Key Laboratory of Efficient Utilization and Processing of Marine Fishery Resources of Hainan Province, Sanya, 572019, China
| | - Chuanpeng Zhou
- Key Laboratory of Aquatic Product Processing, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, China; Key Laboratory of Efficient Utilization and Processing of Marine Fishery Resources of Hainan Province, Sanya, 572019, China.
| |
Collapse
|
44
|
Mohebali N, Weigel M, Hain T, Sütel M, Bull J, Kreikemeyer B, Breitrück A. Faecalibacterium prausnitzii, Bacteroides faecis and Roseburia intestinalis attenuate clinical symptoms of experimental colitis by regulating Treg/Th17 cell balance and intestinal barrier integrity. Biomed Pharmacother 2023; 167:115568. [PMID: 37793274 DOI: 10.1016/j.biopha.2023.115568] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 10/06/2023] Open
Abstract
Ulcerative colitis (UC) is a severe inflammatory bowel disease (IBD) characterized by multifactorial complex disorders triggered by environmental factors, genetic susceptibility, and also gut microbial dysbiosis. Faecalibacterium prausnitzii, Bacteroides faecis, and Roseburia intestinalis are underrepresented species in UC patients, leading to the hypothesis that therapeutic application of those bacteria could ameliorate clinical symptoms and disease severity. Acute colitis was induced in mice by 3.5% DSS, and the commensal bacterial species were administered by oral gavage simultaneously with DSS treatment for up to 7 days. The signs of colonic inflammation, the intestinal barrier integrity, the proportion of regulatory T cells (Tregs), and the expression of pro-inflammatory and anti-inflammatory cytokines were quantified. The concentrations of SCFAs in feces were measured using Gas-liquid chromatography. The gut microbiome was analyzed in all treatment groups at the endpoint of the experiment. Results were benchmarked against a contemporary mesalazine treatment regime. We show that commensal species alone and in combination reduced disease activity index scores, inhibited colon shortening, strengthened the colonic epithelial barrier, and positively modulated tight junction protein expression. The expression level of pro-inflammatory cytokines was significantly reduced. Immune modulation occurred via inhibition of the loss of CD4 +CD25 +Treg cells in the spleen. Our study proofed that therapeutic application of F. prausnitzii, B. faecis, and R. intestinalis significantly ameliorated DSS-induced colitis at the level of clinical symptoms, histological inflammation, and immune status. Our data suggest that these positive effects are mediated by immune-modulatory pathways and influence on Treg/Th17 balance.
Collapse
Affiliation(s)
- Nooshin Mohebali
- Molecular Bacteriology, Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, 18057 Rostock, Germany
| | - Markus Weigel
- Institute of Medical Microbiology, Justus Liebig University, 35392 Giessen, Germany
| | - Torsten Hain
- Institute of Medical Microbiology, Justus Liebig University, 35392 Giessen, Germany; German Center for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, 35392 Giessen, Germany
| | - Mona Sütel
- IMD Institut für Medizinische Diagnostik, Berlin-Potsdam GbR, 12247 Berlin, Germany
| | - Jana Bull
- Molecular Bacteriology, Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, 18057 Rostock, Germany
| | - Bernd Kreikemeyer
- Molecular Bacteriology, Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, 18057 Rostock, Germany.
| | - Anne Breitrück
- Molecular Bacteriology, Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, 18057 Rostock, Germany
| |
Collapse
|
45
|
Lian YZ, Liu YC, Chang CC, Nochi T, Chao JCJ. Combined Lycium barbarum Polysaccharides with Plasmon-Activated Water Affect IFN-γ/TNF-α Induced Inflammation in Caco-2 Cells. Pharmaceuticals (Basel) 2023; 16:1455. [PMID: 37895926 PMCID: PMC10610401 DOI: 10.3390/ph16101455] [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: 09/07/2023] [Revised: 10/05/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
The effects of Lycium barbarum polysaccharides (LBP) and plasmon-activated water (PAW) against IFN-γ/TNF-α induced inflammation in human colon Caco-2 cells were investigated. Cells were divided into the control, induction, LBP treatment (100-500 μg/mL), and combination groups with PAW. Inflammation was induced 24 h with 10 ng/mL IFN-γ when cell confluency reached >90%, and various doses of LBP with or without PAW were treated for 3 h, and subsequently 50 ng/mL TNF-α was added for another 24 h to provoke inflammation. Combination of LBP with PAW significantly decreased the secretion of IL-6 and IL-8. Cyclooxygenase-2 and inducible NO synthase expression was attenuated in all LBP-treated groups with or without PAW. NLRP3 inflammasome and related protein PYCARD expression were inhibited by LBP at the highest dose (500 μg/mL). All doses of LBP alone significantly decreased p-ERK expression, but combination with PAW increased p-ERK expression compared to those without PAW. Additionally, 250 and 500 μg/mL of LBP with or without PAW inhibited procaspase-3/caspase-3 expression. Therefore, LBP possesses anti-inflammation and anti-apoptosis by inhibiting the secretion of inflammatory cytokines and the expression of NLRP3 inflammasome-related protein. The combination with PAW exerts additive or synergistic effect on anti-inflammation.
