1
|
Carneiro P, Vicente MM, Leite MI, Santos ME, Pinho SS, Fernandes Â. The role of N-glycans in regulatory T cells in autoimmunity. Autoimmun Rev 2025; 24:103791. [PMID: 40043894 DOI: 10.1016/j.autrev.2025.103791] [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: 12/04/2024] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 05/17/2025]
Abstract
Regulatory T cells (Tregs) have a key role in the maintenance of immune tolerance and in the prevention of autoimmunity. Recent studies have shown an association between decreased Treg frequency and a deficient suppressive activity with the development of many autoimmune diseases. Although glycosylation, which consists in the addition of glycans to proteins and lipids on the cell surface, is recognized as a critical modification for T cell development and function, the relevance of glycans in Treg biology and activity, as well as their impact in the immunopathogenesis of autoimmune diseases, deserves more attention, as it is far from being fully understood. This review discusses the biological impact of N-glycans in Treg biology, highlighting their potential to uncover novel pathogenic mechanisms in autoimmunity and new targets for promising therapeutic approaches with clinical applications in autoimmune disease patients.
Collapse
Affiliation(s)
- Pedro Carneiro
- i3s - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
| | - Manuel M Vicente
- i3s - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal; Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Maria Isabel Leite
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Maria Ernestina Santos
- CHUdSA - Centro Hospitalar Universitário de Santo António, Department of Neurology, Porto, Portugal
| | - Salomé S Pinho
- i3s - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal; ICBAS, School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal.
| | - Ângela Fernandes
- i3s - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal.
| |
Collapse
|
2
|
Yue S, Gong L, Tan Y, Zhang X, Liao F. IsoalloLCA-intervened regulatory T cell exosomes alleviate inflammatory bowel disease by inhibiting NF-κB-associated inflammation in intestinal epithelial cells. Int Immunopharmacol 2025; 154:114501. [PMID: 40174336 DOI: 10.1016/j.intimp.2025.114501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/11/2025] [Accepted: 03/14/2025] [Indexed: 04/04/2025]
Abstract
Regulatory T cells (Tregs) are the principal immune cells that exert anti-inflammatory effects within the organism. Their exosomes exhibit therapeutic efficacy across a broad spectrum of diseases owing to their high stability, low immunogenicity, and substantial penetration capacity. Recent research have indicated that isoallolithocholic acid (isoalloLCA), a metabolite associated with bile acid metabolism, may enhance Treg activity by upregulating forkhead box protein3 (Foxp3) expression. Hence, metabolite-based strategies for reinforcing Tregs may offer novel intervention options for treating related diseases. In this study, tumor necrosis factor (TNF)-α and dextran sulfate sodium (DSS) were employed to establish cellular and animal models of inflammatory bowel disease (IBD), further evaluating the therapeutic efficacy of isoalloLCA-intervened regulatory T cell exosomes (isoalloLCA-Exo) within this model. Our findings demonstrated that isoalloLCA-Exo effectively inhibit colitis progression in a murine model, as indicated by reduced inflammation, decreased apoptosis of intestinal epithelial cells, and improved intestinal barrier function. Furthermore, in vitro analyses elucidated the molecular mechanisms underlying the anti-inflammatory effects of isoalloLCA-Exo, revealing that the intervention effectively reversed TNF-α-induced inflammation and apoptosis in intestinal epithelial cells by modulating the NF-κB pathway. In conclusion, isoalloLCA-Exo can decelerate inflammatory bowel disease progression and suppress inflammatory response in intestinal epithelial cells by inhibiting NF-κB pathway. Notably, isoalloLCA-Exo exhibit superior efficacy to the traditional drug mesalazine and conventional treg exosome(NC-Exo). These findings have significant implications for optimizing Treg-derived exosome-based therapies for inflammation-related diseases.
Collapse
Affiliation(s)
- Simei Yue
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lingjiao Gong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yulin Tan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaodan Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fei Liao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Wuhan University Shenzhen Research Institute, Shenzhen, Guangdong 518000, China; Central Laboratory of Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
3
|
Maeda E, Okimura H, Tanaka Y, Fujii M, Tarumi Y, Kataoka H, Koshiba A, Hamaguchi M, Fukui M, Mori T, Kitawaki J. Adoptive transfer of regulatory T cells inhibits the progression of endometriosis-like lesions in regulatory T-cell-depleted mice. Hum Reprod 2025; 40:926-937. [PMID: 40180333 DOI: 10.1093/humrep/deaf054] [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: 09/26/2024] [Revised: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
STUDY QUESTION Does the restoration of regulatory T cells (Tregs) suppress the progression of endometriosis? SUMMARY ANSWER Adoptive transfer of Tregs suppresses the progression of endometriosis and reduces the levels of helper T (Th)-cell-related and proinflammatory cytokines in mice. WHAT IS KNOWN ALREADY Endometriosis is a chronic inflammatory gynecological disease, which involves multiple immune components. Activated Treg counts decrease in the endometrioma and endometrium of patients with endometriosis, and depletion of Tregs exacerbates endometriosis in mice. STUDY DESIGN, SIZE, DURATION We evaluated the effects of adoptive transfer of Tregs on the progression of endometriosis in mice. We used Foxp3tm3Ayr/J (Foxp3DTR) mice with temporarily ablated Tregs by injecting diphtheria toxin to develop an endometriosis model, which was generated by ovariectomy, estradiol administration and transplantation of uterine fragments from donor mice. Foxp3DTR mice were randomly divided into Treg adoptive transfer (n = 12) and control (n = 11) groups. Tregs were isolated from lymph nodes and spleens of wild-type (WT) mice and were adoptively transferred into mice that were temporarily Treg-depleted. Control mice were injected with vehicle. Treg adoptive transfer was performed on the day of uterine implantation, and a second adoptive transfer was performed after 14 days. Mice were euthanized 28 days after uterine implantation, and blood, peritoneal fluid, spleen, and endometriosis-like lesion samples were collected. PARTICIPANTS/MATERIALS, SETTING, METHODS Foxp3DTR mice were intravenously injected with Tregs isolated from WT mice. The number, total weight, and total volume of the endometriosis-like lesions were evaluated on Day 28 following implantation of uterine fragments. The proportion of Tregs in endometriosis-like lesions, ascites, and peripheral blood was analyzed by flow cytometry. Inflammation in lesions and serum was examined using real-time PCR and ELISA. MAIN RESULTS AND THE ROLE OF CHANCE Injection of Tregs increased their total count and decreased the number (P < 0.0001), weight (P = 0.0021), and volume (P = 0.0010) of endometriosis-like lesions in Foxp3DTR Treg-depleted mice. Furthermore, injection of Tregs decreased the mRNA expression of Th 1-, 2-, and 17-related cytokines, including interferon gamma (P = 0.0101), interleukin (IL)-4 (P = 0.0051), and IL-17 (P = 0.0177), as well as the levels of the proinflammatory cytokine IL-6 (P = 0.0002), in endometriosis-like lesions of Foxp3DTR Treg-depleted mice. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Treg-related immune mechanisms in mice may not precisely reflect those in humans. WIDER IMPLICATIONS OF THE FINDINGS Restoration of Tregs may be a useful therapeutic strategy for inhibiting the progression of endometriosis in cases where the decrease in the Treg population is an exacerbating factor. STUDY FUNDING/COMPETING INTEREST(S) This study was partially supported by the Grants-in-Aid for Scientific Research (grant numbers 18K16808 and 20K22983) from the Ministry of Education, Culture, Sports, Science, and Technology, Japan. The sponsor had no role in the study design, collection, analysis and interpretation of data, writing of the report, and decision to submit the article for publication. The authors have no conflicts of interest to disclose.
Collapse
Affiliation(s)
- Eiko Maeda
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroyuki Okimura
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yukiko Tanaka
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Maya Fujii
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yosuke Tarumi
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hisashi Kataoka
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akemi Koshiba
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masahide Hamaguchi
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Taisuke Mori
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Jo Kitawaki
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
4
|
Wang L, Shao L, Gao YC, Liu J, Li XD, Zhou J, Li SF, Song YL, Liu B, Zhang W, Huang WH. Panax notoginseng Saponins Alleviate Inflammatory Bowel Disease via Alteration of Gut Microbiota-Bile Acid Metabolism. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2025; 53:567-596. [PMID: 40165428 DOI: 10.1142/s0192415x25500223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Bile acid metabolism mediated by gut microbiota is significantly related to immunity regulation that plays an important role in the development and treatment of inflammatory bowel disease (IBD). Our previous study has demonstrated that Panax notoginseng saponins (PNS) alleviate colitis due to the regulation of T helper 17/Regulatory T cells (Th17/Treg) balance via gut microbiota. However, the effects and mechanism of PNS on colitis pertinent to bile acid metabolism mediated by gut microbiota remain elusive. This study aims to investigate the anti-colitis mechanism of PNS by regulating the Th17/Treg balance pertinent to gut microbiota-bile acid metabolism. Results showed that PNS significantly decreased the relative abundance of Allobaculum, Dubosiella, Muribaculum, and Alistipes, and up-regulated the relative contents of conjugated bile acids, such as TCA and TCDCA. Fecal microbiota transplantation (FMT) showed that the gut microbiota remodeled by PNS had a regulatory effect on bile acid metabolism, and up-regulated the relative contents of TCA and TCDCA, which alleviated IBD and promoted Treg cell expression in vivo and in vitro. Taken together, PNS could reshape the profiling of gut microbiota to generate more TCA and TCDCA, which improve the balance of Th17/Treg to exert anti-IBD effects.
Collapse
Affiliation(s)
- Lin Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics and National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan 410008, P. R. China
| | - Li Shao
- Department of Pharmacognosy, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410128, P. R. China
| | - Yong-Chao Gao
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics and National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan 410008, P. R. China
| | - Jing Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics and National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan 410008, P. R. China
| | - Xu-Dong Li
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics and National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan 410008, P. R. China
| | - Jie Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics and National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan 410008, P. R. China
| | - Shuang-Feng Li
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics and National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan 410008, P. R. China
| | - Yue-Lin Song
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, P. R. China
| | - Bo Liu
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P. R. China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics and National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan 410008, P. R. China
| | - Wei-Hua Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics and National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan 410008, P. R. China
| |
Collapse
|
5
|
Sun X, Yun L, Xie K, Liu R, Ren X, Zeng B, Cao X, Li Z, Zhou G, Liu B, Peng L, Yuan L. Probiotic Bacillus pumilus LV149 enhances gut repair, modulates microbiota, and alters transcriptome in DSS-induced colitis mice. Front Microbiol 2025; 15:1507979. [PMID: 39845056 PMCID: PMC11753000 DOI: 10.3389/fmicb.2024.1507979] [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: 10/08/2024] [Accepted: 12/11/2024] [Indexed: 01/24/2025] Open
Abstract
Purpose Gut microbiota dysbiosis significantly impacts ulcerative colitis (UC) progression and exacerbation. Probiotics show promise in UC management. This study evaluated the effects of different doses of Bacillus pumilus LV149, an aquatic-derived probiotic, on gut injury repair in male C57BL/6 mice with dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) and investigated the underlying mechanisms. Methods UC was induced by allowing mice free access to a 3% DSS solution for 7 days, with concurrent daily oral gavage of either a low (LV149-L, 1 × 108 CFU/day/mouse) or high (LV149-H, 1 × 109 CFU/day/mouse) dose of LV149. The effects were assessed through physiological parameters, intestinal barrier integrity, inflammation, gut microbiota composition, and transcriptomic changes. Results LV149 significantly improved pathological symptoms, including weight loss and disease activity index (DAI), and reduced colon shortening in a dose-dependent manner and inflammatory damage. The intervention also restored gut barrier function by upregulating mucins, goblet cell counts, and tight junction proteins (ZO-1, occludin, and claudin-1) in colonic tissue, along with reducing serum lipopolysaccharide (LPS) levels. Notably, only the LV149-H significantly decreased the expression of pro-inflammatory cytokines TNF-α, IL-1β, and IL-6, while both doses increased the expression of the anti-inflammatory cytokine IL-10 in a dose-dependent in colonic tissue. LV149 further modulated the gut microbiota, increasing beneficial bacteria and reducing pathogenic populations. Transcriptomic analysis indicated that LV149-L may exert gut repair effects via the IL-17 signaling pathway, whereas LV149-H appears to act through the JAK-STAT signaling pathway. Conclusion This study demonstrated that LV149, particularly at a higher dose, effectively mitigated DSS-induced colonic injury by modulating gut microbiota, enhancing gut barrier integrity, and reducing inflammation. The dose-dependent effects underscored LV149-H's potential as a therapeutic agent for UC due to its stronger anti-inflammatory properties and gut-protective effects.
Collapse
Affiliation(s)
- Xinyu Sun
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Long Yun
- Huzhou Key Laboratory of Translational Medicine, First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Keming Xie
- Medical College of Jiaying University, Jiaying University, Meizhou, China
| | - Renhui Liu
- School of Sports Medicine, Wuhan Sports University, Wuhan, China
| | - Xinyue Ren
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Bokun Zeng
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xudong Cao
- Department of Chemical and Biological Engineering, University of Ottawa, Ottawa, ON, Canada
| | - Zhi Li
- Key Laboratory of Aquacultural Biotechnology Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, China
| | - Guihao Zhou
- Division of Medicine, University College London, London, United Kingdom
| | - Bang Liu
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Luo Peng
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS), Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Lihong Yuan
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
6
|
Nicolò S, Faggiani I, Errico C, D'Amico F, Parigi TL, Danese S, Ungaro F. Translational characterization of immune pathways in inflammatory bowel disease: insights for targeted treatments. Expert Rev Clin Immunol 2025; 21:55-72. [PMID: 39313992 DOI: 10.1080/1744666x.2024.2400300] [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: 01/15/2024] [Accepted: 08/30/2024] [Indexed: 09/25/2024]
Abstract
INTRODUCTION The pathogenesis of inflammatory bowel disease (IBD) involves the dysregulation of multiple inflammatory pathways. The understanding of these mechanisms allows their selective targeting for therapeutic purposes. The discovery of Tumor Necrosis Factor-alpha's (TNF-α) role in mucosal inflammation ushered an exciting new era of drug development which now comprises agents targeting multiple pro-inflammatory signaling pathways, integrins, and leukocyte trafficking regulators. AREA COVERED This review provides an overview of the main molecular players of IBD, their translation into therapeutic targets and the successful development of the advanced agents modulating them. We combine basic science with clinical trials data to present a critical review of both the successful and failed drug development programs. A PubMed literature search was conducted to delve into the available literature and clinical trials. EXPERT OPINION The treatment landscape for IBD has rapidly expanded, particularly with the development of biologics targeting TNF-α, integrins, and S1P modulators, as well as newer agents such as IL-12/IL-23 inhibitors and JAK inhibitors, offering robust efficacy and safety profiles. However, challenges persist in understanding and effectively treating difficult-to-treat IBD, highlighting the need for continued research to uncover novel therapeutic targets and optimize patient outcomes.
