1
|
Van Acker N, Frenois FX, Gravelle P, Tosolini M, Syrykh C, Laurent C, Brousset P. Spatial mapping of innate lymphoid cells in human lymphoid tissues and lymphoma at single-cell resolution. Nat Commun 2025; 16:4545. [PMID: 40374674 PMCID: PMC12081901 DOI: 10.1038/s41467-025-59811-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 05/06/2025] [Indexed: 05/17/2025] Open
Abstract
Innate lymphoid cells (ILC) distribution and compartmentalization in human lymphoid tissues are incompletely described. Through combined multiplex immunofluorescence, multispectral imaging, and advanced computer vision methods, we provide a map of ILCs at the whole-slide single-cell resolution level, and study their proximity to T helper (Th) cells. The results show that ILC2 predominates in thymic medulla; by contrast, immature Th cells prevail in the cortex. Unexpectedly, we find that Th2-like and Th17-like phenotypes appear before complete T cell receptor gene rearrangements in these immature thymocytes. In the periphery, ILC2 are more abundant in lymph nodes and tonsils, penetrating lymphoid follicles. NK cells are uncommon in lymphoid tissues but abundant in the spleen, whereas ILC1 and ILC3 predominate in the ileum and appendix. Under pathogenic conditions, a deep perturbation of both ILC and Th populations is seen in follicular lymphoma compared with non-neoplastic conditions. Lastly, all ILCs are preferentially in close proximity to their Th counterparts. In summary, our histopathology tool help present a spatial mapping of human ILCs and Th cells, in normal and neoplastic lymphoid tissues.
Collapse
Affiliation(s)
- Nathalie Van Acker
- Department of Pathology, CHU of Toulouse, Imag'IN Platform, IUCT-Oncopole, Toulouse, France
- Cancer Research Center of Toulouse (INSERM), Team 9 NoLymIT and Labex TOUCAN, Toulouse, France
| | - François-Xavier Frenois
- Department of Pathology, CHU of Toulouse, Imag'IN Platform, IUCT-Oncopole, Toulouse, France
- Cancer Research Center of Toulouse (INSERM), Team 9 NoLymIT and Labex TOUCAN, Toulouse, France
| | - Pauline Gravelle
- Department of Pathology, CHU of Toulouse, Imag'IN Platform, IUCT-Oncopole, Toulouse, France
- Cancer Research Center of Toulouse (INSERM), Team 9 NoLymIT and Labex TOUCAN, Toulouse, France
| | - Marie Tosolini
- Cancer Research Center of Toulouse (INSERM), Team 9 NoLymIT and Labex TOUCAN, Toulouse, France
| | - Charlotte Syrykh
- Department of Pathology, CHU of Toulouse, Imag'IN Platform, IUCT-Oncopole, Toulouse, France
- Cancer Research Center of Toulouse (INSERM), Team 9 NoLymIT and Labex TOUCAN, Toulouse, France
| | - Camille Laurent
- Department of Pathology, CHU of Toulouse, Imag'IN Platform, IUCT-Oncopole, Toulouse, France
- Cancer Research Center of Toulouse (INSERM), Team 9 NoLymIT and Labex TOUCAN, Toulouse, France
| | - Pierre Brousset
- Department of Pathology, CHU of Toulouse, Imag'IN Platform, IUCT-Oncopole, Toulouse, France.
- Cancer Research Center of Toulouse (INSERM), Team 9 NoLymIT and Labex TOUCAN, Toulouse, France.
| |
Collapse
|
2
|
Tulic MK, Bzioueche H. Promising Exopolysaccharide as Complement to Conventional Therapy for Patients with Vitiligo. J Invest Dermatol 2025:S0022-202X(25)00073-9. [PMID: 39969457 DOI: 10.1016/j.jid.2025.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 01/17/2025] [Indexed: 02/20/2025]
Affiliation(s)
- Meri K Tulic
- INSERM U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Université Côte d'Azur, Nice, France.
| | - Hanene Bzioueche
- INSERM U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Université Côte d'Azur, Nice, France
| |
Collapse
|
3
|
Aljohani H, Anbarserry D, Mosli M, Ujaimi A, Bakhshwin D, Elango R, Alharthi S. High-Throughput Whole-Exome Sequencing and Large-Scale Computational Analysis to Identify the Genetic Biomarkers to Predict the Vedolizumab Response Status in Inflammatory Bowel Disease Patients from Saudi Arabia. Biomedicines 2025; 13:459. [PMID: 40002872 PMCID: PMC11852680 DOI: 10.3390/biomedicines13020459] [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: 01/18/2025] [Revised: 02/04/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Vedolizumab (VDZ) is the new monoclonal drug targeting α4β7 integrin for patients with moderate/severe IBD. Between 30 and 45% of patients fail to respond to VDZ after 14-16 weeks of treatment. The aim of the study was to explore the genetic profile of vedolizumab-treated Arab IBD patients in Saudi Arabia to identify the potential biomarkers to differentiate the responders from non-responders. Methods: A cohort of 16 patients with IBD, including 4 with Crohn's disease and 12 with ulcerative colitis, were recruited. Following 16 weeks of VDZ treatment, nine were found to be responders and seven non-responders. Blood samples were collected for the whole exome sequencing of DNA from all patients. The variants in the whole-exome sequencing data were analyzed with a variety of bioinformatics tools and databases, such as Polyphen2, Mutation Taster, CADD, FATHMM, Open Target Platform, TOPPFun, STRING, and GTEx. Results: More than 1.6 million variants from 16 samples were analyzed. The rare variant analysis prioritized NOD2, IL23, IL10, IL27, and TRAF1 genes in non-responders. NOD2, IL23, IL10, IL27, and TRAF1 were found to be the significant IBD risk factors in multiple genome-wide association studies, and their pro-inflammatory activity might contribute to the inherent resistance to VDZ. Rare variants of CARD9, TYK2, IL4, and NLRP1 genes present in VDZ responders enhance the anti-inflammatory/immune modulation effects. Conclusions: This investigation is the first to apply whole-exome sequencing to identify the potential drug response biomarkers for the IBD drug VDZ in Saudi Arabia.
