51
|
Fujiwara M, Raheja R, Garo LP, Ajay AK, Kadowaki-Saga R, Karandikar SH, Gabriely G, Krishnan R, Beynon V, Paul A, Patel A, Saxena S, Hu D, Healy BC, Chitnis T, Gandhi R, Weiner HL, Murugaiyan G. microRNA-92a promotes CNS autoimmunity by modulating the regulatory and inflammatory T cell balance. J Clin Invest 2022; 132:e155693. [PMID: 35298438 PMCID: PMC9106347 DOI: 10.1172/jci155693] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/16/2022] [Indexed: 01/11/2023] Open
Abstract
A disequilibrium between immunosuppressive Tregs and inflammatory IL-17-producing Th17 cells is a hallmark of autoimmune diseases, including multiple sclerosis (MS). However, the molecular mechanisms underlying the Treg and Th17 imbalance in CNS autoimmunity remain largely unclear. Identifying the factors that drive this imbalance is of high clinical interest. Here, we report a major disease-promoting role for microRNA-92a (miR-92a) in CNS autoimmunity. miR-92a was elevated in experimental autoimmune encephalomyelitis (EAE), and its loss attenuated EAE. Mechanistically, miR-92a mediated EAE susceptibility in a T cell-intrinsic manner by restricting Treg induction and suppressive capacity, while supporting Th17 responses, by directly repressing the transcription factor Foxo1. Although miR-92a did not directly alter Th1 differentiation, it appeared to indirectly promote Th1 cells by inhibiting Treg responses. Correspondingly, miR-92a inhibitor therapy ameliorated EAE by concomitantly boosting Treg responses and dampening inflammatory T cell responses. Analogous to our findings in mice, miR-92a was elevated in CD4+ T cells from patients with MS, and miR-92a silencing in patients' T cells promoted Treg development but limited Th17 differentiation. Together, our results demonstrate that miR-92a drives CNS autoimmunity by sustaining the Treg/Th17 imbalance and implicate miR-92a as a potential therapeutic target for MS.
Collapse
Affiliation(s)
- Mai Fujiwara
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Radhika Raheja
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Lucien P. Garo
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Amrendra K. Ajay
- Renal Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ryoko Kadowaki-Saga
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sukrut H. Karandikar
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Galina Gabriely
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Rajesh Krishnan
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Vanessa Beynon
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Anu Paul
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Amee Patel
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Shrishti Saxena
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Dan Hu
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Brian C. Healy
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Tanuja Chitnis
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Roopali Gandhi
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Howard L. Weiner
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gopal Murugaiyan
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
52
|
Rao Q, Ma GC, Wu H, Li M, Xu W, Wang GJ, Wang D, Zhang CE, Ma ZJ, Zhang ZT. Dendritic cell combination therapy reduces the toxicity of triptolide and ameliorates colitis in murine models. Drug Deliv 2022; 29:679-691. [PMID: 35225120 PMCID: PMC8890574 DOI: 10.1080/10717544.2022.2044935] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Triptolide (TP) exerts a promising effect in the treatment of ulcerative colitis (UC). However, its toxicity seriously hinders its application in the clinic. Previous studies indicated that dendritic cells (DCs) are the main target through which TP exerts its immunoregulatory effect. Thus, we designed an approach to target DCs in vitro to avoid the direct exposure of organs to TP. Our results revealed that DCs pretreated with TP (DCTP) exerted satisfactory therapeutic effects in mice with colitis, resulting in improved colonic inflammation and alleviated local lesion damage. In addition, no obvious toxicity was observed. DCTP also reshaped the immune milieu by decreasing CD4+ T cell numbers and increasing regulatory T cell numbers in the spleen, mesenteric lymph nodes, peripheral blood and colon; these effects were further confirmed in vitro. Downregulation of CD80/86, ICAM-1, MHCI, TLR2/4, TNF-α, and IL-6 expression and upregulation of programmed cell death ligand 1 (PDL1) and IL-10 expression were observed, indicating that DCs were converted into tolerogenic DCs. In conclusion, DCTP can effectively reduce toxicity and alleviate colonic inflammation and local lesion damage in mice with colitis. The immune mechanism underlying the effects of DCTP included the conversion of DCs into tolerogenic DCs and the alteration of T cell differentiation to produce immunoinhibitory rather than immunostimulatory T cells.