Collapse
Affiliation(s)
- Sabrina Nicolò
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Ilaria Faggiani
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Carmela Errico
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Ferdinando D'Amico
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Tommaso Lorenzo Parigi
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Silvio Danese
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Federica Ungaro
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
7
|
Vitali R, Novelli F, Palone F, Cucchiara S, Stronati L, Pioli C. PARP1 inactivation increases regulatory T / Th17 cell proportion in intestinal inflammation. Role of HMGB1. Immunol Lett 2024; 270:106912. [PMID: 39237041 DOI: 10.1016/j.imlet.2024.106912] [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: 12/29/2023] [Revised: 08/12/2024] [Accepted: 09/01/2024] [Indexed: 09/07/2024]
Abstract
Inflammatory bowel diseases (IBD) are chronic relapsing disorders with increasing prevalence. Knowledge gaps still limit the possibility to develop more specific and effective therapies. Using a dextran sodium sulfate colitis mouse model, we found that inflammation increased the total number and altered the frequencies of leukocytes within colon mesenteric lymph nodes (cMLNs). Although the inflammation reduced the frequency of regulatory T (Treg) cells, their absolute numbers were increased. Increased frequency of colitogenic Th17 cells was also observed. Noteworthy, untreated mice lacking Poly(ADP-ribose)-Polimerase-1 functional gene (PARP-1KO) displayed higher frequency of Treg cells and lower percentage of Th17 cells in cMLNs. In colitic PARP-1KO mice the inflammation driven expansion of the Foxp3 Treg population was more pronounced than in WT mice. Conversely, colitis increased Th17 cells to a lower extent in PARP-1KO mice compared with WT mice, resulting in a more protective Treg/Th17 cell ratio. Consequently PARP-1KO mice developed less severe colitis with reduced expression of inflammatory cytokines. In ex vivo experiments PARP-1KO and WT CD11c dendritic cells (DCs) promoted naïve CD4 T cell differentiation differently, the former sustaining more efficiently the generation of Treg cells, the latter that of Th17 cells. Addition of HMGB1 B box or of dipotassium glycyrrhizate, which sequesters extracellular HMGB1, revealed a role for this alarmin in the regulation exerted by PARP-1 on the stimulating vs. tolerogenic function of DCs during colitis. Moreover, a higher percentage of CD11c DC from PARP-1KO mice expressed CD103, a marker associated with the ability of DC to induce Treg cells, compared with WT DC. Conversely, PARP-1KO DC were including a reduced percentage of CX3CR1+ DC, described to induce Th17 cells. These findings were observed in both splenic and colon lamina propria DC.
Collapse
Affiliation(s)
| | | | | | - Salvatore Cucchiara
- Department of Maternal Infantile and Urological Sciences, Sapienza University, Rome, Italy
| | - Laura Stronati
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | | |
Collapse
|
8
|
van Lingen E, Nooij S, Terveer EM, Crossette E, Prince AL, Bhattarai SK, Watson A, Galazzo G, Menon R, Szabady RL, Bucci V, Norman JM, van der Woude CJ, van der Marel S, Verspaget HW, van der Meulen-de Jong AE, Keller JJ. Faecal Microbiota Transplantation Engraftment After Budesonide or Placebo in Patients With Active Ulcerative Colitis Using Pre-selected Donors: A Randomized Pilot Study. J Crohns Colitis 2024; 18:1381-1393. [PMID: 38572716 PMCID: PMC11369067 DOI: 10.1093/ecco-jcc/jjae043] [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: 06/01/2023] [Revised: 10/30/2023] [Indexed: 04/05/2024]
Abstract
BACKGROUND Faecal microbiota transplantation [FMT] shows some efficacy in treating patients with ulcerative colitis [UC], although variability has been observed among donors and treatment regimens. We investigated the effect of FMT using rationally selected donors after pretreatment with budesonide or placebo in active UC. METHODS Patients ≥18 years old with mild to moderate active UC were randomly assigned to 3 weeks of budesonide [9 mg] or placebo followed by 4-weekly infusions of a donor faeces suspension. Two donors were selected based on microbiota composition, regulatory T cell induction and short-chain fatty acid production in mice. The primary endpoint was engraftment of donor microbiota after FMT. In addition, clinical efficacy was assessed. RESULTS In total, 24 patients were enrolled. Pretreatment with budesonide did not increase donor microbiota engraftment [p = 0.56] nor clinical response, and engraftment was not associated with clinical response. At week 14, 10/24 [42%] patients achieved [partial] remission. Remarkably, patients treated with FMT suspensions from one donor were associated with clinical response [80% of responders, p < 0.05] but had lower overall engraftment of donor microbiota. Furthermore, differences in the taxonomic composition of the donors and the engraftment of certain taxa were associated with clinical response. CONCLUSION In this small study, pretreatment with budesonide did not significantly influence engraftment or clinical response after FMT. However, clinical response appeared to be donor-dependent. Response to FMT may be related to transfer of specific strains instead of overall engraftment, demonstrating the need to characterize mechanisms of actions of strains that maximize therapeutic benefit in UC.
Collapse
Affiliation(s)
- Emilie van Lingen
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sam Nooij
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Elisabeth M Terveer
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Shakti K Bhattarai
- University of Massachusetts Chan Medical School, Department of Microbiology and Physiological Systems, Worcester, MA, USA
| | | | | | | | - Rose L Szabady
- Vedanta Biosciences, Cambridge, MA, USA
- Ferring Pharmaceuticals, San Diego, CA, USA
| | - Vanni Bucci
- University of Massachusetts Chan Medical School, Department of Microbiology and Physiological Systems, Worcester, MA, USA
| | | | - C Janneke van der Woude
- Department of Gastroenterology and Hepatology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sander van der Marel
- Department of Gastroenterology and Hepatology, Haaglanden Medisch Centrum, den Haag, The Netherlands
| | - Hein W Verspaget
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Josbert J Keller
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Gastroenterology and Hepatology, Haaglanden Medisch Centrum, den Haag, The Netherlands
| |
Collapse
|
9
|
Zhang LL, Xu JY, Xing Y, Wu P, Jin YW, Wei W, Zhao L, Yang J, Chen GC, Qin LQ. Lactobacillus rhamnosus GG alleviates radiation-induced intestinal injury by modulating intestinal immunity and remodeling gut microbiota. Microbiol Res 2024; 286:127821. [PMID: 38941923 DOI: 10.1016/j.micres.2024.127821] [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: 05/06/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 06/30/2024]
Abstract
Radiation injury to the intestine is one of the most common complications in patients undergoing abdominal or pelvic cavity radiotherapy. In this study, we investigated the potential protective effect of Lactobacillus rhamnosus GG (LGG) on radiation-induced intestinal injury and its underlying mechanisms. Mice were assigned to a control group, a 10 Gy total abdominal irradiation (TAI) group, or a group pretreated with 108 CFU LGG for three days before TAI. Small intestine and gut microbiota were analyzed 3.5 days post-exposure. LGG intervention improved intestinal structure, reduced jejunal DNA damage, and inhibited the inflammatory cGAS/STING pathway. Furthermore, LGG reduced M1 proinflammatory macrophage and CD8+ T cell infiltration, restoring the balance between Th17 and Treg cells in the inflamed jejunum. LGG also partially restored the gut microbiota. These findings suggest the possible therapeutic radioprotective effect of probiotics LGG in alleviating radiation-induced intestinal injury by maintaining immune homeostasis and reshaping gut microbiota.
Collapse
Affiliation(s)
- Li-Li Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, 199 Ren'ai Road, Suzhou 215123, China
| | - Jia-Ying Xu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, 199 Ren'ai Road, Suzhou 215123, China
| | - Yifei Xing
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, 199 Ren'ai Road, Suzhou 215123, China
| | - Pengcheng Wu
- Zhangjiagang Center for Disease Control and Prevention, 18 Zhizhong Road, Zhangjiagang 215600, China
| | - Yi-Wen Jin
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, 199 Ren'ai Road, Suzhou 215123, China
| | - Wei Wei
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, 199 Ren'ai Road, Suzhou 215123, China
| | - Lin Zhao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, 199 Ren'ai Road, Suzhou 215123, China
| | - Jing Yang
- Department of Clinical Nutrition, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, China
| | - Guo-Chong Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, 199 Ren'ai Road, Suzhou 215123, China
| | - Li-Qiang Qin
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, 199 Ren'ai Road, Suzhou 215123, China.
| |
Collapse
|
10
|
Zhang D, Jiang L, Yu F, Yan P, Liu Y, Wu Y, Yang X. PepT1-targeted nanodrug based on co-assembly of anti-inflammatory peptide and immunosuppressant for combined treatment of acute and chronic DSS-induced ColitiS. Front Pharmacol 2024; 15:1442876. [PMID: 39211778 PMCID: PMC11357942 DOI: 10.3389/fphar.2024.1442876] [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: 06/03/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
INTRODUCTION Inflammatory bowel disease (IBD), as a chronic and recurrent inflammatory bowel diseases with limited therapeutic outcomes, is characterized by immune disorders and intestinal barrier dysfunction. Currently, the most medications used to cure IBD in clinic just temporarily induce and maintain remission with poor response rates and limited outcomes. Therefore, it is urgently necessary to develop an appropriate therapeutic candidate with preferable efficacy and less adverse reaction for curing IBD. METHODS Five groups of mice were utilized: control that received saline, DSS group (mice received 2.5% DSS or 4% DSS), KPV group (mice received KPV), FK506 group (mice received FK506) and NPs groups (mice received NPs). The effect of NP on the inflammatory factors of macrophage was evaluated using CCK-8, quantitative polymerase chain reaction (PCR), Elisa and Western blot (WB). Immunofluorescent staining revealed the targeting relationship between NP and Petp-1. Immunohistochemistry staining showed the effect of NP on tight junction proteins. Moreover, in vivo animal experiments confirmed that NPs reduced inflammatory levels in IBD. RESULTS AND DISCUSSION After administering with NPs, mice with DSS-induced acute or chronic colitis exhibited significant improvement in body weight, colon length, and disease activity index, decreased the level of the factors associated with oxidative stress (MPO, NO and ROS) and the inflammatory cytokines (TNF-α, IL-1β and IL-6), which implied that NPs could ameliorate murine colitis effectively. Furthermore, treating by NPs revealed a notable reduction of the expressions of CD68 and CD3, restoring the expression levels of tight junction proteins (Claudin-5, Occludin-1, and ZO-1) were significantly restored, surpassing those observed in the KPV and FK506 groups. which indicated that NPs can reduce inflammation and enhance epithelial barrier integrity by decreasing the infiltration of macrophages and T-lymphocytes. Collectively, those results demonstrated the effectively therapeutic outcome after using NPs in both acute and chronic colitis, suggesting that the newly co-assembled of NPs can be as a potential therapeutic candidate for colitis.
Collapse
Affiliation(s)
- Daifang Zhang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Longqi Jiang
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Vascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Fengxu Yu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Pijun Yan
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yong Liu
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Vascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Ya Wu
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Vascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xi Yang
- Department of Vascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
11
|
Din MAU, Wan A, Chu Y, Zhou J, Yan Y, Xu Z. Therapeutic role of extracellular vesicles from human umbilical cord mesenchymal stem cells and their wide therapeutic implications in inflammatory bowel disease and other inflammatory disorder. Front Med (Lausanne) 2024; 11:1406547. [PMID: 39139783 PMCID: PMC11319305 DOI: 10.3389/fmed.2024.1406547] [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: 03/25/2024] [Accepted: 07/18/2024] [Indexed: 08/15/2024] Open
Abstract
The chronic immune-mediated inflammatory condition known as inflammatory bowel disease (IBD) significantly affects the gastrointestinal system. While the precise etiology of IBD remains elusive, extensive research suggests that a range of pathophysiological pathways and immunopathological mechanisms may significantly contribute as potential factors. Mesenchymal stem cells (MSCs) have shown significant potential in the development of novel therapeutic approaches for various medical conditions. However, some MSCs have been found to exhibit tumorigenic characteristics, which limit their potential for medical treatments. The extracellular vesicles (EVs), paracrine factors play a crucial role in the therapeutic benefits conferred by MSCs. The EVs consist of proteins, microRNAs, and lipids, and are instrumental in facilitating intercellular communication. Due to the ease of maintenance, and decreased immunogenicity, tumorigenicity the EVs have become a new and exciting option for whole cell treatment. This review comprehensively assesses recent preclinical research on human umbilical cord mesenchymal stem cell (hUC-MSC)-derived EVs as a potential IBD therapy. It comprehensively addresses key aspects of various conditions, including diabetes, cancer, dermal injuries, neurological disorders, cardiovascular issues, liver and kidney diseases, and bone-related afflictions.
Collapse
Affiliation(s)
- Muhammad Azhar Ud Din
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine Jiangsu University, Zhenjiang, China
| | | | - Ying Chu
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China
| | - Jing Zhou
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China
| | - Yongmin Yan
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China
| | - Zhiliang Xu
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China
| |
Collapse
|
12
|
Honing DY, Luiten RM, Matos TR. Regulatory T Cell Dysfunction in Autoimmune Diseases. Int J Mol Sci 2024; 25:7171. [PMID: 39000278 PMCID: PMC11241405 DOI: 10.3390/ijms25137171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Regulatory T cells (Tregs), a suppressive subpopulation of T cells, are potent mediators of peripheral tolerance, responsible for immune homeostasis. Many autoimmune diseases exhibit disruptions in Treg function or quantity, resulting in an imbalance between protective and pathogenic immune cells. Selective expansion or manipulation of Tregs is a promising therapeutic approach for autoimmune diseases. However, the extensive diversity of Treg subpopulations and the multiple approaches used for Treg identification leads to high complexity, making it difficult to develop a successful treatment capable of modulating Tregs. In this review, we describe the suppressive mechanisms, subpopulations, classification, and identification methodology for Tregs, and their role in the pathogenesis of autoimmune diseases.
Collapse
Affiliation(s)
- Dionne Y Honing
- Department of Dermatology, Netherlands Institute for Pigment Disorders, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, 1081 HV Amsterdam, The Netherlands
| | - Rosalie M Luiten
- Department of Dermatology, Netherlands Institute for Pigment Disorders, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, 1081 HV Amsterdam, The Netherlands
| | - Tiago R Matos
- Department of Dermatology, Netherlands Institute for Pigment Disorders, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Sanofi, 1105 BP Amsterdam, The Netherlands
| |
Collapse
|
13
|
Huang XL, Wu LN, Huang Q, Zhou Y, Qing L, Xiong F, Dong HP, Zhou TM, Wang KL, Liu J. Unraveling the mechanism of malancao in treating ulcerative colitis: A multi-omics approach. World J Clin Cases 2024; 12:3105-3122. [PMID: 38898844 PMCID: PMC11185383 DOI: 10.12998/wjcc.v12.i17.3105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/11/2024] [Accepted: 04/23/2024] [Indexed: 06/04/2024] Open
Abstract
BACKGROUND Malancao (MLC) is a traditional Chinese medicine with a long history of utilization in treating ulcerative colitis (UC). Nevertheless, the precise molecular mechanisms underlying its efficacy remain elusive. This study leveraged ultra-high-performance liquid chromatography coupled with exactive mass spectrometry (UHPLC-QE-MS), network pharmacology, molecular docking (MD), and gene microarray analysis to discern the bioactive constituents and the potential mechanism of action of MLC in UC management. AIM To determine the ingredients related to MLC for treatment of UC using multiple databases to obtain potential targets for fishing. METHODS This research employs UHPLC-QE-MS for the identification of bioactive compounds present in MLC plant samples. Furthermore, the study integrates the identified MLC compound-related targets with publicly available databases to elucidate common drug disease targets. Additionally, the R programming language is utilized to predict the central targets and molecular pathways that MLC may impact in the treatment of UC. Finally, MD are conducted using AutoDock Vina software to assess the affinity of bioactive components to the main targets and confirm their therapeutic potential. RESULTS Firstly, through a comprehensive analysis of UHPLC-QE-MS data and public database resources, we identified 146 drug-disease cross targets related to 11 bioactive components. The Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis highlighted that common disease drug targets are primarily involved in oxidative stress management, lipid metabolism, atherosclerosis, and other processes. They also affect AGE-RAGE and apoptosis signaling pathways. Secondly, by analyzing the differences in diseases, we identified key research targets. These core targets are related to 11 active substances, including active ingredients such as quercetin and luteolin. Finally, MD analysis revealed the stability of compound-protein binding, particularly between JUN-Luteolin, JUN-Quercetin, HSP90AA1-Wogonin, and HSP90AA1-Rhein. Therefore, this suggests that MLC may help alleviate intestinal inflammation in UC, restore abnormal lipid accumulation, and regulate the expression levels of core proteins in the intestine. CONCLUSION The utilization of MLC has demonstrated notable therapeutic efficacy in the management of UC by means of the compound target interaction pathway. The amalgamation of botanical resources, metabolomics, natural products, MD, and gene chip technology presents a propitious methodology for investigating therapeutic targets of herbal medicines and discerning novel bioactive constituents.