Collapse
Affiliation(s)
- Hanin Aljohani
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (H.A.); (D.A.); (D.B.)
| | - Doaa Anbarserry
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (H.A.); (D.A.); (D.B.)
| | - Mahmoud Mosli
- Department of Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia;
- Inflammatory Bowel Disease Research Group, King Abdulaziz University, Jeddah 22252, Saudi Arabia
- Inflammatory Bowel Disease Unit, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Amani Ujaimi
- Princess Al-Jawahara Cernter of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah 22252, Saudi Arabia;
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Duaa Bakhshwin
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (H.A.); (D.A.); (D.B.)
| | - Ramu Elango
- Princess Al-Jawahara Cernter of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah 22252, Saudi Arabia;
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Sameer Alharthi
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (H.A.); (D.A.); (D.B.)
- Inflammatory Bowel Disease Research Group, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| |
Collapse
|
4
|
Chen Y, Gu X, Cao K, Tu M, Liu W, Ju J. The role of innate lymphoid cells in systemic lupus erythematosus. Cytokine 2024; 179:156623. [PMID: 38685155 DOI: 10.1016/j.cyto.2024.156623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 05/02/2024]
Abstract
Systemic lupus erythematosus (SLE) is a connective tissue disorder that affects various body systems. Both the innate and adaptive immunity contribute to the onset and progression of SLE. The main mechanism of SLE is an excessive immune response of immune cells to autoantigens, which leads to systemic inflammation and inflammation-induced organ damage. Notably, a subset of innate immune cells known as innate lymphoid cells (ILCs) has recently emerged. ILCs are pivotal in the early stages of infection; participate in immune responses, inflammation, and tissue repair; and regulate the immune function of the body by resisting pathogens and regulating autoimmune inflammation and metabolic homeostasis. Thus, ILCs dysfunction can lead to autoimmune diseases. This review discusses the maturation of ILCs, the potential mechanisms by which ILCs exacerbate SLE pathogenesis, and their contributions to organ inflammatory deterioration in SLE.
Collapse
Affiliation(s)
- Yong Chen
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China
| | - Xiaotian Gu
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China
| | - Kunyu Cao
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China
| | - Miao Tu
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China
| | - Wan Liu
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China.
| | - Jiyu Ju
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
5
|
Hu Y, Tang J, Xie Y, Xu W, Zhu W, Xia L, Fang J, Yu D, Liu J, Zheng Z, Zhou Q, Shou Q, Zhang W. Gegen Qinlian decoction ameliorates TNBS-induced ulcerative colitis by regulating Th2/Th1 and Tregs/Th17 cells balance, inhibiting NLRP3 inflammasome activation and reshaping gut microbiota. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:117956. [PMID: 38428658 DOI: 10.1016/j.jep.2024.117956] [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: 11/20/2023] [Revised: 02/14/2024] [Accepted: 02/19/2024] [Indexed: 03/03/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chinese herbal medicine Gegen Qinlian Decoction (GQD) has been clinically shown to be an effective treatment of ulcerative colitis (UC) in China. However, the underlying mechanism of GQD's anti-ulcerative colitis properties and its effect on gut microbiota still deserve further exploration. AIM OF THE STUDY This study observed the regulatory effects of GQD on Th2/Th1 and Tregs/Th17 cells balance, the NOD-like receptor family pyrin domain containing 3 (NLRP3) infammasome and gut microbiota in TNBS-induced UC in BALB/c mice. MATERIALS AND METHODS 61 main chemical compounds in the GQD were determined by UPLC-Q-TOF/MS. The UC BALB/c model was established by intrarectal administration of trinitrobenzene sulfonic acid (TNBS), and GQD was orally administered at low and high dosages of 2.96 and 11.83 g/kg/day, respectively. The anti-inflammatory effects of GQD for ulcerative colitis were evaluated by survival rate, body weight, disease activity index (DAI) score, colonic weight and index, spleen index, hematoxylin-eosin (HE) staining and histopathological scores. Flow cytometry was used to detect the percentage of CD4, Th1, Th2, Th17 and Tregs cells. The levels of Th1-/Th2-/Th17-/Tregs-related inflammatory cytokines and additional proinflammatory cytokines (IL-1β, IL-18) were detected by CBA, ELISA, and RT-PCR. The expressions of GATA3, T-bet, NLRP3, Caspase-1, IL-Iβ, Occludin and Zonula occludens-1 (ZO-1) on colon tissues were detected by Western blot and RT-PCR. Transcriptome sequencing was performed using colon tissue and 16S rRNA gene sequencing was performed on intestinal contents. Fecal microbiota transplantation (FMT) was employed to assess the contribution of intestinal microbiota and its correlation with CD4 T cells and the NLRP3 inflammasome. RESULTS GQD increased the survival rate of TNBS-induced UC in BALB/c mice, and significantly improved their body weight, DAI score, colonic weight and index, spleen index, and histological characteristics. The intestinal barrier dysfunction was repaired after GQD administration through promoting the expression of tight junction proteins (Occludin and ZO-1). GQD restored the balance of Th2/Th1 and Tregs/Th17 cells immune response of colitis mice, primarily inhibiting the increase in Th2/Th1 ratio and their transcription factor production (GATA3 and T-bet). Morever, GQD changed the secretion of Th1-/Th2-/Th17-/Tregs-related cytokines (IL-2, IL-12, IL-5, IL-13, IL-6, IL-10, and IL-17A) and reduced the expressions of IL-1β, IL-18. Transcriptome results suggested that GQD could also remodel the immune inflammatory response of colitis by inhibiting NOD-like receptor signaling pathway, and Western blot, immunohistochemistry and RT-PCR further revealed that GQD exerted anti-inflammatory effects by inhibiting the NLRP3 inflammasome, such as down-regulating the expression of NLRP3, Caspase-1 and IL-1β. More interestingly, GQD regulated gut microbiota dysbiosis, suppressed the overgrowth of conditional pathogenic gut bacteria like Helicobacter, Proteobacteria, and Mucispirillum, while the probiotic gut microbiota, such as Lactobacillus, Muribaculaceae, Ruminiclostridium_6, Akkermansia, and Ruminococcaceae_unclassified were increased. We further confirmed that GQD-treated gut microbiota was sufficient to relieve TNBS-induced colitis by FMT, involving the modulation of Th2/Th1 and Tregs/Th17 balance, inhibition of NLRP3 inflammasome activation, and enhancement of colonic barrier function. CONCLUSIONS GQD might alleviate TNBS-induced UC via regulating Th2/Th1 and Tregs/Th17 cells Balance, inhibiting NLRP3 inflammasome and reshaping gut microbiota, which may provide a novel strategy for patients with colitis.