Collapse
Affiliation(s)
- Yu Zhi Lian
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110301, Taiwan;
| | - Yu-Chuan Liu
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, Taipei Medical University, Taipei 110301, Taiwan;
- Cell Physiology and Molecular Image Research Center, Taipei Municipal Wan Fang Hospital, Taipei Medical University, Taipei 110301, Taiwan
| | - Chun-Chao Chang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 110301, Taiwan;
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Tomonori Nochi
- Laboratory of Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8577, Japan;
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8577, Japan
| | - Jane C.-J. Chao
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110301, Taiwan;
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Master Program in Global Health and Health Security, Taipei Medical University, Taipei 110301, Taiwan
- Nutrition Research Center, Taipei Medical University Hospital, Taipei 110301, Taiwan
| |
Collapse
|
46
|
Zhou F, Wu NZ, Xie Y, Zhou XJ. Intestinal barrier in inflammatory bowel disease: A bibliometric and knowledge-map analysis. World J Gastroenterol 2023; 29:5254-5267. [PMID: 37901448 PMCID: PMC10600957 DOI: 10.3748/wjg.v29.i36.5254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/11/2023] [Accepted: 09/08/2023] [Indexed: 09/20/2023] Open
Abstract
BACKGROUND Barrier surfaces composed of specialized epithelial cells separate the host body from the external environment, and are essential for maintaining proper intestinal physiologic and immune homeostasis. AIM To explore the development trends and research hotspots of intestinal barrier research in inflammatory bowel disease (IBD). METHODS The publications related to the intestinal barrier in IBD were obtained from the Web of Science Core Collection database. Bibliometric analysis and visualization were conducted using VOSviewer, CiteSpace and R software. RESULTS A total of 4482 articles published between 2002 and 2022 were identified. The United States is dominant in intestinal barrier research, whereas the University of Chicago is the most active institution. Jerrold from Harvard Medical School was the most productive authors with the most citations. The journals Inflammatory Bowel Disease and Gastroenterology have made significant contributions in this field. The keywords appearing at high frequency related to the intestinal barrier in IBD were detected, including nuclear factor kappa B, tumor necrosis factor-α, apoptosis, oxidative stress and probiotics. Among them, antioxidants, Akkermansia muciniphila, nanoparticles, short-chain fatty acids and extracellular vesicles have received growing interest in recent research. CONCLUSION The intestinal barrier field is developing rapidly with extensive cooperation. Targeting the gut microbiota and dietary metabolism to regulate the intestinal barrier has shown promising prospective applications and has generated broad interest. The importance of the intestinal barrier in IBD is gradually being fully recognized, providing a new therapeutic perspective for improving inflammation and prognosis.