Collapse
Affiliation(s)
- Xing-Long Huang
- Hospital of Traditional Chinese Medicine in Qijiang District, Chongqing 401420, China
- Xing-Long Huang and Lu-Na Wu
| | - Lu-Na Wu
- Hospital of Traditional Chinese Medicine in Qijiang District, Chongqing 401420, China
- Xing-Long Huang and Lu-Na Wu
| | - Qin Huang
- Hospital of Traditional Chinese Medicine in Qijiang District, Chongqing 401420, China
| | - Yue Zhou
- Hospital of Traditional Chinese Medicine in Qijiang District, Chongqing 401420, China
| | - Lei Qing
- Qijiang Health Center for Maternal and Child Care, Chongqing 401420, China
| | - Feng Xiong
- Hospital of Traditional Chinese Medicine in Qijiang District, Chongqing 401420, China
| | - Hui-Ping Dong
- Hospital of Traditional Chinese Medicine in Qijiang District, Chongqing 401420, China
| | - Tai-Min Zhou
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, Guizhou Province, China
| | - Kai-Li Wang
- Hospital of Traditional Chinese Medicine in Qijiang District, Chongqing 401420, China
| | - Jue Liu
- Hospital of Traditional Chinese Medicine in Qijiang District, Chongqing 401420, China
| |
Collapse
|
14
|
Curvino EJ, Roe EF, Freire Haddad H, Anderson AR, Woodruff ME, Votaw NL, Segura T, Hale LP, Collier JH. Engaging natural antibody responses for the treatment of inflammatory bowel disease via phosphorylcholine-presenting nanofibres. Nat Biomed Eng 2024; 8:628-649. [PMID: 38012308 PMCID: PMC11128482 DOI: 10.1038/s41551-023-01139-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/17/2023] [Indexed: 11/29/2023]
Abstract
Inflammatory bowel disease lacks a long-lasting and broadly effective therapy. Here, by taking advantage of the anti-infection and anti-inflammatory properties of natural antibodies against the small-molecule epitope phosphorylcholine (PC), we show in multiple mouse models of colitis that immunization of the animals with self-assembling supramolecular peptide nanofibres bearing PC epitopes induced sustained levels of anti-PC antibodies that were both protective and therapeutic. The strength and type of immune responses elicited by the nanofibres could be controlled through the relative valency of PC epitopes and exogenous T-cell epitopes on the nanofibres and via the addition of the adjuvant CpG. The nanomaterial-assisted induction of the production of therapeutic antibodies may represent a durable therapy for inflammatory bowel disease.
Collapse
Affiliation(s)
| | - Emily F Roe
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Alexa R Anderson
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Mia E Woodruff
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nicole L Votaw
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Tatiana Segura
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Laura P Hale
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
15
|
Zhang B, Cheng Y, Jian Q, Xiang S, Xu Q, Wang C, Yang C, Lin J, Zheng C. Sishen Pill and its active phytochemicals in treating inflammatory bowel disease and colon cancer: an overview. Front Pharmacol 2024; 15:1375585. [PMID: 38650627 PMCID: PMC11033398 DOI: 10.3389/fphar.2024.1375585] [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: 01/24/2024] [Accepted: 03/21/2024] [Indexed: 04/25/2024] Open
Abstract
The incidence of inflammatory bowel disease (IBD) and the associated risk of colon cancer are increasing globally. Traditional Chinese medicine (TCM) treatment has unique advantages. The Sishen Pill, a common Chinese patented drug used to treat abdominal pain and diarrhea, consists mainly of Psoraleae Fructus, Myristicae Semen, Euodiae Fructus, and Schisandra Chinensis. Modern research has confirmed that Sishen Pill and its active secondary metabolites, such as psoralen, myristicin, evodiamine, and schisandrin, can improve intestinal inflammation and exert antitumor pharmacological effects. Common mechanisms in treating IBD and colon cancer mainly include regulating inflammation-related signaling pathways such as nuclear factor-kappa B, mitogen-activated protein kinase, phosphatidylinositol 3-kinase, NOD-like receptor heat protein domain-related protein 3, and wingless-type MMTV integration site family; NF-E2-related factor 2 and hypoxia-inducible factor 1α to inhibit oxidative stress; mitochondrial autophagy and endoplasmic reticulum stress; intestinal immune cell differentiation and function through the Janus kinase/signal transducer and activator of transcription pathway; and improving the gut microbiota and intestinal barrier. Overall, existing evidence suggests the potential of the Sishen pill to improve IBD and suppress inflammation-to-cancer transformation. However, large-scale randomized controlled clinical studies and research on the safety of these clinical applications are urgently required.
Collapse
Affiliation(s)
- Boxun Zhang
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yingying Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qin Jian
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sirui Xiang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qi Xu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuchu Wang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuan Yang
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Junzhi Lin
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuan Zheng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Sichuan Provincial Engineering Research Center of Innovative Re-development of Famous Classical Formulas, Tianfu TCM Innovation Harbour, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
16
|
Alqudah A, Qnais E, Gammoh O, Bseiso Y, Wedyan M, Alqudah M, Hatahet T. Cirsimaritin Alleviates Dextran Sodium Sulfate-Induced Acute Colitis in Experimental Animals: A Therapeutic Approach for Inflammatory Bowel Disease. Prev Nutr Food Sci 2024; 29:31-39. [PMID: 38576881 PMCID: PMC10987388 DOI: 10.3746/pnf.2024.29.1.31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 04/06/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic disease that affects the entire digestive tract. IBD can be classified as ulcerative colitis or Crohn's disease. The key symptoms of IBD include the emergence of abscesses or pustules, pronounced abdominal discomfort, diarrhea, fistulas, and intestinal narrowing, all of which can greatly affect a patient's daily well-being. Several factors, including bacterial infections, immune response irregularities, and changes in the intestinal milieu, can contribute to the onset of IBD. The aim of this study was investigating the role of cirsimaritin in reducing the severity of colitis in animal model. To induce colitis in laboratory Swiss albino mice, a 4% dextran sulfate sodium (DSS) concoction was provided in their hydration source for a duration of six days. Before the onset of colitis, mice were treated with cirsimaritin (10 mg/kg) once daily to evaluate its potential treatment effects against DSS-induced inflammation. The results showed that 10 mg/kg of cirsimaritin decreased colitis severity (P<0.05). Moreover, cirsimaritin successfully reversed the detrimental effects induced by DSS, including weight reduction, colon truncation, tissue-related damage, increased levels of inflammatory cells in the affected region, and secretion of proinflammatory cytokines. Our findings suggest that cirsimaritin can effectively alleviate acute colitis triggered by DSS.
Collapse
Affiliation(s)
- Abdelrahim Alqudah
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa 13133, Jordan
| | - Esam Qnais
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa 13133, Jordan
| | - Omar Gammoh
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, Irbid 21163, Jordan
| | - Yousra Bseiso
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa 13133, Jordan
| | - Mohammed Wedyan
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa 13133, Jordan
| | - Mohammad Alqudah
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
- Physiology Department, School of Medicine and Biomedical Sciences, Arabian Gulf University, Manama 26671, Bahrain
| | - Taher Hatahet
- School of Pharmacy, Queen’s University Belfast, Belfast BT7 1NN, UK
| |
Collapse
|
17
|
Koh YC, Chang YC, Lin WS, Leung SY, Chen WJ, Wu SH, Wei YS, Gung CL, Chou YC, Pan MH. Efficacy and Mechanism of the Action of Live and Heat-Killed Bacillus coagulans BC198 as Potential Probiotic in Ameliorating Dextran Sulfate Sodium-Induced Colitis in Mice. ACS OMEGA 2024; 9:10253-10266. [PMID: 38463297 PMCID: PMC10918820 DOI: 10.1021/acsomega.3c07529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/01/2024] [Accepted: 02/12/2024] [Indexed: 03/12/2024]
Abstract
Inflammatory bowel disease alters the gut microbiota, causes defects in mucosal barrier function, and leads to dysregulation of the immune response to microbial stimulation. This study investigated and compared the efficacy of a candidate probiotic strain, Bacillus coagulans BC198, and its heat-killed form in treating dextran sulfate sodium-induced colitis. Both live and heat-killed B. coagulans BC198 increased gut barrier-associated protein expression, reduced neutrophil and M1 macrophage infiltration of colon tissue, and corrected gut microbial dysbiosis induced by colitis. However, only live B. coagulans BC198 could alleviate the general symptoms of colitis, prevent colon shortening, and suppress inflammation and tissue damage. At the molecular level, live B. coagulans BC198 was able to inhibit Th17 cells while promoting Treg cells in mice with colitis, reduce pro-inflammatory MCP-1 production, and increase anti-inflammatory IL-10 expression in the colonic mucosa. The live form of B. coagulans BC198 functioned more effectively than the heat-killed form in ameliorating colitis by enhancing the anti-inflammatory response and promoting Treg cell accumulation in the colon.
Collapse
Affiliation(s)
- Yen-Chun Koh
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
| | - Ya-Chu Chang
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
| | - Wei-Sheng Lin
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
- Department
of Food Science, National Quemoy University, Quemoy 892, Taiwan
| | - Siu-Yi Leung
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
| | - Wei-Jen Chen
- Biotech
Department, Syngen Biotech Co., Ltd., Tainan 744094, Taiwan
| | - Shiuan-Huei Wu
- Biotech
Department, Syngen Biotech Co., Ltd., Tainan 744094, Taiwan
| | - Yu-Shan Wei
- Research
and Development Department, Syngen Biotech
Co., Ltd., Tainan 744094, Taiwan
| | - Chiau-Ling Gung
- Research
and Development Department, Syngen Biotech
Co., Ltd., Tainan 744094, Taiwan
| | - Ya-Chun Chou
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
| | - Min-Hsiung Pan
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
- Department
of Medical Research, China Medical University Hospital, China Medical University, Taichung City 40402, Taiwan
- Department
of Health and Nutrition Biotechnology, Asia
University, Taichung City 41354, Taiwan
| |
Collapse
|
18
|
Yao Y, Liu Y, Xu Q, Mao L. Short Chain Fatty Acids: Essential Weapons of Traditional Medicine in Treating Inflammatory Bowel Disease. Molecules 2024; 29:379. [PMID: 38257292 PMCID: PMC10818876 DOI: 10.3390/molecules29020379] [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: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic and recurrent intestinal inflammatory disease, mainly including Crohn's disease (CD) and ulcerative colitis (UC). In recent years, the incidence and prevalence of IBD have been on the rise worldwide and have become a significant concern of health and a huge economic burden on patients. The occurrence and development of IBD involve a variety of pathogenic factors. The changes in short-chain fatty acids (SCFAs) are considered to be an important pathogenic mechanism of this disease. SCFAs are important metabolites in the intestinal microbial environment, which are closely involved in regulating immune, anti-tumor, and anti-inflammatory activities. Changes in metabolite levels can reflect the homeostasis of the intestinal microflora. Recent studies have shown that SCFAs provide energy for host cells and intestinal microflora, shape the intestinal environment, and regulate the immune system, thereby regulating intestinal physiology. SCFAs can effectively reduce the incidence of enteritis, cardiovascular disease, colon cancer, obesity, and diabetes, and also play an important role in maintaining the balance of energy metabolism (mainly glucose metabolism) and improving insulin tolerance. In recent years, many studies have shown that numerous decoctions and natural compounds of traditional Chinese medicine have shown promising therapeutic activities in multiple animal models of colitis and thus attracted increasing attention from scientists in the study of IBD treatment. Some of these traditional Chinese medicines or compounds can effectively alleviate colonic inflammation and clinical symptoms by regulating the generation of SCFAs. This study reviews the effects of various traditional Chinese medicines or bioactive substances on the production of SCFAs and their potential impacts on the severity of colonic inflammation. On this basis, we discussed the mechanism of SCFAs in regulating IBD-associated inflammation, as well as the related regulatory factors and signaling pathways. In addition, we provide our understanding of the limitations of current research and the prospects for future studies on the development of new IBD therapies by targeting SCFAs. This review may widen our understanding of the effect of traditional medicine from the view of SCFAs and their role in alleviating IBD animal models, thus contributing to the studies of IBD researchers.
Collapse
Affiliation(s)
- Yuan Yao
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China; (Y.Y.); (Y.L.)
| | - Yongchao Liu
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China; (Y.Y.); (Y.L.)
| | - Qiuyun Xu
- Basic Medical Research Center, School of Medicine, Nantong University, Nantong 226019, China
| | - Liming Mao
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China; (Y.Y.); (Y.L.)
| |
Collapse
|
19
|
Fasano A, Matera M. Probiotics to Prevent Celiac Disease and Inflammatory Bowel Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1449:95-111. [PMID: 39060733 DOI: 10.1007/978-3-031-58572-2_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
The incidence of chronic inflammatory diseases (CIDs) is dramatically increasing in the developed world, resulting in an increased burden of disease in childhood. Currently, there are limited effective strategies for treating or preventing these conditions. To date, myriads of cross-sectional studies have described alterations in the composition of the gut microbiota in a variety of disease states, after the disease has already occurred. We suggest that to mechanically link these microbiome changes with disease pathogenesis, a prospective cohort design is needed to capture changes that precede or coincide with disease onset and symptoms. In addition, these prospective studies must integrate microbiological, metagenomic, meta transcriptomic and metabolomic data with minimal and standardized clinical and environmental metadata that allow to correctly compare and interpret the results of the analysis of the human microbiota in order to build a system-level model of the interactions between the host and the development of the disease. The creation of new biological computational models thus constructed will allow us to finally move from the detection of simple elements of "association" to the identification of elements of real "causality" allowing to provide a mechanistic approach to the exploration of the development of CIDs.This can only be done when these diseases are studied as complex biological networks. In this chapter we discuss the current knowledge regarding the contribution of the microbiome to CID in childhood, focusing on celiac disease and inflammatory bowel disease, with the overall aim of identifying pathways to shift research from descriptive to mechanistic approaches. We then examine how some components of the microbiota, through epigenetic reprogramming, can start the march from genetic predisposition to clinical expression of CIDs, thus opening up new possibilities for intervention, through microbiota therapy targeting the manipulation of the composition and function of the microbiota, for future applications of precision medicine and primary prevention.
Collapse
Affiliation(s)
- Alessio Fasano
- Research Centre for Immunology and Mucosal Biology and Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital for Children - Harvard Medical School, Boston, USA, MA.