Collapse
Affiliation(s)
- Yingnan Hu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jingyi Tang
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yongfeng Xie
- Department of Burn Plastic Surgery, Huai'an Hospital Affiliated to Xuzhou Medical University, Jiangsu, 223001, China
| | - Wenjun Xu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Weihan Zhu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Linying Xia
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Jintao Fang
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Dian Yu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jingjing Liu
- Department of General Surgery, Haining City Central Hospital, Jiaxing, 314408, China
| | - Zhipeng Zheng
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China
| | - Qiujing Zhou
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China
| | - Qiyang Shou
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China.
| | - Wei Zhang
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China; The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China.
| |
Collapse
|
6
|
Wei Z, Tang X, Yi C, Ocansey DKW, Mao F, Mao Z. HucMSC-Ex alleviates DSS-induced colitis in mice by decreasing mast cell activation via the IL-33/ST2 axis. Am J Transl Res 2024; 16:2727-2744. [PMID: 39006299 PMCID: PMC11236658 DOI: 10.62347/exze5413] [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: 05/22/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a chronic inflammatory disease that poses challenges in terms of treatment. The precise mechanism underlying the role of human umbilical cord mesenchymal stem cell-derived exosome (HucMSC-Ex) in the inflammatory repair process of IBD remains elusive. Mucosal mast cells accumulate within the intestinal tract and exert regulatory functions in IBD, thus presenting a novel target for addressing this intestinal disease. METHODS A mouse model of Dextran Sulfate Sodium (DSS)-induced colitis was established and hucMSC-Ex were administered to investigate their impact on the regulation of intestinal mast cells. An in vitro co-culture model using the human clonal colorectal adenocarcinoma cell line (Caco-2) and human mast cell line (LAD2) was also established for further exploration of the effect of hucMSC-Ex. RESULTS We observed the accumulation of mast cells in the intestines of patients with IBD as well as mice. In colitis mice, there was an upregulation of mast cell-related tryptase, interleukin-33 (IL-33), and suppression of tumorigenicity 2 receptor (ST2 or IL1RL1), and the function of the intestinal mucosal barrier related to intestinal tight junction protein was weakened. HucMSC-Ex treatment significantly reduced mast cell infiltration and intestinal damage. In the co-culture model, a substantial number of mast cells interact with the epithelial barrier, triggering activation of the IL-33/IL1RL1 (ST2) pathway and subsequent release of inflammatory factors and trypsin. This disruption leads to aberrant expression of tight junction proteins, which can be alleviated by supplementation with hucMSC-Ex. CONCLUSION Our results suggest that hucMSC-Ex may reduce the release of mast cell mediators via the IL-33/IL1RL1 (ST2) axis, thereby mitigating its detrimental effects on intestinal barrier function.
Collapse
Affiliation(s)
- Zhiping Wei
- Department of Laboratory Medicine, The Affiliated People’s Hospital, Jiangsu UniversityZhenjiang 212002, Jiangsu, P. R. China
- Department of Clinical Laboratory, The Third People’s Hospital of Xindu DistrictChengdu 610500, Sichuan, P. R. China
| | - Xiaohua Tang
- Department of Orthopaedics, The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong UniversityZhenjiang 212300, Jiangsu, P. R. China
| | - Chengxue Yi
- School of Medical Technology, Zhenjiang CollegeZhenjiang 212028, Jiangsu, P. R. China
| | - Dickson Kofi Wiredu Ocansey
- Department of Laboratory Medicine, The Affiliated People’s Hospital, Jiangsu UniversityZhenjiang 212002, Jiangsu, P. R. China
- Department of Medical Laboratory Science, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape CoastCape Coast CC0959347, Ghana
| | - Fei Mao
- Department of Laboratory Medicine, The Affiliated People’s Hospital, Jiangsu UniversityZhenjiang 212002, Jiangsu, P. R. China
| | - Zhenwei Mao
- The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Affiliated People’s Hospital of Jiangsu UniversityZhenjiang 212002, Jiangsu, P. R. China
| |
Collapse
|
7
|
Cantorna MT, Arora J. Vitamin D, microbiota, and inflammatory bowel disease. FELDMAN AND PIKE'S VITAMIN D 2024:1057-1073. [DOI: 10.1016/b978-0-323-91338-6.00047-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
8
|
Selvakumar B, Eladham MW, Hafezi S, Ramakrishnan R, Hachim IY, Bayram OS, Sharif-Askari NS, Sharif-Askari FS, Ibrahim SM, Halwani R. Allergic Airway Inflammation Emerges from Gut Inflammation and Leakage in Mouse Model of Asthma. Adv Biol (Weinh) 2024; 8:e2300350. [PMID: 37752729 DOI: 10.1002/adbi.202300350] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/16/2023] [Indexed: 09/28/2023]
Abstract
Asthma is an allergic airway inflammatory disease characterized by type 2 immune responses. Growing evidence suggests an association between allergic airways and intestinal diseases. However, the primary site of disease origin and initial mechanisms involved in the development of allergic airway inflammation (AAI) is not yet understood. Therefore, the initial contributing organs and mechanisms involved in the development of AAI are investigated using a mouse model of asthma. This study, without a local allergen challenge into the lungs, demonstrates a significant increase in intestinal inflammation with signature type-2 mediators including IL-4, IL-13, STAT6, eosinophils, and Th2 cells. In addition, gut leakage and mRNA expressions of gut leakage markers significantly increase in the intestine. Moreover, reduced mRNA expressions of tight junction proteins are observed in gut and interestingly, in lung tissues. Furthermore, in lung tissues, an increased pulmonary barrier permeability and IL-4 and IL-13 levels associated with significant increase of lipopolysaccharide-binding protein (LBP-gut leakage marker) and eosinophils are observed. However, with local allergen challenges into the lungs, these mechanisms are further enhanced in both gut and lungs. In conclusion, the primary gut originated inflammatory responses translocates into the lungs to orchestrate AAI in a mouse model of asthma.