Collapse
Affiliation(s)
- Feng Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, China
| | - Nan-Zhen Wu
- Department of Gastrointestinal Surgery, Fengcheng People's Hospital, Fengcheng 331100, Jiangxi Province, China
| | - Yong Xie
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, China
| | - Xiao-Jiang Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, China
| |
Collapse
|
47
|
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
|
48
|
Kim H, Yoo MS, Jeon H, Shim JJ, Park WJ, Kim JY, Lee JL. Probiotic Properties and Safety Evaluation of Lactobacillus plantarum HY7718 with Superior Storage Stability Isolated from Fermented Squid. Microorganisms 2023; 11:2254. [PMID: 37764098 PMCID: PMC10534859 DOI: 10.3390/microorganisms11092254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
The aim of this study was to identify new potential probiotics with improved storage stability and to evaluate their efficacy and safety. Sixty lactic acid bacteria strains were isolated from Korean traditional fermented foods, and their survival was tested under extreme conditions. Lactobacillus plantarum HY7718 (HY7718) showed the greatest stability during storage. HY7718 also showed a stable growth curve under industrial conditions. Whole genome sequencing revealed that the HY7718 genome comprises 3.26 Mbp, with 44.5% G + C content, and 3056 annotated Protein-coding DNA sequences (CDSs). HY7718 adhered to intestinal epithelial cells and was tolerant to gastric fluids. Additionally, HY7718 exhibited no hemolytic activity and was not resistant to antibiotics, confirming that it has probiotic properties and is safe for consumption. Additionally, we evaluated its effects on intestinal health using TNF-induced Caco-2 cells. HY7718 restored the expression of tight junction proteins such as zonular occludens (ZO-1, ZO-2), occludin (OCLN), and claudins (CLDN1, CLDN4), and regulated the expression of myosin light-chain kinase (MLCK), Elk-1, and nuclear factor kappa B subunit 1 (NFKB1). Moreover, HY7718 reduced the secretion of proinflammatory cytokines such as interleukin-6 (IL-6) and IL-8, as well as reducing the levels of peroxide-induced reactive oxygen species. In conclusion, HY7718 has probiotic properties, is safe, is stable under extreme storage conditions, and exerts positive effects on intestinal cells. These results suggest that L. plantarum HY7718 is a potential probiotic for use as a functional supplement in the food industry.
Collapse
Affiliation(s)
- Hyeonji Kim
- R&BD Center, hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (H.K.); (M.-S.Y.); (H.J.); (J.-J.S.)
| | - Myeong-Seok Yoo
- R&BD Center, hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (H.K.); (M.-S.Y.); (H.J.); (J.-J.S.)
| | - Hyejin Jeon
- R&BD Center, hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (H.K.); (M.-S.Y.); (H.J.); (J.-J.S.)
| | - Jae-Jung Shim
- R&BD Center, hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (H.K.); (M.-S.Y.); (H.J.); (J.-J.S.)
| | - Woo-Jung Park
- Department of Marine Food Science and Technology, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea;
| | - Joo-Yun Kim
- R&BD Center, hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (H.K.); (M.-S.Y.); (H.J.); (J.-J.S.)
| | - Jung-Lyoul Lee
- R&BD Center, hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (H.K.); (M.-S.Y.); (H.J.); (J.-J.S.)
| |
Collapse
|
49
|
Ding N, Xiao H, Zhen L, Li H, Zhang Z, Ge J, Jia H. Systemic cytokines inhibition with Imp7 siRNA nanoparticle ameliorates gut injury in a mouse model of ventilator-induced lung injury. Biomed Pharmacother 2023; 165:115237. [PMID: 37516020 DOI: 10.1016/j.biopha.2023.115237] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/22/2023] [Accepted: 07/25/2023] [Indexed: 07/31/2023] Open
Abstract
Mechanical ventilation (MV) may negatively affect the lungs and cause the release of inflammatory mediators, resulting in extra-pulmonary organ dysfunction. Studies have revealed systemically elevated levels of proinflammatory cytokines in animal models of ventilator-induced lung injury (VILI); however, whether these cytokines have an effect on gut injury and the mechanisms involved remain unknown. In this study, VILI was generated in mice with high tidal volume mechanical ventilation (20 ml/kg). Tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and IL-6 concentrations in serum and gut measured by ELISA showed significant elevation in the VILI mice. Significant increases in gut injury and PANoptosis were observed in the VILI mice, which were positively correlated with the serum levels of TNF-α, IL-1β, and IL-6. The VILI mice displayed intestinal barrier defects, decreased expressions of occludin and zonula occludin-1 (ZO-1), and increased expression of claudin-2 and the activation of myosin light chain (MLC). Importantly, intratracheal administration of Imp7 siRNA nanoparticle effectively inhibited cytokines production and protected mice from VILI-induced gut injury. These data provide evidence of systemic cytokines contributing to gut injury following VILI and highlight the possibility of targeting cytokines inhibition via Imp7 siRNA nanoparticle as a potential therapeutic intervention for alleviating gut injury following VILI.