- Mucosal Immunology and Biology Research Center and Division of Pediatric Gastroenterology and Nutrition, Mass General for Children - Harvard Medical School, Boston, MA, USA.
| | - Mariarosaria Matera
- Neonatologist, Neurodevelopmental Clinics and Pediatric Clinical Microbiomic - Misericordia Hospital, Grosseto, Italy
| |
Collapse
|
20
|
Chen X, Zhang M, Zhou F, Gu Z, Li Y, Yu T, Peng C, Zhou L, Li X, Zhu D, Zhang X, Yu C. SIRT3 Activator Honokiol Inhibits Th17 Cell Differentiation and Alleviates Colitis. Inflamm Bowel Dis 2023; 29:1929-1940. [PMID: 37335900 PMCID: PMC10697418 DOI: 10.1093/ibd/izad099] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Indexed: 06/21/2023]
Abstract
BACKGROUND Honokiol (HKL), a natural extract of the bark of the magnolia tree and an activator of the mitochondrial protein sirtuin-3 (SIRT3), has been proposed to possess anti-inflammatory effects. This study investigated the inhibitory effects of HKL on T helper (Th) 17 cell differentiation in colitis. METHODS Serum and biopsies from 20 participants with ulcerative colitis (UC) and 18 healthy volunteers were collected for the test of serum cytokines, flow cytometry analysis (FACS), and relative messenger RNA (mRNA) levels of T cell subsets, as well as the expression of SIRT3 and phosphorylated signal transducer and activator of transcription/retinoic acid-related orphan nuclear receptor γt (p-STAT3/RORγt) signal pathway in colon tissues. In vitro, naïve clusters of differentiation (CD) 4 + T cells isolated from the mouse spleen differentiated to subsets including Th1, Th2, Th17, and regulatory T (Treg) cells. Peripheral blood monocytes (PBMCs) from healthy volunteers were induced to the polarization of Th17 cells. After HKL treatment, changes in T cell subsets, related cytokines, and transcription factors were measured. The dextran sulfate sodium (DSS)-induced colitis and interleukin (IL)-10-deficient mice were intraperitoneally injected with HKL. These experiments were conducted to study the effect of HKL on the development, cytokines, and expression of signaling pathway proteins in colitis. RESULTS Patients with UC had higher serum IL-17 and a higher proportion of Th17 differentiation in blood compared with healthy participants; while IL-10 level and the proportion of Treg cells were lower. Higher relative mRNA levels of RORγt and a lower SIRT3 expression in colon tissues were observed. In vitro, HKL had little effect on the differentiation of naïve CD4+ T cells to Th1, Th2, or Treg cells, but it downregulated IL-17 levels and the Th17 cell ratio in CD4+ T cells from the mouse spleen and human PBMCs under Th17 polarization. Even with a STAT3 activator, HKL still significantly inhibited IL-17 levels. In DSS-induced colitis mice and IL-10 deficient mice treated with HKL, the length of the colon, weight loss, disease activity index, and histopathological scores were improved, IL-17 and IL-21 levels, and the proportion of Th17 cells were decreased. Sirtuin-3 expression was increased, whereas STAT3 phosphorylation and RORγt expression were inhibited in the colon tissue of mice after HKL treatment. CONCLUSIONS Our study demonstrated that HKL could partially protect against colitis by regulating Th17 differentiation through activating SIRT3, leading to inhibition of the STAT3/RORγt signaling pathway. These results provide new insights into the protective effects of HKL against colitis and may facilitate the research of new drugs for inflammatory bowel disease.
Collapse
Affiliation(s)
- Xiaotian Chen
- Department of Clinical Nutrition, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210008, P.R. China
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210008, P.R. China
| | - Mingming Zhang
- Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai 200001, P.R. China
| | - Fan Zhou
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, P.R. China
| | - Zhengrong Gu
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210017, P.R. China
| | - Yuan Li
- Department of Clinical Nutrition, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210008, P.R. China
| | - Ting Yu
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Chunyan Peng
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, P.R. China
| | - Lixing Zhou
- The Center of Gerontology and Geriatrics/National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, P.R. China
| | - Xiangrui Li
- Department of Clinical Nutrition, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210008, P.R. China
| | - Dandan Zhu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, P.R. China
| | - Xiaoqi Zhang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, P.R. China
| | - Chenggong Yu
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210008, P.R. China
| |
Collapse
|
21
|
Fan Q, Dai W, Li M, Wang T, Li X, Deng Z, Li W, Li M. Inhibition of α2,6-sialyltransferase relieves symptoms of ulcerative colitis by regulating Th17 cells polarization. Int Immunopharmacol 2023; 125:111130. [PMID: 37897948 DOI: 10.1016/j.intimp.2023.111130] [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: 08/03/2023] [Revised: 10/08/2023] [Accepted: 10/22/2023] [Indexed: 10/30/2023]
Abstract
Ulcerative colitis (UC) is a chronic, relapsing inflammatory disease that affects human intestines. Immune imbalance is one of the important factors inducing UC. After the activation of CD4+ T cells, pro-inflammatory cytokines are produced to induce colonic inflammation. α2,6-Sialylation, catalyzed by α2,6-sialyltransferase (ST6GAL1), affects the proliferation, activation, and T cell receptor (TCR) signaling of CD4+ T cells, but its role in CD4+ T cell polarization, regulation of Th17 / Treg balance, and its role in UC are still unclear. We found the number of CD4+ T and Th17 cells increased in colonic tissue with UC. The level of α2,6-sialylation of CD4+ T cells in patients with UC was significantly increased. De-α2,6-sialylation significantly reduced the symptoms of UC in rats. ST6GAL1 gene knockout inhibited the polarization of CD4+ T cells to Th17 cells, and promoted the polarization of CD4+ T cells to Treg cells. ST6GAL1 knockout significantly inhibited the IL-17 signaling pathway in CD4+ T cells and inhibited the secretion of pro-inflammatory cytokine IL-17a. ST6GAL1 and IL-17a are highly expressed in patients with UC, and there is a positive correlation between them. In conclusion, reduced α2,6-sialylation inhibits the polarization of CD4+ T cells to Th17 cells, inhibits IL-17a signaling pathway and reduces the level of pro-inflammatory cytokine IL-17a to alleviate the symptoms of UC, which is a potential novel target for the clinical treatment of UC.
Collapse
Affiliation(s)
- Qingjie Fan
- College of Basic Medical Science, Dalian Medical University, 9-Western Section, Lvshun South Road, Dalian, Liaoning 116044, China
| | - Wenjie Dai
- Department of Gastroenterology, Xinhua Hospital Affiliated to Dalian University, No. 156, Wansui Street, Dalian, Liaoning 116044, China
| | - Mechou Li
- College of Basic Medical Science, Dalian Medical University, 9-Western Section, Lvshun South Road, Dalian, Liaoning 116044, China
| | - Tiantong Wang
- College of Basic Medical Science, Dalian Medical University, 9-Western Section, Lvshun South Road, Dalian, Liaoning 116044, China
| | - Xinran Li
- College of Basic Medical Science, Dalian Medical University, 9-Western Section, Lvshun South Road, Dalian, Liaoning 116044, China
| | - Zhaobin Deng
- Department of Gastroenterology, Xinhua Hospital Affiliated to Dalian University, No. 156, Wansui Street, Dalian, Liaoning 116044, China.
| | - Wenzhe Li
- Department of Thoracic Surgery, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China.
| | - Ming Li
- College of Basic Medical Science, Dalian Medical University, 9-Western Section, Lvshun South Road, Dalian, Liaoning 116044, China.
| |
Collapse
|
22
|
Choi SI, Shin YC, Lee JS, Yoon YC, Kim JM, Sung MK. N-Acetylglucosamine and its dimer ameliorate inflammation in murine colitis by strengthening the gut barrier function. Food Funct 2023; 14:8533-8544. [PMID: 37655824 DOI: 10.1039/d3fo00282a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Ulcerative colitis (UC) is a chronic gastrointestinal disease whose incidence is increasing rapidly worldwide. Anti-inflammatory medications, including 5-aminosalicylic acid (5-ASA), corticosteroids, and immunosuppressants, are used for its treatment; however, new alternatives would be required due to the serious side effects of some of these medications. N-Acetylglucosamine (NAG) is an amino sugar composed of mucin that is secreted by intestinal epithelial cells. It is also used to promote the growth of intestinal bacteria. The current study aimed to determine the efficacy of NAG against dextran sulfate sodium (DSS)-induced chronic colitis and elucidate its mechanism of action. Mice were randomly divided into control, DSS, 0.1% sulfasalazine, 0.1% NAG, 0.3% NAG, and 0.3% NAG-dimer (NAG-D) groups, and results showed that colitis-induced body weight loss, disease activity, colonic tissue damage, colon length shortening, and the loss of mucin-secreting area were significantly improved in the NAG-D group. The intestinal permeability indicator, serum CD 14 level, and expression of the tight junction protein, occludin, were both improved in the 0.3% NAG group. Inflammatory biomarkers, including GATA3, IFN-γ, p-IκBα, COX2, TGF-β1, and Smad7, were significantly lower in the 0.3% NAG and NAG-D groups than in the DSS group. The intestinal microbial composition was most significantly altered in the 0.3% NAG group, showing decreased ratios of pathogenic bacteria, such as Betaproteobacteria, especially Burkholderiales. The results overall suggested that NAG or NAG-D supplementation can alleviate inflammation by strengthening the intestinal barrier function and maintaining gut microbiota homeostasis in a DSS-induced colitis mouse model.
Collapse
Affiliation(s)
- Sung-In Choi
- Department of Food and Nutrition, College of Human Ecology, Sookmyung Women's University, Chungpa-ro 47-gil 100, Yongsan-gu, Seoul 04310, Republic of Korea.
| | | | - Joong Su Lee
- Amicogen Inc., Jinju-si 52621, Republic of Korea
| | - Yeo Cho Yoon
- Amicogen Inc., Jinju-si 52621, Republic of Korea
| | - Ju Myung Kim
- Amicogen Inc., Jinju-si 52621, Republic of Korea
| | - Mi-Kyung Sung
- Department of Food and Nutrition, College of Human Ecology, Sookmyung Women's University, Chungpa-ro 47-gil 100, Yongsan-gu, Seoul 04310, Republic of Korea.
| |
Collapse
|
23
|
Hidalgo-García L, Ruiz-Malagon AJ, Huertas F, Rodríguez-Sojo MJ, Molina-Tijeras JA, Diez-Echave P, Becerra P, Mirón B, Morón R, Rodríguez-Nogales A, Gálvez J, Rodríguez-Cabezas ME, Anderson P. Administration of intestinal mesenchymal stromal cells reduces colitis-associated cancer in C57BL/6J mice modulating the immune response and gut dysbiosis. Pharmacol Res 2023; 195:106891. [PMID: 37586618 DOI: 10.1016/j.phrs.2023.106891] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/22/2023] [Accepted: 08/12/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND Patients with inflammatory bowel disease (IBD) have a higher risk of developing colitis-associated colorectal cancer (CAC) with poor prognosis. IBD etiology remains undefined but involves environmental factors, genetic predisposition, microbiota imbalance (dysbiosis) and mucosal immune defects. Mesenchymal stromal cell (MSC) injections have shown good efficacy in reducing intestinal inflammation in animal and human studies. However, their effect on tumor growth in CAC and their capacity to restore gut dysbiosis are not clear. METHODS The outcome of systemic administrations of in vitro expanded human intestinal MSCs (iMSCs) on tumor growth in vivo was evaluated using the AOM/DSS model of CAC in C57BL/6J mice. Innate and adaptive immune responses in blood, mesenteric lymph nodes (MLNs) and colonic tissue were analyzed by flow cytometry. Intestinal microbiota composition was evaluated by 16S rRNA amplicon sequencing. RESULTS iMSCs significantly inhibited colitis and intestinal tumor development, reducing IL-6 and COX-2 expression, and IL-6/STAT3 and PI3K/Akt signaling. iMSCs decreased colonic immune cell infiltration, and partly restored intestinal monocyte homing and differentiation. iMSC administration increased the numbers of Tregs and IFN-γ+CD8+ T cells in the MLNs while decreasing the IL-4+Th2 response. It also ameliorated intestinal dysbiosis in CAC mice, increasing diversity and Bacillota/Bacteroidota ratio, as well as Akkermansia abundance, while reducing Alistipes and Turicibacter, genera associated with inflammation. CONCLUSION Administration of iMSCs protects against CAC, ameliorating colitis and partially reverting intestinal dysbiosis, supporting the use of MSCs for the treatment of IBD.
Collapse
Affiliation(s)
- Laura Hidalgo-García
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Antonio Jesús Ruiz-Malagon
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Francisco Huertas
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain; Servicio de Cirugía, Hospital Universitario Virgen de las Nieves, 18012 Granada, Spain
| | - María Jesús Rodríguez-Sojo
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - José Alberto Molina-Tijeras
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Patricia Diez-Echave
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Patricia Becerra
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain; Servicio de Anatomía Patológica, Hospital Universitario Clínico San Cecilio, 18014 Granada, Spain
| | - Benito Mirón
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain; Servicio de Cirugía, Hospital Universitario Clínico San Cecilio, 18016 Granada, Spain
| | - Rocío Morón
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain; Servicio Farmacia Hospitalaria, Hospital Universitario Clínico San Cecilio, 18016 Granada, Spain
| | - Alba Rodríguez-Nogales
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain.
| | - Julio Gálvez
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain; Centro de Investigación Biomédica En Red para Enfermedades Hepáticas y Digestivas (CIBER-EHD), School of Pharmacy, University of Granada, 18071 Granada, Spain.
| | - María Elena Rodríguez-Cabezas
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Per Anderson
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain; Servicio de Análisis Clínicos e Inmunología, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain; Departamento de Bioquímica, Biología Molecular e Inmunología III, University of Granada, 18016 Granada, Spain
| |
Collapse
|
24
|
Linggi B, Cremer J, Wang Z, Van Viegen T, Vermeire S, Lefevre P, Shackelton LM, Jairath V, Teft W, Vande Casteele N, Verstockt B. Effect of storage time on peripheral blood mononuclear cell isolation from blood collected in vacutainer CPT™ tubes. J Immunol Methods 2023; 519:113504. [PMID: 37257687 DOI: 10.1016/j.jim.2023.113504] [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: 10/20/2022] [Revised: 05/12/2023] [Accepted: 05/25/2023] [Indexed: 06/02/2023]
Abstract
BACKGROUND Clinical trials of novel therapies for the treatment of ulcerative colitis (UC) may benefit from immune cell profiling, however implementation of this methodology is limited in the multicenter trial setting by necessity of timely (within 6 to 8 h) isolation and processing of peripheral blood mononuclear cells (PBMC) from whole blood samples. Becton Dickinson Vacutainer CPT™ Cell Preparation Tubes (CPT™) limit required processing prior to shipping to a central lab to an initial centrifugation step within 24 h of sample collection. As shipping may delay final processing beyond 24 h, we analyzed cell viability and T cell composition in whole blood stored in CPT™ to determine if their use may accommodate processing delays typical for multicenter clinical trials. METHODS Whole blood samples from 3 patients with UC were collected in CPT™ (15 tubes/patient) and PBMC were processed at various timepoints (24-96 h). Cell viability and T cell composition (26 types) were evaluated by flow cytometry. Variability between technical and biological replicates was evaluated in the context of cell-type abundance, delayed processing time, and data normalization. RESULTS Total cell viability was <50% when processing was delayed to 48 h after collection and was further reduced at later processing timepoints. The effect of delayed processing on cell abundance varied widely across cell types, with CD4+, CD8+, naïve effector CD8+, and Tcm CD4 + T cells displaying the least variability in abundance with delayed processing. Normalization of cell counts to cell types other than total T cells corrected for the effect of delayed processing for several cell types, particularly Th17. CONCLUSIONS Based on these data, processing of PBMC in CPT™ should ideally be performed within 48 h. Delayed processing of PBMC in CPT™ may be considered for cell types that are robust to these conditions. Normalization of cell abundance to different parental cell-types may reduce variability in quantitation and should be used in conjunction with the expected effect size to meet the experimental goals of a multicenter clinical trial.