Collapse
Affiliation(s)
- Balachandar Selvakumar
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, UAE
| | - Mariam Wed Eladham
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, UAE
| | - Shirin Hafezi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, UAE
| | - Rakhee Ramakrishnan
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, UAE
| | - Ibrahim Yaseen Hachim
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, UAE
| | - Ola Salam Bayram
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, UAE
| | - Narjes Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, UAE
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, UAE
| | - Fatemeh Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, UAE
- Department of Pharmacy Practice and Pharmaceutics, College of Pharmacy, University of Sharjah, Sharjah, 27272, UAE
| | - Saleh Mohamed Ibrahim
- Institute of Experimental Dermatology, University of Lübeck, 23562, Lübeck, Germany
- Deapartment of Biotechnology, Khalifa University, Abu Dhabi, 127788, UAE
| | - Rabih Halwani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, UAE
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, UAE
- Prince Abdullah Ben Khaled Celiac Disease Research Chair, Department of Pediatrics, Faculty of Medicine, King Saud University, Riyadh, 11461, Saudi Arabia
| |
Collapse
|
9
|
Vebr M, Pomahačová R, Sýkora J, Schwarz J. A Narrative Review of Cytokine Networks: Pathophysiological and Therapeutic Implications for Inflammatory Bowel Disease Pathogenesis. Biomedicines 2023; 11:3229. [PMID: 38137450 PMCID: PMC10740682 DOI: 10.3390/biomedicines11123229] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/11/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a lifelong inflammatory immune mediated disorder, encompassing Crohn's disease (CD) and ulcerative colitis (UC); however, the cause and specific pathogenesis of IBD is yet incompletely understood. Multiple cytokines produced by different immune cell types results in complex functional networks that constitute a highly regulated messaging network of signaling pathways. Applying biological mechanisms underlying IBD at the single omic level, technologies and genetic engineering enable the quantification of the pattern of released cytokines and new insights into the cytokine landscape of IBD. We focus on the existing literature dealing with the biology of pro- or anti-inflammatory cytokines and interactions that facilitate cell-based modulation of the immune system for IBD inflammation. We summarize the main roles of substantial cytokines in IBD related to homeostatic tissue functions and the remodeling of cytokine networks in IBD, which may be specifically valuable for successful cytokine-targeted therapies via marketed products. Cytokines and their receptors are validated targets for multiple therapeutic areas, we review the current strategies for therapeutic intervention and developing cytokine-targeted therapies. New biologics have shown efficacy in the last few decades for the management of IBD; unfortunately, many patients are nonresponsive or develop therapy resistance over time, creating a need for novel therapeutics. Thus, the treatment options for IBD beyond the immune-modifying anti-TNF agents or combination therapies are expanding rapidly. Further studies are needed to fully understand the immune response, networks of cytokines, and the direct pathogenetic relevance regarding individually tailored, safe and efficient targeted-biotherapeutics.
Collapse
Affiliation(s)
- Marek Vebr
- Departments of Pediatrics, Faculty Hospital, Faculty of Medicine in Pilsen, Charles University of Prague, 323 00 Pilsen, Czech Republic; (R.P.); (J.S.); (J.S.)
| | | | | | | |
Collapse
|
10
|
Wang Y, Li Q, Zhang J, Liu P, Zheng H, Chen L, Wang Z, Tan C, Zhang M, Zhang H, Miao W, Wang Y, Xuan X, Yi G, Wang P. Ring1a protects against colitis through regulating mucosal immune system and colonic microbial ecology. Gut Microbes 2023; 15:2251646. [PMID: 37655448 PMCID: PMC10478745 DOI: 10.1080/19490976.2023.2251646] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/19/2023] [Accepted: 08/21/2023] [Indexed: 09/02/2023] Open
Abstract
Inflammatory bowel disease (IBD) represents a prominent chronic immune-mediated inflammatory disorder, yet its etiology remains poorly comprehended, encompassing intricate interactions between genetics, immunity, and the gut microbiome. This study uncovers a novel colitis-associated risk gene, namely Ring1a, which regulates the mucosal immune response and intestinal microbiota. Ring1a deficiency exacerbates colitis by impairing the immune system. Concomitantly, Ring1a deficiency led to a Prevotella genus-dominated pathogenic microenvironment, which can be horizontally transmitted to co-housed wild type (WT) mice, consequently intensifying dextran sodium sulfate (DSS)-induced colitis. Furthermore, we identified a potential mechanism linking the altered microbiota in Ring1aKO mice to decreased levels of IgA, and we demonstrated that metronidazole administration could ameliorate colitis progression in Ring1aKO mice, likely by reducing the abundance of the Prevotella genus. We also elucidated the immune landscape of DSS colitis and revealed the disruption of intestinal immune homeostasis associated with Ring1a deficiency. Collectively, these findings highlight Ring1a as a prospective candidate risk gene for colitis and suggest metronidazole as a potential therapeutic option for clinically managing Prevotella genus-dominated colitis.