Collapse
Affiliation(s)
- Ning Ding
- Key Laboratory of Intensive Care Rehabilitation of Shandong, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China; Department of Anesthesiology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China.
| | - Hui Xiao
- Key Laboratory of Intensive Care Rehabilitation of Shandong, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
| | - Lixiao Zhen
- Key Laboratory of Intensive Care Rehabilitation of Shandong, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
| | - Huiqing Li
- Key Laboratory of Intensive Care Rehabilitation of Shandong, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China; Department of Anesthesiology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
| | - Zengzhen Zhang
- Key Laboratory of Intensive Care Rehabilitation of Shandong, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China; Department of Anesthesiology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
| | - Junke Ge
- Key Laboratory of Intensive Care Rehabilitation of Shandong, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China; Department of Intensive Care Medicine, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Haiyan Jia
- Key Laboratory of Intensive Care Rehabilitation of Shandong, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China; Department of Intensive Care Medicine, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
50
|
Barni S, Mori F, Giovannini M, Liotti L, Mastrorilli C, Pecoraro L, Saretta F, Castagnoli R, Arasi S, Caminiti L, Gelsomino M, Klain A, del Giudice MM, Novembre E. Allergic Proctocolitis: Literature Review and Proposal of a Diagnostic-Therapeutic Algorithm. Life (Basel) 2023; 13:1824. [PMID: 37763228 PMCID: PMC10533178 DOI: 10.3390/life13091824] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Allergic proctocolitis (AP) is a benign condition, frequent in childhood, that is classified as a non-IgE-mediated food allergy. The prevalence is unknown; however, its frequency appears to be increasing, especially in exclusively breastfed infants. Clinical manifestations typically begin in the first few months of life with the appearance of bright red blood (hematochezia), with or without mucus, in the stool of apparently healthy, thriving infants. Most cases of AP are caused by cow's milk proteins; however, other allergens, such as soy, egg, corn, and wheat, may be potential triggers. Diagnosis is based on the patient's clinical history and on the resolution of signs and symptoms with the elimination of the suspected food antigen from the diet and their reappearance when the food is reintroduced into the diet. The treatment of AP is based on an elimination diet of the trigger food, with resolution of the symptoms within 72-96 h from the beginning of the diet. The prognosis of AP is good; it is a self-limiting condition, because most children can tolerate the trigger food within one year of life, with an excellent long-term prognosis. The purpose of this review is to provide an update on the current knowledge and recommendations in epidemiological, diagnostic, and therapeutic terms to the pediatricians, allergists, and gastroenterologists who may find themselves managing a patient with AP.
Collapse
Affiliation(s)
- Simona Barni
- Allergy Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy; (S.B.); (F.M.); (M.G.); (E.N.)
| | - Francesca Mori
- Allergy Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy; (S.B.); (F.M.); (M.G.); (E.N.)
| | - Mattia Giovannini
- Allergy Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy; (S.B.); (F.M.); (M.G.); (E.N.)
- Department of Health Sciences, University of Florence, 50139 Florence, Italy
| | - Lucia Liotti
- Pediatric Unit, Department of Mother and Child Health, Salesi Children’s Hospital, 60123 Ancona, Italy;
| | - Carla Mastrorilli
- Pediatric and Emergency Department, Pediatric Hospital Giovanni XXIII, AOU Policlinic of Bari, 70126 Bari, Italy;
| | - Luca Pecoraro
- Pediatric Unit, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, 37126 Verona, Italy;
| | - Francesca Saretta
- Pediatric Department, Latisana-Palmanova Hospital, Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy;
| | - Riccardo Castagnoli
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy;
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Stefania Arasi
- Translational Research in Pediatric Specialties Area, Division of Allergy, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Lucia Caminiti
- Allergy Unit, Department of Pediatrics, AOU Policlinico Gaetano Martino, 98124 Messina, Italy;
| | - Mariannita Gelsomino
- Department of Life Sciences and Public Health, Pediatric Allergy Unit, University Foundation Policlinico Gemelli IRCCS, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Angela Klain
- Department of Woman, Child and General and Specialized Surgery, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.K.); (M.M.d.G.)
| | - Michele Miraglia del Giudice
- Department of Woman, Child and General and Specialized Surgery, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.K.); (M.M.d.G.)
| | - Elio Novembre
- Allergy Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy; (S.B.); (F.M.); (M.G.); (E.N.)
| |
Collapse
|