Collapse
Affiliation(s)
- Bryan Linggi
- Alimentiv Inc., 100 Dundas Street, Suite 200, London, ON, Canada.
| | - Jonathan Cremer
- Department of Microbiology and Immunology, Laboratory of Allergy and Clinical Immunology, KU Leuven, Herestraat 49, Leuven, Belgium; Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases & Metabolism (CHROMETA), KU Leuven, Herestraat 49, Leuven, Belgium.
| | - Zhongya Wang
- Alimentiv Inc., 100 Dundas Street, Suite 200, London, ON, Canada.
| | - Tanja Van Viegen
- Alimentiv Inc., 100 Dundas Street, Suite 200, London, ON, Canada.
| | - Séverine Vermeire
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases & Metabolism (CHROMETA), KU Leuven, Herestraat 49, Leuven, Belgium; Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Herestraat 49, Leuven, Belgium.
| | - Pavine Lefevre
- Alimentiv Inc., 100 Dundas Street, Suite 200, London, ON, Canada.
| | | | - Vipul Jairath
- Alimentiv Inc., 100 Dundas Street, Suite 200, London, ON, Canada; Departments of Medicine and Epidemiology and Biostatistics, Western University, 1151 Richmond St, London, ON, Canada.
| | - Wendy Teft
- Alimentiv Inc., 100 Dundas Street, Suite 200, London, ON, Canada.
| | - Niels Vande Casteele
- Alimentiv Inc., 100 Dundas Street, Suite 200, London, ON, Canada; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, USA.
| | - Bram Verstockt
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases & Metabolism (CHROMETA), KU Leuven, Herestraat 49, Leuven, Belgium; Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Herestraat 49, Leuven, Belgium.
| |
Collapse
|
25
|
Niu Y, Zhang J, Shi D, Zang W, Niu J. Glycosides as Potential Medicinal Components for Ulcerative Colitis: A Review. Molecules 2023; 28:5210. [PMID: 37446872 DOI: 10.3390/molecules28135210] [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: 05/29/2023] [Revised: 06/21/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Ulcerative colitis (UC) is a chronic, non-specific disease of unknown etiology. The disease develops mainly in the rectum or colon, and the main clinical symptoms include abdominal pain, diarrhea, and purulent bloody stools, with a wide variation in severity. The specific causative factors and pathogenesis of the disease are not yet clear, but most scholars believe that the disease is caused by the interaction of genetic, environmental, infectious, immune, and intestinal flora factors. As for the treatment of UC, medications are commonly used in clinical practice, mainly including aminosalicylates, glucocorticoids, and immunosuppressive drugs. However, due to the many complications associated with conventional drug therapy and the tendency for UC to recur, there is an urgent need to discover new, safer, and more effective drugs. Natural compounds with biodiversity and chemical structure diversity from medicinal plants are the most reliable source for the development of new drug precursors. Evidence suggests that glycosides may reduce the development and progression of UC by modulating anti-inflammatory responses, inhibiting oxidative stress, suppressing abnormal immune responses, and regulating signal transduction. In this manuscript, we provide a review of the epidemiology of UC and the available drugs for disease prevention and treatment. In addition, we demonstrate the protective or therapeutic role of glycosides in UC and describe the possible mechanisms of action to provide a theoretical basis for preclinical studies in drug development.
Collapse
Affiliation(s)
- Yating Niu
- School of Basic Medical Science, Ningxia Medical University, Yinchuan 750004, China
| | - Jun Zhang
- Shandong Academy of Chinese Medicine, Jinan 250014, China
| | - Dianhua Shi
- Shandong Academy of Chinese Medicine, Jinan 250014, China
| | - Weibiao Zang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Jianguo Niu
- School of Basic Medical Science, Ningxia Medical University, Yinchuan 750004, China
| |
Collapse
|
26
|
Cheru N, Hafler DA, Sumida TS. Regulatory T cells in peripheral tissue tolerance and diseases. Front Immunol 2023; 14:1154575. [PMID: 37197653 PMCID: PMC10183596 DOI: 10.3389/fimmu.2023.1154575] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/13/2023] [Indexed: 05/19/2023] Open
Abstract
Maintenance of peripheral tolerance by CD4+Foxp3+ regulatory T cells (Tregs) is essential for regulating autoreactive T cells. The loss of function of Foxp3 leads to autoimmune disease in both animals and humans. An example is the rare, X-linked recessive disorder known as IPEX (Immune Dysregulation, Polyendocrinopathy, Enteropathy X-linked) syndrome. In more common human autoimmune diseases, defects in Treg function are accompanied with aberrant effector cytokines such as IFNγ. It has recently become appreciated that Tregs plays an important role in not only maintaining immune homeostasis but also in establishing the tissue microenvironment and homeostasis of non-lymphoid tissues. Tissue resident Tregs show profiles that are unique to their local environments which are composed of both immune and non-immune cells. Core tissue-residence gene signatures are shared across different tissue Tregs and are crucial to homeostatic regulation and maintaining the tissue Treg pool in a steady state. Through interaction with immunocytes and non-immunocytes, tissue Tregs exert a suppressive function via conventional ways involving contact dependent and independent processes. In addition, tissue resident Tregs communicate with other tissue resident cells which allows Tregs to adopt to their local microenvironment. These bidirectional interactions are dependent on the specific tissue environment. Here, we summarize the recent advancements of tissue Treg studies in both human and mice, and discuss the molecular mechanisms that maintain tissue homeostasis and prevent pathogenesis.
Collapse
Affiliation(s)
- Nardos Cheru
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
| | - David A. Hafler
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Tomokazu S. Sumida
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
27
|
Geng B, Ding X, Li X, Liu H, Zhao W, Gong H, Tian Z, Guo J. Peripheral blood T-lymphocyte subsets are potential biomarkers of disease severity and clinical outcomes in patients with ulcerative colitis: a retrospective study. BMC Gastroenterol 2023; 23:136. [PMID: 37106335 PMCID: PMC10134527 DOI: 10.1186/s12876-023-02769-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is considered an immune-mediated disease. The disorder of T-lymphocyte subsets plays an important role in the pathogenesis of UC. The aim of this study was to evaluate the significance of peripheral blood T-lymphocyte subsets in assessing disease severity and predicting clinical outcomes in UC patients. METHODS The retrospective case-control study was performed in 116 UC patients with active disease and 90 healthy controls (HC). The UC patients included were followed up for 180 days. Analyses of t-test, Spearman's correlation coefficient, multivariable Cox regression analysis, receiver operating characteristic (ROC) curves and cumulative survival analysis were done. RESULTS The UC patients had lower proportions of CD4+T cells (42.85%±9.77% vs 45.71%±7.94%, P=0.021) and higher proportion of CD8+T cells (27.88%±8.86% vs 25.00%±6.47%, P=0.008) than HC. The severely active UC patients had higher proportion of CD3+HLA-DR+ T cells (8.83%±6.55% vs 2.80%±1.55%, P<0.001; 8.83%±6.55% vs 4.06%±5.01%, P<0.001) and CD8+T cells (31.35%±8.49% vs 26.98%±7.98%, P=0.029; 31.35%±8.49% vs 25.46%±9.15%, P=0.003) than mild and moderate group, whereas lower proportion of CD4+CD25+T cells (2.86%±1.35% vs 3.46%±1.07%, P=0.034) than mild group and CD4+T cells (40.40%±9.36% vs 44.73%±10.39%, P=0.049) than moderate group. The area under the curve (AUC) of CD3+HLA-DR+ T cells for assessing severely active UC was 0.885, with the cut-off value of 5.33%. The sensitivity was 76.32% and specificity was 89.74%. The combination of CD3+HLA-DR+ T cells and CRP had stronger assessment value with AUC of 0.929. The AUC of CD8+T cells, CD4+/CD8+ ratio and CD4+CD25+T cells for assessing disease severity was 0.677, 0.669 and 0.631 respectively. Within the 180 days follow-up, 24 patients (20.69%) had UC-related readmission or surgery, with higher proportion of CD3+HLA-DR+ T cells (10.66%±9.52% vs 3.88%±2.56%, P=0.003) and CD8+T cells (31.19%±10.59% vs 27.01%±8.20%, P=0.039) than those without readmission and surgery. The proportion of CD3+HLA-DR+ T cells was the independent predictor of UC-related readmission or surgery (HR=1.109, P=0.002). The AUC of CD3+HLA-DR+ T cells for predicting readmission or surgery was 0.796 with the cut-off value of 5.38%. UC patients with CD3+HLA-DR+T cells proportion>5.38% had a shorter time to readmission or surgery (log-rank test, P<0.001). CONCLUSIONS The combination of CD3+HLA-DR+T cells and CRP may be potential biomarker of disease severity in UC patients. The high proportion of CD3+HLA-DR+T cells may be associated with an increased risk of readmission or surgery in UC patients.
Collapse
Affiliation(s)
- Bailu Geng
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China
| | - Xueli Ding
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China
| | - Xiaoyu Li
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China
| | - Hua Liu
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China
| | - Wenjun Zhao
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China
| | - Haihong Gong
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zibin Tian
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China.
| | - Jing Guo
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China.
| |
Collapse
|
28
|
Li H, Xing H. Interleukin-35 Enhances Regulatory T Cell Function by Potentially Suppressing Their Transdifferentiation into a T Helper 17-Like Phenotype in Kawasaki Disease. Immunol Invest 2023:1-16. [PMID: 37052682 DOI: 10.1080/08820139.2023.2201283] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Interleukin-35 (IL-35) modulates immune cell activity in inflammation and autoimmune disorders. However, its specific effects on regulatory T cells (Tregs) in Kawasaki disease remain ambiguous. We enrolled 37 patients with Kawasaki disease and 20 healthy controls in this study. The percentages of CD4+CD25+CD127dim/- Tregs and CD4+IL-17A+ T helper 17 (Th17) cells were determined via flow cytometry. Tregs were enriched and stimulated by recombinant IL-35. Immunosuppressive activity of Tregs was via co-culture with autologous CD4+CD25- T cells. Purified Tregs were cultured for Th17 polarization, and the influence of IL-35 on Tregs transdifferentiation into a Th17-like phenotype was determined. The percentage of Tregs was elevated in patients with Kawasaki disease and positively correlated with C-reactive protein levels. There was no significant difference in the percentage of Th17 cells between the two groups. IL-35 stimulation increased the percentage of Tregs in both groups, but decreased the percentage of Tregs Th17 cells in affected patients. IL-35 enhanced the immunosuppressive activity of Tregs in both groups, resulting in decreased cellular proliferation and increased IL-35 subunit mRNA relative levels in co-culture system. IL-35 did not affect the immune checkpoint molecule expression in Tregs, but inhibited the transdifferentiation of Tregs into a Th17-like phenotype in affected patients, indicating by the down-regulations of C-C motif chemokine receptor-4/6 expression, retinoid-related orphan nuclear receptor γt mRNA levels, and IL-17 secretion. IL-35 contributes to the immunosuppressive function of Tregs by inhibiting the cellular proliferation and transdifferentiation of Tregs into a Th17-like phenotype, which may be a protective mechanism against Kawasaki disease.
Collapse
Affiliation(s)
- Hua Li
- Department of Disease Prevention and Control, Xi'an Children's Hospital, The Children's Hospital Affiliated to Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Haijian Xing
- Department of General Internal Medicine, Xi'an Children's Hospital, The Children's Hospital Affiliated to Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
29
|
Włodarczyk J, Krajewska J, Szeleszczuk Ł, Szałwińska P, Gurba A, Lipiec S, Taciak P, Szczepaniak R, Mlynarczuk-Bialy I, Fichna J. A New Gold(III) Complex, TGS 703, Shows Potent Anti-Inflammatory Activity in Colitis via the Enzymatic and Non-Enzymatic Antioxidant System-An In Vitro, In Silico, and In Vivo Study. Int J Mol Sci 2023; 24:ijms24087025. [PMID: 37108188 PMCID: PMC10138903 DOI: 10.3390/ijms24087025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/02/2023] [Accepted: 04/08/2023] [Indexed: 04/29/2023] Open
Abstract
Inflammatory bowel diseases (IBD) and their main representatives, Crohn's disease and ulcerative colitis, are worldwide health-care problems with constantly increasing frequency and still not fully understood pathogenesis. IBD treatment involves drugs such as corticosteroids, derivatives of 5-aminosalicylic acid, thiopurines, and others, with the goal to achieve and maintain remission of the disease. Nowadays, as our knowledge about IBD is continually growing, more specific and effective therapies at the molecular level are wanted. In our study, we tested novel gold complexes and their potential effect on inflammation and IBD in vitro, in silico, and in vivo. A series of new gold(III) complexes (TGS 404, 512, 701, 702, and 703) were designed and screened in the in vitro inflammation studies. In silico modeling was used to study the gold complexes' structure vs. their activity and stability. Dextran sulphate sodium (DSS)-induced mouse model of colitis was employed to characterize the anti-inflammatory activity in vivo. Lipopolysaccharide (LPS)-stimulated RAW264.7 cell experiments proved the anti-inflammatory potential of all tested complexes. Selected on the bases of in vitro and in silico analyses, TGS 703 significantly alleviated inflammation in the DSS-induced mouse model of colitis, which was confirmed by a statistically significant decrease in the macro- and microscopic score of inflammation. The mechanism of action of TGS 703 was linked to the enzymatic and non-enzymatic antioxidant systems. TGS 703 and other gold(III) complexes present anti-inflammatory potential and may be applied therapeutically in the treatment of IBD.
Collapse
Affiliation(s)
- Jakub Włodarczyk
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland
- Department of General and Oncological Surgery, Medical University of Lodz, Pomorska 251, 92-213 Lodz, Poland
| | - Julia Krajewska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland
| | - Łukasz Szeleszczuk
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-093 Warsaw, Poland
| | - Patrycja Szałwińska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland
| | - Agata Gurba
- Department of Pharmacodynamics, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-093 Warsaw, Poland
| | - Szymon Lipiec
- Department for Histology and Embryology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland
| | - Przemysław Taciak
- Department of Pharmacodynamics, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-093 Warsaw, Poland
| | | | - Izabela Mlynarczuk-Bialy
- Department for Histology and Embryology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland
| |
Collapse
|
30
|
Tian CM, Yang MF, Xu HM, Zhu MZ, Zhang Y, Yao J, Wang LS, Liang YJ, Li DF. Mesenchymal Stem Cell-derived Exosomes: Novel Therapeutic Approach for Inflammatory Bowel Diseases. Stem Cells Int 2023; 2023:4245704. [PMID: 37056457 PMCID: PMC10089786 DOI: 10.1155/2023/4245704] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 02/19/2023] [Accepted: 03/22/2023] [Indexed: 04/07/2023] Open
Abstract
As double membrane-encapsulated nanovesicles (30-150 nm), exosomes (Exos) shuttle between different cells to mediate intercellular communication and transport active cargoes of paracrine factors. The anti-inflammatory and immunomodulatory activities of mesenchymal stem cell (MSC)-derived Exos (MSC-Exos) provide a rationale for novel cell-free therapies for inflammatory bowel disease (IBD). Growing evidence has shown that MSC-Exos can be a potential candidate for treating IBD. In the present review, we summarized the most critical advances in the properties of MSC-Exos, provided the research progress of MSC-Exos in treating IBD, and discussed the molecular mechanisms underlying these effects. Collectively, MSC-Exos had great potential for cell-free therapy in IBD. However, further studies are required to understand the full dimensions of the complex Exo system and how to optimize its effects.