Collapse
Affiliation(s)
- Yashu Wang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Infection, Inflammation and Immunity Center, the Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Qianru Li
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Infection, Inflammation and Immunity Center, the Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Jiayu Zhang
- Lab of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Pingping Liu
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Infection, Inflammation and Immunity Center, the Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Huaixin Zheng
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Infection, Inflammation and Immunity Center, the Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Lijuan Chen
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Zhen Wang
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Chen Tan
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Infection, Inflammation and Immunity Center, the Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Min Zhang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Infection, Inflammation and Immunity Center, the Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Hongxia Zhang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Infection, Inflammation and Immunity Center, the Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Wenqing Miao
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Infection, Inflammation and Immunity Center, the Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Yuke Wang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Infection, Inflammation and Immunity Center, the Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Xiaoyan Xuan
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Infection, Inflammation and Immunity Center, the Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Guoqiang Yi
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Peng Wang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Infection, Inflammation and Immunity Center, the Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Eigenschink M, Wessely I, Dijmarescu M, Förster-Waldl E, Farr A, Kiss H, Berger A, Wisgrill L. Transcriptomic analysis identifies lactoferrin-induced quiescent circuits in neonatal macrophages. Front Immunol 2023; 14:1276173. [PMID: 37868991 PMCID: PMC10590118 DOI: 10.3389/fimmu.2023.1276173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
Introduction Upon birth, a hitherto naïve immune system is confronted with a plethora of microbial antigens due to intestinal bacterial colonization. To prevent excessive inflammation and disruption of the epithelial barrier, physiological mechanisms must promote immune-anergy within the neonatal gut. As high concentrations of human lactoferrin (hLF), a transferrin glycoprotein shown to modulate macrophage function, are frequently encountered in colostrum, its direct interaction with intestinal macrophages may satisfy this physiological need. Thus, the primary objective of this study was to investigate transcriptional changes induced by human lactoferrin in neonatal monocyte-derived macrophages. Methods Cord blood-derived monocytes were differentiated with M-CSF in presence or absence of 500 µg/mL hLF for 7 days and afterwards stimulated with 1 ng/mL LPS or left untreated. RNA was then isolated and subjected to microarray analysis. Results Differentiation of cord blood-derived monocytes in presence of hLF induced a distinct transcriptional program defined by cell cycle arrest in the G2/M phase, induction of IL-4/IL-13-like signaling, altered extracellular matrix interaction, and enhanced propensity for cell-cell interaction. Moreover, near-complete abrogation of transcriptional changes induced by TLR4 engagement with LPS was observed in hLF-treated samples. Discussion The global transition towards an M2-like homeostatic phenotype and the acquisition of quiescence elegantly demonstrate the ontogenetical relevance of hLF in attenuating pro-inflammatory signaling within the developing neonatal intestine. The marked anergy towards proinflammatory stimuli such as LPS further underlines the glycoprotein's potential therapeutic relevance.
Collapse
Affiliation(s)
- Michael Eigenschink
- Division of Neonatology, Pediatric Intensive Care and Neuropaediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Isabelle Wessely
- Division of Neonatology, Pediatric Intensive Care and Neuropaediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Marco Dijmarescu
- Division of Neonatology, Pediatric Intensive Care and Neuropaediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Elisabeth Förster-Waldl
- Division of Neonatology, Pediatric Intensive Care and Neuropaediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Alex Farr
- Department of Obstetrics and Gynecology, Division of Obstetrics and Feto-Maternal Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Herbert Kiss
- Department of Obstetrics and Gynecology, Division of Obstetrics and Feto-Maternal Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Angelika Berger
- Division of Neonatology, Pediatric Intensive Care and Neuropaediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Lukas Wisgrill
- Division of Neonatology, Pediatric Intensive Care and Neuropaediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
12
|
Jing S, Chen H, Liu E, Zhang M, Zeng F, Shen H, Fang Y, Muhitdinov B, Huang Y. Oral pectin/oligochitosan microspheres for colon-specific controlled release of quercetin to treat inflammatory bowel disease. Carbohydr Polym 2023; 316:121025. [PMID: 37321723 DOI: 10.1016/j.carbpol.2023.121025] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/29/2023] [Accepted: 05/11/2023] [Indexed: 06/17/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic, life quality-reducing disease with no cures available yet. To develop an effective medication suitable for long-term use is an urgent but unmet need. Quercetin (QT) is a natural dietary flavonoid with good safety and multifaceted pharmacological activities against inflammation. However, orally administrated quercetin yields unproductive outcomes for IBD treatment because of its poor solubility and extensive metabolism in the gastrointestinal tract. In this work, a colon-targeted QT delivery system (termed COS-CaP-QT) was developed, of which the pectin (PEC)/Ca2+ microspheres were prepared and then crosslinked by oligochitosan (COS). The drug release profile of COS-CaP-QT was pH-dependent and colon microenvironment-responsive, and COS-CaP-QT showed preferential distribution in the colon. The mechanism study showed that QT triggered the Notch pathway to regulate the proliferation of T helper 2 (Th2) cells and group 3 innate lymphoid cells (ILC3s) and the inflammatory microenvironment was remodeled. The in vivo therapeutic results revealed that COS-CaP-QT could relieve the colitis symptoms and maintain the colon length and intestinal barrier integrity.
Collapse
Affiliation(s)
- Shisuo Jing
- School of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Huayuan Chen
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ergang Liu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China.
| | - Meng Zhang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Feng Zeng
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510450, China
| | - Huan Shen
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; Shanghai Institute of Materia Medica, CAS, Shanghai 201203, China
| | - Yuefei Fang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Bahtiyor Muhitdinov
- Shanghai Institute of Materia Medica, CAS, Shanghai 201203, China; Institute of Bioorganic Chemistry, Uzbekistan Academy of Sciences, Tashkent 100125, Uzbekistan
| | - Yongzhuo Huang
- School of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; Shanghai Institute of Materia Medica, CAS, Shanghai 201203, China.
| |
Collapse
|
13
|
Hu C, Liao S, Lv L, Li C, Mei Z. Intestinal Immune Imbalance is an Alarm in the Development of IBD. Mediators Inflamm 2023; 2023:1073984. [PMID: 37554552 PMCID: PMC10406561 DOI: 10.1155/2023/1073984] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/06/2023] [Accepted: 07/08/2023] [Indexed: 08/10/2023] Open
Abstract
Immune regulation plays a crucial role in human health and disease. Inflammatory bowel disease (IBD) is a chronic relapse bowel disease with an increasing incidence worldwide. Clinical treatments for IBD are limited and inefficient. However, the pathogenesis of immune-mediated IBD remains unclear. This review describes the activation of innate and adaptive immune functions by intestinal immune cells to regulate intestinal immune balance and maintain intestinal mucosal integrity. Changes in susceptible genes, autophagy, energy metabolism, and other factors interact in a complex manner with the immune system, eventually leading to intestinal immune imbalance and the onset of IBD. These events indicate that intestinal immune imbalance is an alarm for IBD development, further opening new possibilities for the unprecedented development of immunotherapy for IBD.
Collapse
Affiliation(s)
- Chunli Hu
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Shengtao Liao
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Lin Lv
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Chuanfei Li
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Zhechuan Mei
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
14
|
Palmer G, Ramanathan S, Mortier E. Editorial: Modulating cytokines as treatment for autoimmune diseases and cancer: volume II. Front Immunol 2023; 14:1225699. [PMID: 37342326 PMCID: PMC10277794 DOI: 10.3389/fimmu.2023.1225699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 05/30/2023] [Indexed: 06/22/2023] Open
Affiliation(s)
- Gaby Palmer
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sheela Ramanathan
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Erwan Mortier
- Nantes Université, CNRS, Inserm, CRCI2NA, Nantes, France
- LabEX IGO, Immuno-Onco-Greffe, Nantes, France
| |
Collapse
|
15
|
Ionescu EM, Olteanu AO, Tieranu CG, Popa LO, Andrei SI, Preda CM, Dutescu MI, Bojinca M, Tieranu I, Popa OM. Interleukin-4 Gene Polymorphisms in Romanian Patients with Inflammatory Bowel Diseases: Association with Disease Risk and Clinical Features. Diagnostics (Basel) 2023; 13:1465. [PMID: 37189566 PMCID: PMC10137844 DOI: 10.3390/diagnostics13081465] [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: 03/10/2023] [Revised: 04/09/2023] [Accepted: 04/16/2023] [Indexed: 05/17/2023] Open
Abstract
1. INTRODUCTION Multiple cytokines have been studied for their role in the propagation of the inflammatory process related to inflammatory bowel diseases (IBD), but the role of interleukin-4 remains controversial. The aim of this study was to evaluate the role of two IL-4 gene single nucleotide polymorphisms (SNPs) in disease susceptibility and phenotypic expression. 2. MATERIALS AND METHODS A group of 160 patients with IBD (86CD/74UC) and 160 healthy controls were genotyped for IL-4 rs2243250/-590C/T and rs2070874/-34C/T using real-time polymerase chain reaction with TaqMan assay. 3. RESULTS The analysis of IBD patients and controls revealed a significantly reduced frequency of the minor allele T of both SNPs in CD patients (p = 0.03, OR 0.55 and p = 0.02, OR 0.52) and for the entire IBD group (p = 0.01, OR 0.57 and p = 0.01, OR 0.55). Haplotype analysis identified the most frequent haplotype (rs2243250/rs2070874 CC) associated with a high risk for developing IBD (either UC or CD) (p = 0.003). IBD patients with extraintestinal manifestations had significantly increased frequency of the minor alleles T. We also found an association between the presence of allele C of rs2070874 and response to antiTNF treatment. 4. CONCLUSIONS This is the first study to investigate the IL-4 gene's relation to IBD susceptibility conducted in Romania. Both SNPs were found to be associated with disease susceptibility and phenotypic features, such as extraintestinal manifestations and response to antiTNF agents.