Collapse
Affiliation(s)
- Cheng-mei Tian
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- Department of Emergency, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong, China
| | - Mei-feng Yang
- Department of Hematology, Yantian District People's Hospital, Shenzhen, Guangdong, China
| | - Hao-ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Min-zheng Zhu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou, Guangdong, China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Li-sheng Wang
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Yu-jie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen, Guangdong, China
| | - De-feng Li
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| |
Collapse
|
31
|
HIURA K, MARUYAMA T, WATANABE M, NAKANO K, OKAMURA T, SASAKI H, SASAKI N. Mitotic spindle positioning protein (MISP) deficiency exacerbates dextran sulfate sodium (DSS)-induced colitis in mice. J Vet Med Sci 2023; 85:167-174. [PMID: 36596561 PMCID: PMC10017287 DOI: 10.1292/jvms.22-0483] [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: 10/25/2022] [Accepted: 12/07/2022] [Indexed: 12/31/2022] Open
Abstract
Inflammatory bowel disease (IBD) is classified into two types: Crohn's disease and ulcerative colitis. In IBD, the imbalance between the pro-inflammatory and anti-inflammatory cytokines prevents recovery from the inflammatory state, resulting in chronic inflammation in the colon. The mitotic spindle positioning protein (MISP) is localized to the apical membrane in the colon. In this study, we observed increased expression of MISP in the intestinal epithelial cells in dextran sulfate sodium (DSS)-induced colitis in mice. MISP-deficient mice receiving DSS showed significant exacerbation of colitis (e.g., weight loss, loss of the crypts). The intestinal epithelial cells of the MISP-deficient mice showed a trend towards decreased cell proliferation after DSS treatment. Reverse transcription followed by quantitative polymerase chain reaction revealed that the expression levels of Tgfb1, an anti-inflammatory cytokine, were significantly reduced in the colon of MISP-deficient mice compared with the wild-type mice regardless of DSS treatment. These findings indicate that MISP may play a role in the recovery of the colon after inflammation through its anti-inflammatory and proliferative activities, suggesting that MISP may be a new therapeutic target for IBD.
Collapse
Affiliation(s)
- Koki HIURA
- Laboratory of Laboratory Animal Science and Medicine, School
of Veterinary Medicine, Kitasato University, Aomori, Japan
| | - Takumi MARUYAMA
- Laboratory of Laboratory Animal Science and Medicine, School
of Veterinary Medicine, Kitasato University, Aomori, Japan
- Department of Laboratory Animal Medicine, Research
Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Masaki WATANABE
- Laboratory of Laboratory Animal Science and Medicine, School
of Veterinary Medicine, Kitasato University, Aomori, Japan
| | - Kenta NAKANO
- Department of Laboratory Animal Medicine, Research
Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tadashi OKAMURA
- Department of Laboratory Animal Medicine, Research
Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hayato SASAKI
- Laboratory of Laboratory Animal Science and Medicine, School
of Veterinary Medicine, Kitasato University, Aomori, Japan
| | - Nobuya SASAKI
- Laboratory of Laboratory Animal Science and Medicine, School
of Veterinary Medicine, Kitasato University, Aomori, Japan
| |
Collapse
|
32
|
Fabian O, Klocperk A, Lerchova T, Jencova P, Stolova L, Belhajova M, Voriskova D, Kazeka D, Vicha A, Hradsky O, Bronsky J. Serum and Mucosal CD30 in Pediatric Inflammatory Bowel Diseases: Useful Biomarker for Diagnosis and Disease Activity Monitoring? Dig Dis Sci 2023; 68:460-470. [PMID: 36056999 DOI: 10.1007/s10620-022-07677-4] [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: 04/12/2022] [Accepted: 08/16/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND Inflammatory bowel diseases (IBD) frequently manifest in pediatric age, but may have atypical clinical, histological and laboratory features. Their underlying immune pathophysiology is incompletely understood, rendering quick diagnosis followed by tailored therapy difficult. The tumor necrosis factor superfamily receptor CD30 has been proposed as a potential marker of ulcerative colitis (UC) and has also been associated with elevated Th2 helper T cells. METHODS A cohort of pediatric patients with UC and Crohn's disease (CD) was evaluated for serum soluble CD30 (sCD30) using ELISA and expression of CD30 and subpopulations of Th1/Th2/Th17 lymphocytes in the gastrointestinal mucosa using flow cytometry (FCM). The dataset is supported by endoscopic and microscopic activity of the disease and basic laboratory markers of inflammation. RESULTS The cohort consisted of 102 observations from 94 patients. sCD30 levels did not differ between patients with CD or UC. However, sCD30 levels correlated with levels of CRP, ESR, fecal calprotectin and albumin and also with clinical activity of the disease in patients with both UC and CD. FCM was not helpful in evaluation of mucosal CD30, which was lowly expressed and not associated with the diagnosis or disease activity. We show augmented Th2 and Th1/17 response in terminal ileum and right-sided colon and decreased Th1/17 response in left-sided colon of UC patients. T lymphocyte subsets were also affected by anti-TNF treatment and patients' age. CONCLUSIONS Neither sCD30 nor mucosal CD30 expression was helpful in differentiating between UC and CD. sCD30 seems to reflect a degree of systemic inflammation and clinical activity in IBD.
Collapse
Affiliation(s)
- Ondrej Fabian
- Clinical and Transplant Pathology Centre, Institute for Clinical and Experimental Medicine, Videnska 1958/9, Prague 4, 140 21, Czech Republic.
- Department of Pathology and Molecular Medicine, 3Rd Faculty of Medicine, Charles University and Thomayer Hospital, Videnska 800, Prague 4, 140 59, Czech Republic.
- Department of Pathology and Molecular Medicine, 2nd Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, Prague 5, 150 06, Czech Republic.
| | - Adam Klocperk
- Department of Immunology, 2Nd Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, Prague 5, 150 06, Czech Republic
| | - Tereza Lerchova
- Gastroenterology and Nutrition Unit, Department of Paediatrics, 2Nd Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, Prague 5, 150 06, Czech Republic
| | - Pavla Jencova
- Department of Paediatric Haematology and Oncology, 2Nd Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, Prague 5, 150 06, Czech Republic
| | - Lucie Stolova
- Department of Paediatric Haematology and Oncology, 2Nd Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, Prague 5, 150 06, Czech Republic
| | - Marie Belhajova
- Department of Paediatric Haematology and Oncology, 2Nd Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, Prague 5, 150 06, Czech Republic
| | - Dagmar Voriskova
- Department of Paediatric Haematology and Oncology, 2Nd Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, Prague 5, 150 06, Czech Republic
| | - Denis Kazeka
- Gastroenterology and Nutrition Unit, Department of Paediatrics, 2Nd Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, Prague 5, 150 06, Czech Republic
| | - Ales Vicha
- Department of Paediatric Haematology and Oncology, 2Nd Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, Prague 5, 150 06, Czech Republic
| | - Ondrej Hradsky
- Gastroenterology and Nutrition Unit, Department of Paediatrics, 2Nd Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, Prague 5, 150 06, Czech Republic
| | - Jiri Bronsky
- Gastroenterology and Nutrition Unit, Department of Paediatrics, 2Nd Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, Prague 5, 150 06, Czech Republic
| |
Collapse
|
33
|
Decreased ratio of FOXP3 +/FOXP3 -CD45RA +CD4 + T cells in peripheral blood is associated with unexplained infertility and ART failure. J Reprod Immunol 2023; 155:103793. [PMID: 36603467 DOI: 10.1016/j.jri.2022.103793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/17/2022] [Accepted: 12/29/2022] [Indexed: 01/01/2023]
Abstract
Unexplained infertility has a huge social impact and is a significant challenge for both clinicians and researchers. Previous studies have shown the involvement of multiple factors in infertility. Among these, the subset of regulatory T cells is of particular interest for the maternal tolerance towards the semi-allogenic fetus. We investigated circulating CD45RA+ regulatory and non-regulatory CD4+ T cells in healthy women and patients with unexplained infertility in the context of thymic output and peripheral proliferation. The proportion of FOXP3+ and FOXP3-CD45RA+CD4+ T cells in peripheral blood was studied in control groups of healthy parous and nulliparous (never-pregnant) women and in patients with unexplained infertility. In the same groups thymic output and peripheral proliferation were defined by the sj/βTREC ratio, and signal joint T-cell receptor excision circles (sjTREC) and Ki67 expression, respectively. In parous women a decrease in sjTREC/105 cells and CD45RA+ T lymphocytes, compared to nulliparous group was found. At the same time, the proportion of FOXP3-CD45RA+CD4+ cells, but not FOXP3+CD45RA+ Tregs was reduced. In contrast, in patients with unsuccessful pregnancy, proportions of both regulatory and non-regulatory T cell counterparts were lower. Taken together, our results provide evidence for group-specific properties in the CD45RA+ T cell compartment between healthy parous, nulliparous and women with unexplained infertility.
Collapse
|
34
|
Lin SJH, Helm ET, Gabler NK, Burrough ER. Acute infection with Brachyspira hyodysenteriae affects mucin expression, glycosylation, and fecal MUC5AC. Front Cell Infect Microbiol 2023; 12:1042815. [PMID: 36683692 PMCID: PMC9852840 DOI: 10.3389/fcimb.2022.1042815] [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: 09/13/2022] [Accepted: 11/28/2022] [Indexed: 01/08/2023] Open
Abstract
Introduction Infection with strongly β-hemolytic strains of Brachyspira hyodysenteriae leads to swine dysentery (SD), a production-limiting disease that causes mucohemorrhagic diarrhea and typhlocolitis in pigs. This pathogen has strong chemotactic activity toward mucin, and infected pigs often have a disorganized mucus layer and marked de novo expression of MUC5AC, which is not constitutively expressed in the colon. It has been shown that fucose is chemoattractant for B. hyodysenteriae, and a highly fermentable fiber diet can mitigate and delay the onset of SD. Methods We used lectins targeting sialic acids in α-2,6 or α-2,3 linkages, N-acetylglucosamine (GlcNAc), α-linked L-fucose, and an immunohistochemical stain targeting N-glycolylneuraminic acid (NeuGc) to investigate the local expression of these mucin glycans in colonic tissues of pigs with acute SD. We used a commercial enzyme-linked immunosorbent assay (ELISA) to quantify fecal MUC5AC in infected pigs and assess its potential as a diagnostic monitoring tool and RNA in situ hybridization to detect IL-17A in the colonic mucosa. Results Colonic mucin glycosylation during SD has an overall increase in fucose, a spatially different distribution of GlcNAc with more expression within the crypt lumens of the upper colonic mucosa, and decreased expression or a decreased trend of sialic acids in α-2,6 or α-2,3 linkages, and NeuGc compared to the controls. The degree of increased fucosylation was less in the colonic mucosa of pigs with SD and fed the highly fermentable fiber diet. There was a significant increase in MUC5AC in fecal and colonic samples of pigs with SD at the endpoint compared to the controls, but the predictive value for disease progression was limited. Discussion Fucosylation and the impact of dietary fiber may play important roles in the pathogenesis of SD. The lack of predictive value for fecal MUC5AC quantification by ELISA is possibly due to the presence of other non-colonic sources of MUC5AC in the feces. The moderate correlation between IL-17A, neutrophils and MUC5AC confirms its immunoregulatory and mucin stimulatory role. Our study characterizes local alteration of mucin glycosylation in the colonic mucosa of pigs with SD after B. hyodysenteriae infection and may provide insight into host-pathogen interaction.
Collapse
Affiliation(s)
- Susanne Je-Han Lin
- Department of Veterinary Pathology, Iowa State University, Ames, IA, United States
| | - Emma T Helm
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Nicholas K Gabler
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Eric R Burrough
- Department of Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
35
|
Litta F, Papait A, Lucchetti D, Farigu S, Parello A, Tenore CR, Campennì P, Silini AR, Giustiniani MC, Parolini O, Sgambato A, Ratto C. The pathogenesis of cryptoglandular anal fistula: New insight into the immunological profile. Colorectal Dis 2022; 24:1567-1575. [PMID: 35916639 PMCID: PMC10087133 DOI: 10.1111/codi.16290] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/08/2022] [Accepted: 07/27/2022] [Indexed: 01/07/2023]
Abstract
AIM The aetiology of cryptoglandular anal fistula (AF) is poorly understood. Evidence suggests that persistence and/or recurrence of the disease is more related to inflammatory than infectious factors. The aim of this study was to investigate the immune profile of cryptoglandular AF and to perform a histopathological characterization. METHOD Fistulectomy was performed in all patients; healthy ischioanal fat from the same patients was used as a control. Samples were evaluated by the Luminex xMAP system for the detection of 27 analytes. AF tissues were analysed using immunofluorescence. Staining was performed using primary antibodies to identify M1 inflammatory and M2 anti-inflammatory macrophages. Selective staining of total T lymphocytes and different T lymphocyte subsets was performed. RESULTS Twenty patients with AF underwent a fistulectomy. Specific cytokine pathways differentiated AF from healthy tissue: pro-inflammatory cytokines interleukin (IL)-1β, IL-4, IL-8 and IL-17 and the anti-inflammatory cytokine IL-10 were overexpressed in AF compared with controls. Chemokines involved in macrophage recruitment (CCL2, CCL3, CCL4) were higher in AF than in healthy fatty tissue. Moreover, we showed that Tc17 cells characterize AF patients, thus confirming the enzyme-linked immunosorbent assay data. Furthermore, elevated infiltration of CD68+ myeloid cells and a reduction of the M1/M2 ratio characterize AF patients. CONCLUSION A combination of inflammatory cytokines, chemokines and growth factors reside in the wound microenvironment of AF patients. For the first time an important prevalence of Tc17 cells and a reduction in the M1/M2 ratio was observed, thus suggesting new insights into the immunological characterization of AF patients.
Collapse
Affiliation(s)
- Francesco Litta
- Proctology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Andrea Papait
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario 'Agostino Gemelli' IRCCS, Rome, Italy
| | - Donatella Lucchetti
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Serafina Farigu
- Centro di Ricerca E. Menni, Fondazione Poliambulanza, Brescia, Italy
| | - Angelo Parello
- Proctology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Claudio Ricciardi Tenore
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Paola Campennì
- Proctology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | | | - Ornella Parolini
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario 'Agostino Gemelli' IRCCS, Rome, Italy
| | - Alessandro Sgambato
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Carlo Ratto
- Proctology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
36
|
Cui C, Zhang D, Sun K, Zhu Y, Xu J, Kang Y, Zhang G, Cai Y, Mao S, Long R, Ma J, Dong S, Sun Y. Propofol maintains Th17/Treg cell balance in elderly patients undergoing lung cancer surgery through GABAA receptor. BMC Immunol 2022; 23:58. [PMID: 36434505 PMCID: PMC9701037 DOI: 10.1186/s12865-022-00490-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 03/17/2022] [Indexed: 11/27/2022] Open
Abstract
Propofol is widely used in clinical anesthesia due to its advantages of rapid onset and less adverse reactions. This study focused on the role of propofol in the balance of Th17/Treg in elderly patients with lung cancer during perioperative period. Patients undergoing lung cancer surgery were anesthetized by propofol or sevoflurane. Veinal blood was collected at different time points to evaluate the changes of Th17/Treg cell. Propofol better maintained the balance of Th17/Treg in vivo. The peripheral blood of patients with lung cancer was collected in vitro before surgery. Cluster of differentiation (CD)4+ T cells were obtained and then treated with propofol at different concentrations and γ-aminobutyric acid A (GABAA) receptor antagonists. Propofol affected Th17/Treg cell balance by increasing Th17 cells, decreasing Treg cells, thus elevating Th17/Treg ratio, and inhibited invasion and migration of lung cancer cells through GABAA receptor, which was counteracted by GABAA receptor inhibitors. Subsequently, tumor in situ model of lung cancer in aged mice was established. Propofol anesthetized mice had lower change of Th17/Treg ratio, higher survival rate and less metastasis. In brief, propofol regulated balance of Th17/Treg in elderly patients undergoing lung cancer surgery through GABAA receptor. Additionally, propofol could inhibit metastasis of lung cancer.