Collapse
Affiliation(s)
- Elena Mirela Ionescu
- Department of Gastroenterology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Gastroenterology, “Elias” Emergency University Hospital, 011461 Bucharest, Romania
| | - Andrei Ovidiu Olteanu
- Department of Gastroenterology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Gastroenterology, “Elias” Emergency University Hospital, 011461 Bucharest, Romania
| | - Cristian George Tieranu
- Department of Gastroenterology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Gastroenterology, “Elias” Emergency University Hospital, 011461 Bucharest, Romania
| | - Luis Ovidiu Popa
- Molecular Biology Department, “Grigore Antipa” National Museum of Natural History, 011341 Bucharest, Romania
| | - Silvia Ioana Andrei
- Clinic of Internal Medicine II, Thüringen-Kliniken “Georgius Agricola“, 07318 Saalfeld, Germany
| | - Carmen Monica Preda
- Department of Gastroenterology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Monica Irina Dutescu
- “Prof. Dr. C. T. Nicolau” National Institute of Blood Transfusion, 011155 Bucharest, Romania
| | - Mihai Bojinca
- Department of Rheumatology and Internal Medicine, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Ioana Tieranu
- Department of Pediatrics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Olivia Mihaela Popa
- Department of Immunology and Pathophysiology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
16
|
Yang R, Shan S, Shi J, Li H, An N, Li S, Cui K, Guo H, Li Z. Coprococcus eutactus, a Potent Probiotic, Alleviates Colitis via Acetate-Mediated IgA Response and Microbiota Restoration. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:3273-3284. [PMID: 36786768 DOI: 10.1021/acs.jafc.2c06697] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Inflammatory bowel disease (IBD) is a complex disease characterized by relapsing episodes of inflammation of the colonic mucosa. Research into IBD suggests that this disease condition is caused by alterations in resident mucosal bacterial populations. Our previous study showed that Coprococcus was significantly elevated during the improvement of IBD. Human metagenome database GMrepo also indicates Coprococcus, in particular, Coprococcus eutactus (C. eutactus), which was negatively associated with IBD. The current study implied the alleviated effects and mechanisms of C. eutactus on dextran sodium sulfate-induced experimental colitis mice. Gavage with C. eutactus-ameliorated acute colitis, as evidenced, relieved weight loss, decreased concentrations of proinflammatory cytokines TNF-α, IL-1β, and IL-6, and increased anti-inflammatory factors, IL-4, IL-5, and IL-10. In addition, C. eutactus enhanced the maturation of goblet cells and the expressions of mucins and restored the expressions of tight junction proteins such as claudin-1, occludin, and ZO-1. As a short-chain fatty acid-producing bacterium, C. eutactus mainly generates acetic acid. Interestingly, not only high levels of secretory immunoglobulin A (SIgA) but also increased IgA-producing plasma cells were observed in colitis mice during the administration of C. eutactus. Importantly, our data demonstrated that colonic SIgA is specifically coated on pathogens of Enterobacteriaceae. Owing to the selective binding effect of SIgA on microorganisms, the microbial diversity in the intestinal lumen and mucosa of C. eutactus-treated colitis mice was significantly restored, and the microbiota structure was remodeled. These findings provide substantial insight that C. eutactus as a promising probiotic can ameliorate colitis. In conclusion, our findings may deliver a novel approach to the prevention and biotherapy of IBD.
Collapse
Affiliation(s)
- Ruipeng Yang
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Shuhua Shan
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Jiangying Shi
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Hanqing Li
- School of Life Science, Shanxi University, Taiyuan 030006, China
| | - Ning An
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Songtao Li
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Kaili Cui
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Huiqin Guo
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Zhuoyu Li
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
17
|
Chen J, Wang Y, Shen L, Xiu Y, Wang B. Could IL-25 be a potential therapeutic target for intestinal inflammatory diseases? Cytokine Growth Factor Rev 2023; 69:43-50. [PMID: 35840510 DOI: 10.1016/j.cytogfr.2022.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/04/2022] [Indexed: 02/07/2023]
Abstract
As a member of the IL-17 cytokine family, IL-25 (also called IL-17E) induces and sustains type 2 immunity. IL-25, which is mainly produced by intestinal epithelial cells, has been gradually investigated in recent years for its function in intestinal inflammation but is not yet fully understood. This review summarizes the expression and function of IL-25 in the intestine, especially the progression of its regulatory role on type 2 immunity-related cells. Finally, we discuss the dual role of IL-25 based on inflammatory bowel disease to inform research on targeting IL-25 for the treatment of intestinal inflammatory diseases.
Collapse
Affiliation(s)
- Jie Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
| | - Yingshu Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lan Shen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yanfeng Xiu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Bing Wang
- Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
18
|
Jiang W, Wang Z, Zhang J, Li M. Interleukin 25 and its biological features and function in intestinal diseases. Cent Eur J Immunol 2023; 47:362-372. [PMID: 36817397 PMCID: PMC9901255 DOI: 10.5114/ceji.2022.124416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 01/13/2023] [Indexed: 02/04/2023] Open
Abstract
Interleukin 25 (IL-25), also known as IL-17E, is a member of the IL-17 cytokine family and an important regulator of the type 2 immune response. Accumulating evidence suggests that IL-25 interacts with diverse immune as well as non-immune cells and plays a rather complicated role in different backgrounds of multiple organs. IL-25 has been studied in the physiology and pathology of the intestine to some extent. With epithelial cells being an important source in the intestine, IL-25 plays a key role in intestinal immune responses and is associated with inappropriate allergic reactions, autoimmune diseases, and cancer tumorigenesis. In this review, we discuss the emerging comprehension of the biology of IL-25, as well as its cellular sources, targets, and signaling transduction. In particular, we discuss how IL-25 participates in the development of intestinal diseases including helminth infection, inflammatory bowel diseases, food allergy and colorectal cancer, as well as its underlying role in future therapy.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zehui Wang
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jun Zhang
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Minghui Li
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
19
|
Yang R, Shan S, An N, Liu F, Cui K, Shi J, Li H, Li Z. Polyphenols from foxtail millet bran ameliorate DSS-induced colitis by remodeling gut microbiome. Front Nutr 2022; 9:1030744. [PMID: 36479296 PMCID: PMC9719911 DOI: 10.3389/fnut.2022.1030744] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/01/2022] [Indexed: 01/11/2024] Open
Abstract
INTRODUCTION Polyphenols from plants possess the anti-inflammatory and gut microbiota modulated properties. Foxtail millet (Setaria italica L., FM) has potential medical and nutritional functions because of rich phenolic and other phytochemical components. METHODS Here, the study explored the effects of bound polyphenol of inner shell (BPIS) from FM bran on dextran sodium sulfate (DSS)-induced experimental colitis mice. RESULTS Results showed that BPIS administration effectively relieved the weight loss, decreased disease active index (DAI) scores, restrained the secretion of pro-inflammatory cytokines TNF-α, IL-6 and IL-1β, increased anti-inflammatory cytokines IL-10, IL-4, IL-5. BPIS prevented gut barrier damage by enhancing tight junction proteins Claudin1, ZO-1 and Occludin, increasing the number of goblet cells and facilitating the gene expressions of mucin family. In addition, BPIS restored the gut microbiota composition and increased the relative abundance of commensal bacteria such as Lachnospiraceae and Rikenellaceae and restrained the growth of S24-7 and Staphylococcaceae. Concentrations of short-chain-fatty acids (SCFAs) generated by gut microbiota were elevated in BPIS treated colitis mice. CONCLUSION These data suggest that BPIS effectively ameliorates DSS-induced colitis by preventing intestinal barrier damage and promoting gut microbiota community.
Collapse
Affiliation(s)
- Ruipeng Yang
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, China
| | - Shuhua Shan
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, China
| | - Ning An
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, China
| | - Fengming Liu
- School of Life Science, Shanxi University, Taiyuan, China
| | - Kaili Cui
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, China
| | - Jiangying Shi
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, China
| | - Hanqing Li
- School of Life Science, Shanxi University, Taiyuan, China
| | - Zhuoyu Li
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, China
| |
Collapse
|
20
|
Tai M, Shi H, Wang H, Ma X, Gao M, Chang Q, Li F, Zeng Q, Shi Y, Guo Y. Pilot study of peripheral blood chemokines as biomarkers for atrial fibrillation-related thromboembolism and bleeding in elderly patients. Front Public Health 2022; 10:844087. [PMID: 36211709 PMCID: PMC9538109 DOI: 10.3389/fpubh.2022.844087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 08/26/2022] [Indexed: 01/21/2023] Open
Abstract
Background The scoring systems currently used to identify the potential for thrombosis and bleeding events in high-risk atrial fibrillation patients have certain limitations. The aim of this pilot study was to identify inflammatory chemokines with potential utility as sensitive biomarkers for the risk of thrombosis and bleeding in elderly patients with non-valvular atrial fibrillation. Methods From January 1, 2014, to December 31, 2017, 200 consecutive elderly patients with atrial fibrillation (average age: 87.6 ± 7.7 years) were enrolled and followed up for 2 years to observe thromboembolic (arterial and venous) and bleeding events. Serum was collected upon enrollment, and the baseline levels of 27 chemokines were analyzed. During the 2-year follow-up, 12 patients were lost to follow-up. Among the 188 patients, there were 32 cases (17.0%) of AF-related thrombosis, 36 cases (19.1%) of arterial thrombosis, and 35 cases (18.6%) of major bleeding events. Results Among 188 patients, 30 patients without clinical events (control group), 23 with arterial thrombosis, 15 with atrial fibrillation-related venous thromboembolism, and 12 with major bleeding were selected and randomly matched to compare chemokine levels. The baseline levels of interleukin-6, interleukin-10, vascular cell adhesion molecule-1, chemokine C-C-motif ligand, B-lymphocyte chemoattractant 1, interleukin-4, E-selectin, fractalkine, C-X-C motif chemokine 12, and granulocyte chemotactic protein 2 were found to differ statistically among the four groups (p < 0.05). Compared with that in the control group, the level of interleukin-4 in patients with atrial fibrillation-related thrombosis, arterial thrombosis, or major bleeding increased by 53-fold (0.53 vs. 0.01 pg/ml), 17-fold (0.17 vs. 0.01 pg/ml), and 19-fold (0.19 vs. 0.01 pg/ml), respectively. Compared with that in the control group, the level of interleukin-6 in patients with arterial thrombosis increased by six-fold (39.78 vs. 4.98 pg/ml). Conclusions Among elderly patients with atrial fibrillation at high risk of thromboembolism and bleeding, the baseline levels of interleukin-6, interleukin-4, and E-selectin were significantly increased in those that experienced thrombosis and bleeding events during the 2-year follow-up, indicating that these chemokines may serve as potential biomarkers for an increased risk of thrombosis and bleeding in this population. Clinical trial registration number ChiCTR-OCH-13003479.
Collapse
Affiliation(s)
- Meihui Tai
- Chinese PLA Medical College, Pulmonary Vessel and Thrombotic Disease, Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Haiyan Shi
- Department of Gastroenterology, Second Medical Center, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Hao Wang
- Department of Cardiology, Second Medical Center, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Xiao Ma
- Department of Cardiology, Second Medical Center, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Meng Gao
- Department of Cardiology, Second Medical Center, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Qing Chang
- Department of Cardiology, Second Medical Center, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Fang Li
- Department of Gastroenterology, Second Medical Center, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Qiang Zeng
- Health Management Institute, Second Medical Center, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Yang Shi
- Health Management Institute, Second Medical Center, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Yutao Guo
- Chinese PLA Medical College, Pulmonary Vessel and Thrombotic Disease, Sixth Medical Center, Chinese PLA General Hospital, Beijing, China,*Correspondence: Yutao Guo
| |
Collapse
|
21
|
Shi W, Xu N, Wang X, Vallée I, Liu M, Liu X. Helminth Therapy for Immune-Mediated Inflammatory Diseases: Current and Future Perspectives. J Inflamm Res 2022; 15:475-491. [PMID: 35087284 PMCID: PMC8789313 DOI: 10.2147/jir.s348079] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 01/11/2022] [Indexed: 12/17/2022] Open
Affiliation(s)
- Wenjie Shi
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, People’s Republic of China
| | - Ning Xu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, People’s Republic of China
| | - Xuelin Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, People’s Republic of China
| | - Isabelle Vallée
- UMR BIPAR, Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, Laboratoire de Santé Animale, Maisons-Alfort, France
| | - Mingyuan Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, People’s Republic of China
| | - Xiaolei Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, People’s Republic of China
- Correspondence: Xiaolei Liu; Mingyuan Liu, Tel +86-15943092280; +86-13019125996, Email ;
| |
Collapse
|