Collapse
Affiliation(s)
- Can Cui
- grid.413405.70000 0004 1808 0686Department of Anesthesiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 96 DongChuan Road, Guangzhou, 510080 China
| | - Dengwen Zhang
- grid.413405.70000 0004 1808 0686Department of Anesthesiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 96 DongChuan Road, Guangzhou, 510080 China
| | - Ke Sun
- grid.413405.70000 0004 1808 0686Department of Anesthesiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 96 DongChuan Road, Guangzhou, 510080 China
| | - Yi Zhu
- grid.413405.70000 0004 1808 0686Department of Anesthesiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 96 DongChuan Road, Guangzhou, 510080 China
| | - Jindong Xu
- grid.413405.70000 0004 1808 0686Department of Anesthesiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 96 DongChuan Road, Guangzhou, 510080 China
| | - Yin Kang
- grid.413405.70000 0004 1808 0686Department of Anesthesiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 96 DongChuan Road, Guangzhou, 510080 China
| | - Guangyan Zhang
- grid.413405.70000 0004 1808 0686Department of Anesthesiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 96 DongChuan Road, Guangzhou, 510080 China
| | - Yujin Cai
- grid.413405.70000 0004 1808 0686Department of Anesthesiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 96 DongChuan Road, Guangzhou, 510080 China
| | - Songsong Mao
- grid.413405.70000 0004 1808 0686Department of Anesthesiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 96 DongChuan Road, Guangzhou, 510080 China
| | - Ruichun Long
- grid.413405.70000 0004 1808 0686Department of Anesthesiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 96 DongChuan Road, Guangzhou, 510080 China
| | - Jue Ma
- grid.413405.70000 0004 1808 0686Department of Anesthesiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 96 DongChuan Road, Guangzhou, 510080 China
| | - Song Dong
- grid.413405.70000 0004 1808 0686Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 96 DongChuan Road, Guangzhou, 510080 China
| | - Yi Sun
- grid.413405.70000 0004 1808 0686Department of Anesthesiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 96 DongChuan Road, Guangzhou, 510080 China
| |
Collapse
|
37
|
Palomares O, Elewaut D, Irving PM, Jaumont X, Tassinari P. Regulatory T cells and immunoglobulin E: A new therapeutic link for autoimmunity? Allergy 2022; 77:3293-3308. [PMID: 35852798 DOI: 10.1111/all.15449] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 07/07/2022] [Accepted: 07/14/2022] [Indexed: 01/28/2023]
Abstract
Autoimmune diseases have a prevalence of approximately 7 to 9% and are classified as either organ-specific diseases, including type I diabetes, multiple sclerosis, inflammatory bowel disease and myasthenia gravis, or systemic diseases, including systemic lupus erythematosus, rheumatoid arthritis and Sjögren's syndrome. While many advancements have been made in understanding of the mechanisms of autoimmune disease, including the nature of self-tolerance and its breakdown, there remain unmet needs in terms of effective and highly targeted treatments. T regulatory cells (Tregs) are key mediators of peripheral tolerance and are implicated in many autoimmune diseases, either as a result of reduced numbers or altered function. Tregs may be broadly divided into those generated in the thymus (tTregs) and those generated in the periphery (pTregs). Tregs target many different immune cell subsets and tissues to suppress excessive inflammation and to support tissue repair and homeostasis: there is a fine balance between Treg cell stability and the plasticity that is required to adjust Tregs' regulatory purposes to particular immune responses. The central role of immunoglobulin E (IgE) in allergic disease is well recognized, and it is becoming increasingly apparent that this immunoglobulin also has a wider role encompassing other diseases including autoimmune disease. Anti-IgE treatment restores the capacity of plasmacytoid dendritic cells (pDCs) impaired by IgE- high-affinity IgE receptor (FcεR1) cross-linking to induce Tregs in vitro in atopic patients. The finding that anti-IgE therapy restores Treg cell homeostasis, and that this mechanism is associated with clinical improvement in asthma and chronic spontaneous urticaria suggests that anti-IgE therapy may also have a potential role in the treatment of autoimmune diseases in which Tregs are involved.
Collapse
Affiliation(s)
| | - Dirk Elewaut
- Department of Rheumatology, VIB Center for Inflammation Research, Ghent University, Ghent University Hospital, Ghent, Belgium
| | - Peter M Irving
- Guy's and St Thomas' Hospital Foundation Trust, London, UK
- King's College London, London, UK
| | | | | |
Collapse
|
38
|
Chen Y, Hou C, Yang N, Yang Y, Chen Y, Kong D, Jiang Y, Lin M, Zheng S, Li S, Lu M. Regulatory Effect of JAK2/STAT3 on the Immune Function of Endotoxin-tolerant Dendritic Cells and its Involvement in Acute Liver Failure. J Clin Transl Hepatol 2022; 10:879-890. [PMID: 36304491 PMCID: PMC9547265 DOI: 10.14218/jcth.2021.00175] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/19/2021] [Accepted: 11/16/2021] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND AND AIMS Acute liver failure (ALF) is a potentially fatal clinical syndrome with no effective treatment. This study aimed to explore the role of Janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) pathway in modulating the phenotype and immune function of endotoxin-tolerant dendritic cells (ETDCs). In addition, we explored the use of EDTCs in an experimental model of ALF and investigated the associated mechanisms. METHODS In the in vitro experiment, ETDCs were transfected with adenovirus to induce SOCS1+/+ETDCs and SOCS1-/-ETDCs. Thereafter, costimulatory molecules and mixed lymphocyte reaction were assessed. Experimental mice were randomly divided into normal control, ALF, ALF+mock-ETDCs, ALF+SOCS1+/+ETDCs, ALF+AG490, and ALF+AG490+SOCS1+/+ETDCs groups. We examined the therapeutic effect of adoptive cellular immunotherapy by tail-vein injection of target ETDCs 12 h before ALF modeling. AG490, a JAK2/STAT3 inhibitor, was used in the in vivo experiment to further explore the protective mechanism of SOCS1+/+ETDCs. RESULTS Compared with control ETDCs, SOCS1+/+ETDCs had lower expression of costimulatory molecules, weaker allostimulatory ability, lower levels of IL-6 and TNF-α expression and higher IL-10 secretion. SOCS1-/-ETDCs showed the opposite results. In the in vivo experiments, the ALF+SOCS1+/+ETDCs and ALF+AG490+SOCS1+/+ETDCs groups showed less pathological damage and suppressed activation of JAK2/STAT3 pathway. The changes were more pronounced in the ALF+AG490+SOCS1+/+ETDCs group. Infusion of SOCS1+/+ETDCs had a protective effect against ALF possibly via inhibition of JAK2 and STAT3 phosphorylation. CONCLUSIONS The SOCS1 gene had an important role in induction of endotoxin tolerance. SOCS1+/+ETDCs alleviated lipopolysaccharide/D-galactosamine-induced ALF by downregulating the JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Yukai Chen
- Ningbo Puji Hospital, Ningbo, Zhejiang, China
| | - Chaochen Hou
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Naibin Yang
- Department of Infectious Diseases, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Yanyan Yang
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Youran Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Deyong Kong
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuchun Jiang
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Minghao Lin
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Sijie Zheng
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shanshan Li
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Mingqin Lu
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
39
|
Zhong Y, Xiao Q, Kang Z, Huang J, Ge W, Wan Q, Wang H, Zhou W, Zhao H, Liu D. Astragalus polysaccharide alleviates ulcerative colitis by regulating the balance of Tfh/Treg cells. Int Immunopharmacol 2022; 111:109108. [PMID: 35926271 DOI: 10.1016/j.intimp.2022.109108] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/15/2022] [Accepted: 07/26/2022] [Indexed: 11/23/2022]
Abstract
The immunomodulatory function of natural active ingredients has long been a focus of scientific research, with recent hotspots reporting targeted modulation of the follicular helper T cells (Tfh)/regulatory T cells (Treg) balance as an emerging strategy for the treatment of ulcerative colitis (UC). Here, dextran sodium sulfate induced mice UC and Astragalus polysaccharide (APS, 200 mg/kg/day) was administered simultaneously. In this study, APS effectively alleviated colitis in mice by improving survival rate, disease activity index (DAI), the change rate of body weight, colonic length and weight, and histopathological injury of the colon. Moreover, APS regulated the expression of inflammatory cytokines interleukin (IL)-2, IL-6, IL-12p70, IL-23, Tumour necrosis factor (TNF)-ɑ, and transforming growth factor (TGF)-β1 in colonic tissues of colitis mice. Importantly, APS significantly downregulated Tfh cell and the expression of its related nuclear transcription factors Blimp-1 and Bcl-6, and cytokine IL-21. Meanwhile, APS regulated the differentiation of Tfh subpopulations in colitis mice, with Tfh10 and Tfr significantly upregulated while Tfh1, Tfh17, and Tfh21 significantly downregulated. In addition, APS significantly upregulated Treg cells and the levels of its associated nuclear transcription factor Foxp3, and cytokine IL-10 in colitis mice. In conclusion, APS effectively alleviated UC by reshaping the balance of Tfh/Treg cells.
Collapse
Affiliation(s)
- Youbao Zhong
- Formula-Pattern Research Center, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China; Laboratory Animal Research Center for Science and Technology, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China
| | - Qiuping Xiao
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang 330004, Jiangxi Province, China
| | - Zengping Kang
- Formula-Pattern Research Center, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China; Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Jiaqi Huang
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Wei Ge
- Formula-Pattern Research Center, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China; Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Qi Wan
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Haiyan Wang
- Formula-Pattern Research Center, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China; Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Wen Zhou
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China; Nanchang Medical College, Nanchang 330004, Jiangxi Province, China
| | - Haimei Zhao
- Formula-Pattern Research Center, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China; College of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China.
| | - Duanyong Liu
- Formula-Pattern Research Center, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China.
| |
Collapse
|
40
|
Bharti S, Bharti M. The Business of T Cell Subsets and Cytokines in the Immunopathogenesis of Inflammatory Bowel Disease. Cureus 2022; 14:e27290. [PMID: 36039239 PMCID: PMC9407026 DOI: 10.7759/cureus.27290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2022] [Indexed: 12/03/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disorder and one of the most common inflammatory diseases of gastrointestinal (GI) tract in young adults. It is now equally prevalent in western countries as well as in Asian countries. Recently, there has been an increasing IBD burden in low- to middle-income countries as opposed to the earlier notion of this being a disease of the affluents. It occurs due to a variety of factors, namely, local immune alteration, disruption and inflammation of the mucosa, environmental factors, microbial commensals, and pathogen-induced genetic predisposition or genetic alteration in protective factors, etc. So far, an exact etiopathogenesis of IBD is yet to be completely elucidated. Several recent types of research have emphasized the role of altered innate and humoral immunity in its causation, many of them based on animal models of IBD. Due to the poor understanding of its etiopathogenesis, IBD is still a challenge for the treating clinicians leading to persistent and recurrent disease in many cases. Immune dysregulation in the GI tract incited by various pathogenic stimuli has gained great attention from researchers in the field of IBD. This review focuses on highlighting the role of various T cell subsets, their interplay, and associated cytokines involved in the pathogenesis of IBD along with a short description of genetic as well as other immunological factors. A better understanding of the pathogenic factors and subsequent randomized controlled trials targeting these factors is prudent for better therapeutic approaches for IBD.
Collapse
|
41
|
Bednar KJ, Lee JH, Ort T. Tregs in Autoimmunity: Insights Into Intrinsic Brake Mechanism Driving Pathogenesis and Immune Homeostasis. Front Immunol 2022; 13:932485. [PMID: 35844555 PMCID: PMC9280893 DOI: 10.3389/fimmu.2022.932485] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/16/2022] [Indexed: 11/16/2022] Open
Abstract
CD4+CD25highFoxp3+ regulatory T-cells (Tregs) are functionally characterized for their ability to suppress the activation of multiple immune cell types and are indispensable for maintaining immune homeostasis and tolerance. Disruption of this intrinsic brake system assessed by loss of suppressive capacity, cell numbers, and Foxp3 expression, leads to uncontrolled immune responses and tissue damage. The conversion of Tregs to a pathogenic pro-inflammatory phenotype is widely observed in immune mediated diseases. However, the molecular mechanisms that underpin the control of Treg stability and suppressive capacity are incompletely understood. This review summarizes the concepts of Treg cell stability and Treg cell plasticity highlighting underlying mechanisms including translational and epigenetic regulators that may enable translation to new therapeutic strategies. Our enhanced understanding of molecular mechanism controlling Tregs will have important implications into immune homeostasis and therapeutic potential for the treatment of immune-mediated diseases.
Collapse
|
42
|
Resveratrol and resveratrol nano-delivery systems in the treatment of inflammatory bowel disease. J Nutr Biochem 2022; 109:109101. [PMID: 35777588 DOI: 10.1016/j.jnutbio.2022.109101] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 04/04/2022] [Accepted: 06/08/2022] [Indexed: 12/22/2022]
Abstract
Inflammatory bowel disease (IBD) is a chronic disorder associated with the inflammation in the digestive tract. The exact cause of IBD is unknown; nevertheless, in IBD, the homeostasis of key regulatory factors involved in intestinal immunity has been documented to be disrupted. Despite the lack of a viable treatment for IBD, synthetic drugs and monoclonal antibodies are currently used to treat it. However, these treatments have side effects, and the high relapse rate limits their usage. Dietary polyphenols constitute a great variety of compounds and have shown an array of biological properties. Resveratrol is a natural polyphenol found in grapevines and berries. The therapeutic ability of resveratrol against IBD is amply demonstrated in many in vivo studies. Resveratrol can interact with several molecular targets (Nf-kB, SIRT1, mTOR, HIF-1α, miRNAs, and TNF-α) and effectively prevent/ alleviate IBD symptoms with promising results. Although resveratrol has profound anti-inflammatory properties against IBD, its therapeutic employment is limited due to its low water solubility, less chemical stability, less bioavailability, and rapid metabolism in vivo. Hence, resveratrol encapsulation using different carries and its controlled release has become a promising strategy to overcome limitations. Herein, we meticulously review, talk-over the anti-inflammatory effect and mechanisms of resveratrol in IBD. We further provide the latest information on resveratrol formulations and nano-delivery systems used in oral delivery of resveratrol for the treatment of IBD and offer our view on future research on resveratrol in IBD treatment.
Collapse
|
43
|
DNMT3a-Mediated Enterocyte Barrier Dysfunction Contributes to Ulcerative Colitis via Facilitating the Interaction of Enterocytes and B Cells. Mediators Inflamm 2022; 2022:4862763. [PMID: 35574272 PMCID: PMC9106515 DOI: 10.1155/2022/4862763] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 03/04/2022] [Accepted: 04/01/2022] [Indexed: 12/13/2022] Open
Abstract
Objective Dysfunction of the enterocyte barrier is associated with the development of ulcerative colitis (UC). This study was aimed at exploring the effect of DNMT3a on enterocyte barrier function in the progression of UC and the underlying mechanism. Method Mice were given 3.5% dextran sodium sulphate (DSS) in drinking water to induce colitis. The primary intestinal epithelial cells (IECs) were isolated and treated with lipopolysaccharide (LPS) to establish an in vitro inflammatory model. We detected mouse clinical symptoms, histopathological damage, enterocyte barrier function, B cell differentiation, DNA methylation level, and cytokine production. Subsequently, the effect of DNMT3a from IECs on B cell differentiation was explored by a cocultural experiment. Result DSS treatment significantly reduced the body weight and colonic length, increased disease activity index (DAI), and aggravated histopathological damage. In addition, DSS treatment induced downregulation of tight junction (TJ) protein, anti-inflammatory cytokines (IL-10 and TGF-β), and the number of anti-inflammatory B cells (CD1d+) in intestinal epithelial tissues, while upregulated proinflammatory cytokines (IL-6 and TNF-α), proinflammatory B cells (CD138+), and DNA methylation level. Further in vitro results revealed that DNMT3a silencing or TNFSF13 overexpression in IECs partly abolished the result of LPS-induced epithelial barrier dysfunction, as well as abrogated the effect of IEC-regulated B cell differentiation, while si-TACI transfection reversed these effects. Moreover, DNMT3a silencing decreased TNFSF13 methylation level and induced CD1d+ B cell differentiation, and the si-TNFSF13 transfection reversed the trend of B cell differentiation but did not affect TNFSF13 methylation level. Conclusion Our study suggests that DNMT3a induces enterocyte barrier dysfunction to aggravate UC progression via TNFSF13-mediated interaction of enterocyte and B cells.
Collapse
|
44
|
Makled MN, Serrya MS, El-Sheakh AR. Fingolimod ameliorates acetic acid-induced ulcerative colitis: An insight into its modulatory impact on pro/anti-inflammatory cytokines and AKT/mTOR signalling. Basic Clin Pharmacol Toxicol 2022; 130:569-580. [PMID: 35274449 DOI: 10.1111/bcpt.13720] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 02/21/2022] [Accepted: 03/07/2022] [Indexed: 12/20/2022]
Abstract
BACKGROUND Because of the approved immunomodulatory activities of fingolimod, the current study aimed at studying the curative potential of fingolimod against experimentally induced ulcerative colitis (UC) via modulating pro/anti-inflammatory cytokines release and AKT/mTOR signalling. METHODS UC was induced in rats by intracolonic instillation of acetic acid. Fingo (0.5 mg/kg/day, p.o.) was given for 8 consecutive days that started 48 h after UC induction. RESULTS Fingolimod increased body weight growth rate and colon body/weight and colon weight/length indices compared to the UC group. Fingolimod significantly decreased clinical evaluation score and macroscopic score compared to the UC group. The curative potential of fingolimod was further confirmed by histopathological examination revealing marked attenuation of mucosal injury and inflammatory cells infiltration. Fingolimod significantly decreased colon malondialdehyde content and increased colon glutathione contents compared to the UC group. Fingolimod also significantly decreased the expressions of pro-inflammatory cytokines interleukin-9 and T-helper 17 along with increasing the expression of anti-inflammatory interleukin-10 and transforming growth factor-β compared to the UC group. In addition, fingolimod decreased the expressions of AKT and mTOR compared to the UC group. CONCLUSION Fingolimod attenuated acetic acid-induced UC through its immunomodulatory effect by shifting the balance to favour anti-inflammatory cytokine production rather than pro-inflammatory cytokines and modulating the AKT/mTOR signalling.
Collapse
Affiliation(s)
- Mirhan N Makled
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Marwa S Serrya
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Ahmed R El-Sheakh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
45
|
Lee J, Kim D, Min B. Tissue Resident Foxp3+ Regulatory T Cells: Sentinels and Saboteurs in Health and Disease. Front Immunol 2022; 13:865593. [PMID: 35359918 PMCID: PMC8963273 DOI: 10.3389/fimmu.2022.865593] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 02/22/2022] [Indexed: 01/04/2023] Open
Abstract
Foxp3+ regulatory T (Treg) cells are a CD4 T cell subset with unique immune regulatory function that are indispensable in immunity and tolerance. Their indisputable importance has been investigated in numerous disease settings and experimental models. Despite the extensive efforts in determining the cellular and molecular mechanisms operating their functions, our understanding their biology especially in vivo remains limited. There is emerging evidence that Treg cells resident in the non-lymphoid tissues play a central role in regulating tissue homeostasis, inflammation, and repair. Furthermore, tissue-specific properties of those Treg cells that allow them to express tissue specific functions have been explored. In this review, we will discuss the potential mechanisms and key cellular/molecular factors responsible for the homeostasis and functions of tissue resident Treg cells under steady-state and inflammatory conditions.
Collapse
Affiliation(s)
- Juyeun Lee
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Dongkyun Kim
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Booki Min
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- *Correspondence: Booki Min,
| |
Collapse
|
46
|
Hu Y, Ye Z, She Y, Li L, Wu M, Qin K, Li Y, He H, Hu Z, Yang M, Lu F, Ye Q. Efficacy and Safety of Probiotics Combined With Traditional Chinese Medicine for Ulcerative Colitis: A Systematic Review and Meta-Analysis. Front Pharmacol 2022; 13:844961. [PMID: 35321324 PMCID: PMC8936956 DOI: 10.3389/fphar.2022.844961] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/15/2022] [Indexed: 12/24/2022] Open
Abstract
Background: The combination of probiotics and traditional Chinese medicine (TCM) is a prospective therapy for ulcerative colitis (UC), and its efficacy and safety need to be urgently evaluated. Objective: This study aims to comprehensively assess the efficacy and safety of probiotics combined with TCM for the treatment of UC. Methods: The Pubmed, EMBASE, Cochrane library, China Academic Journals (CNKI), Wan-fang database, Chinese biomedical literature service system (CBM), and Chinese Science and Technology Journals (CQVIP) were searched. Subgroup analysis were designed in accordance with different control drugs, treatment courses, and types of probiotics. The Review Manager software (version 5.4.1) was utilized for statistical analysis. Results: 14 original studies containing 1,154 patients were analyzed and showed that probiotics with TCM was more effective than 5-aminosalicylic acid (5-ASA), probiotics or TCM used individually. Moreover, probiotics combined with TCM could inhibit the intestinal inflammation, reduce the recurrence rate and the incidence of adverse events. The subgroup analysis showed that a mixture of different probiotics was more effective than a single strain. Conclusion: It is suggested that probiotics combined with TCM could effectively control clinical symptoms, inhibit intestinal inflammatory response, and finally slow down the disease progress and reduce the disease recurrence with less adverse events. The mixture of different probiotics used in conjunction with individually tailored TCM is a potential clinical strategy for UC.
Collapse
Affiliation(s)
- Yu Hu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhen Ye
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yingqi She
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Linzhen Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mingquan Wu
- Department of Pharmacy, Sichuan Provincial Orthopedic Hospital, Chengdu, China
| | - Kaihua Qin
- Health Preservation and Rehabilitation College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuzheng Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Haiqing He
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhipeng Hu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Maoyi Yang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fating Lu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiaobo Ye
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Qiaobo Ye,
| |
Collapse
|
47
|
Ranjbar R, Ghasemian M, Maniati M, Hossein Khatami S, Jamali N, Taheri-Anganeh M. Gastrointestinal disorder biomarkers. Clin Chim Acta 2022; 530:13-26. [DOI: 10.1016/j.cca.2022.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 01/19/2023]
|
48
|
Angus HCK, Urbano PCM, Laws GA, Fan S, Gadeock S, Schultz M, Butt G, Highton AJ, Kemp RA. An autologous colonic organoid‐derived monolayer model to study immune: bacterial interactions in Crohn's disease patients. Clin Transl Immunology 2022; 11:e1407. [PMID: 35924188 PMCID: PMC9342672 DOI: 10.1002/cti2.1407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/10/2022] [Accepted: 07/13/2022] [Indexed: 12/02/2022] Open
Abstract
Objectives Crohn's disease (CD) initiation and pathogenesis are believed to involve an environmental trigger in a genetically susceptible person that results in an immune response against commensal gut bacteria, leading to a compromised intestinal epithelial barrier and a cycle of inflammation. However, it has been difficult to study the contribution of all factors together in a physiologically relevant model and in a heterogenous patient population. Methods We developed an autologous colonic monolayer model that incorporated the immune response from the same donor and a commensal bacteria, Faecalibacterium prausnitzii. Two‐dimensional monolayers were grown from three‐dimensional organoids generated from intestinal biopsies, and the epithelial integrity of the epithelium was measured using transepithelial electrical resistance. We determined the effect of immune cells alone, bacteria alone and the co‐culture of immune cells and bacteria on integrity. Results Monolayers derived from CD donors had impaired epithelial integrity compared to those from non‐inflammatory bowel disease (IBD) donors. This integrity was further impaired by culture with bacteria, but not immune cells, despite a higher frequency of inflammatory phenotype peripheral T cells in CD donors. Variability in epithelial integrity was higher in CD donors than in non‐IBD donors. Conclusion We have developed a new autologous model to study the complexity of CD, which allows for the comparison of the barrier properties of the colonic epithelium and the ability to study how autologous immune cells directly affect the colonic barrier and whether this is modified by luminal bacteria. This new model allows for the study of individual patients and could inform treatment decisions.
Collapse
Affiliation(s)
- Hamish CK Angus
- Department of Microbiology and Immunology University of Otago Dunedin New Zealand
| | - Paulo CM Urbano
- Department of Microbiology and Immunology University of Otago Dunedin New Zealand
- Department of Medicine University of Otago Dunedin New Zealand
| | - Gemma A Laws
- Department of Microbiology and Immunology University of Otago Dunedin New Zealand
- Department of Medicine University of Otago Dunedin New Zealand
| | - Shijun Fan
- Department of Microbiology and Immunology University of Otago Dunedin New Zealand
- Department of Physiology University of Otago Dunedin New Zealand
| | - Safina Gadeock
- Department of Physiology University of Otago Dunedin New Zealand
- Paediatrics, School of Medicine UC San Diego La Jolla CA USA
| | - Michael Schultz
- Department of Medicine University of Otago Dunedin New Zealand
| | - Grant Butt
- Department of Physiology University of Otago Dunedin New Zealand
| | - Andrew J Highton
- Department of Microbiology and Immunology University of Otago Dunedin New Zealand
- Department of Medicine University of Otago Dunedin New Zealand
| | - Roslyn A Kemp
- Department of Microbiology and Immunology University of Otago Dunedin New Zealand
| |
Collapse
|
49
|
The Effects of Sishen Wan on T Cell Responses in Mice Models of Ulcerative Colitis Induced by Dextran Sodium Sulfate. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:9957709. [PMID: 34956391 PMCID: PMC8702314 DOI: 10.1155/2021/9957709] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 12/24/2022]
Abstract
Currently, it is unclear whether Sishen Wan (SSW) could modulate the balance of Th1 cells, Th17 cells, and Tregs and we evaluated the effects of SSW on T cell responses in mice models of ulcerative colitis (UC). The mice models of acute UC (4% dextran sodium sulfate (DSS), 8 days) and chronic UC (3% DSS, 16 days) with SSW were assayed. Colon tissues were collected for immunohistochemical analysis, enzyme linked immunosorbent assay (ELISA), and flow cytometry (FCM). The expressions of cytokines associated with Tregs, transcription factors of Th17 cells, the frequencies of Th1 cells, Th17 cells, and Tregs, and the functional plasticity of Th17 cells were detected. The frequency of IFN-γ+ T cells was not changed significantly with SSW treatment in acute DSS. In chronic models, the frequency of IFN-γ+ T cells was downregulated with SSW. Meanwhile, the levels of RORγt and the frequency of IL-17A+ Th17 cells showed no significant differences after SSW treatment. Despite no significant effect on the transdifferentiation of Th17 cells in chronic UC models, SSW transdifferentiated Th17 cells into IL-10+ Th17 cells and downregulated IFN-γ+ Th17 cells/IL-10+ Th17 cells in acute DSS. Moreover, there were no significant changes of cytokines secreted by Tregs in acute DSS after SSW treatment, but SSW facilitated the expressions of IL-10 and IL-35, as well as development of IL-10+ Tregs in chronic DSS. SSW showed depressive effects on the immunoreaction of Th17 cells and might promote the conversion of Th17 cells into IL-10+ Th17 cells in acute UC, while it inhibited the excessive reaction of Th1 cells, facilitated the development of Tregs, and enhanced the anti-inflammatory effects in chronic UC.
Collapse
|
50
|
Cheng C, Zhang W, Zhang C, Ji P, Wu X, Sha Z, Chen X, Wang Y, Chen Y, Cheng H, Shi L. Hyperoside Ameliorates DSS-Induced Colitis through MKRN1-Mediated Regulation of PPARγ Signaling and Th17/Treg Balance. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:15240-15251. [PMID: 34878764 DOI: 10.1021/acs.jafc.1c06292] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Hyperoside (HYP), a naturally occurring flavonoid compound, exerts multiple biological functions including myocardial protection, antiredox, and anti-inflammatory activities. However, the role of HYP on inflammatory bowel disease (IBD) and the underlying mechanism need to be further established. Here, we show that HYP treatment profoundly alleviated dextran sulfate sodium-induced ulcerative colitis in mice, characterized by reduced pathological scores, preserved tissue integrity, suppressed colonic inflammation, and balanced Th17/Treg response. Mechanistically, HYP was shown to restrain the expression of the E3 ubiquitin ligase, makorin ring finger protein 1 (MKRN1), which in turn promoted the ubiquitination and proteasomal degradation of peroxisome proliferator-activated receptor gamma (PPARγ), an essential regulator of Th17 and Treg differentiation. Consequently, HYP treatment enhanced PPARγ signaling and hence promoted Treg differentiation while suppressing Th17 cell development during colitis. Thus, our data indicate that HYP acts through the MKRN1/PPARγ axis to modulate the Th17/Treg axis and thereby confers protection against experimental colitis. The findings extend our understanding about HYP action and may provide a potential therapeutic target for IBD.
Collapse
Affiliation(s)
- Cheng Cheng
- Key Laboratory of Inflammation and Immunoregulation, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210046, China
- The First School of Clinical Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Wei Zhang
- Key Laboratory of Inflammation and Immunoregulation, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Cong Zhang
- Key Laboratory of Inflammation and Immunoregulation, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210046, China
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Peng Ji
- Key Laboratory of Inflammation and Immunoregulation, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Xiaohui Wu
- Key Laboratory of Inflammation and Immunoregulation, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Zhou Sha
- Key Laboratory of Inflammation and Immunoregulation, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Xiang Chen
- Key Laboratory of Inflammation and Immunoregulation, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Yongkang Wang
- Key Laboratory of Inflammation and Immunoregulation, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Yugen Chen
- The First School of Clinical Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Haibo Cheng
- The First School of Clinical Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
- Collaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese Medicine, Nanjing 210023, China
| | - Liyun Shi
- Key Laboratory of Inflammation and Immunoregulation, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210046, China
- Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310022, China
| |
Collapse
|