Collapse
Affiliation(s)
- Quan Rao
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Guang-Chao Ma
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Hao Wu
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Meng Li
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wei Xu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Guo-Jun Wang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Dong Wang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Cong-En Zhang
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhi-Jie Ma
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhong-Tao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center for Digestive Diseases, Beijing, China
| |
Collapse
|
53
|
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]
|
54
|
Zhao XX, Ma SB, Wen JP, Hu DT, Gao JS, Peng Q, Zhang YC, Wang JQ, Wang K, Shi XP. Reactive Oxygen Species-Responsive Polyether Micelle Nanomaterials for Targeted Treatment of Ulcerative Colitis. J Biomed Nanotechnol 2022; 18:120-131. [PMID: 35180905 DOI: 10.1166/jbn.2022.3233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
As one of the most challenging inflammatory diseases, the incidence of ulcerative colitis (UC) is increasing year by year, but the existing therapeutic drugs are not effective and lack of targeting. Nanomaterials are expected to become promising delivery system due to their good targeting effects. Here, we designed a nanomaterial sensitive to reactive oxygen species, which can be used to treat IBD, especially UC. It is a self-assembled polyether micelle that can be oxidized at the inflammation site where the concentration of reactive oxygen increases, and effectively release the encapsulated budesonide (Bud). Experiments have proved that for DSS-induced colitis, the synthetic drug-loaded nanoparticles have excellent therapeutic effects, can effectively repair intestinal barrier, and significantly improve the damaged colon tissue. At the same time, it has a beneficial regulatory effect on inflammatory factors. Molecular mechanism studies have shown that it achieves its therapeutic effects by activating the peroxisome proliferators-activated receptors-γ (PPAR-γ) pathway and inhibiting the nuclear factor (NF)-κB pathway. This study proves that oral nano-micelles have an important impact on improving the efficacy of UC treatment drugs and have far-reaching significance for the targeted treatment of gastrointestinal diseases.
Collapse
Affiliation(s)
- Xin-Xin Zhao
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Shan-Bo Ma
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jin-Peng Wen
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Da-Tao Hu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Ju-Shan Gao
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Qiao Peng
- Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yu-Chen Zhang
- Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jin-Qing Wang
- Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Ke Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiao-Peng Shi
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
55
|
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
|
56
|
Hill L, Sharma R, Hart L, Popov J, Moshkovich M, Pai N. The neonatal microbiome in utero and beyond: perinatal influences and long-term impacts. J LAB MED 2021. [DOI: 10.1515/labmed-2021-0131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
The neonatal microbiome offers a valuable model for studying the origins of human health and disease. As the field of metagenomics expands, we also increase our understanding of early life influences on its development. In this review we will describe common techniques used to define and measure the microbiome. We will review in utero influences, normal perinatal development, and known risk factors for abnormal neonatal microbiome development. Finally, we will summarize current evidence that links early life microbial impacts on the development of chronic inflammatory diseases, obesity, and atopy.
Collapse
Affiliation(s)
- Lee Hill
- Department of Paediatrics, Division of Gastroenterology, Hepatology and Nutrition , McMaster Children’s Hospital, McMaster University , Hamilton , Canada
- Department of Human Biology, Division of Exercise Science and Sports Medicine , University of Cape Town , Cape Town , South Africa
| | - Ruchika Sharma
- Department of Paediatrics, Division of Gastroenterology, Hepatology and Nutrition , McMaster Children’s Hospital, McMaster University , Hamilton , Canada
- McMaster University , Hamilton , Canada
| | - Lara Hart
- Department of Paediatrics, Division of Gastroenterology, Hepatology and Nutrition , McMaster Children’s Hospital, McMaster University , Hamilton , Canada
| | - Jelena Popov
- Department of Paediatrics, Division of Gastroenterology, Hepatology and Nutrition , McMaster Children’s Hospital, McMaster University , Hamilton , Canada
- University College Cork, College of Medicine and Health , Cork , Ireland
| | - Michal Moshkovich
- Department of Paediatrics, Division of Gastroenterology, Hepatology and Nutrition , McMaster Children’s Hospital, McMaster University , Hamilton , Canada
- Faculty of Health Sciences , McMaster University , Hamilton , Canada
| | - Nikhil Pai
- Department of Paediatrics, Division of Gastroenterology, Hepatology and Nutrition , McMaster Children’s Hospital, McMaster University , Hamilton , Canada
- Farncombe Family Digestive Health Research Institute , McMaster University , Hamilton , Canada
| |
Collapse
|
57
|
Casalegno Garduño R, Däbritz J. New Insights on CD8 + T Cells in Inflammatory Bowel Disease and Therapeutic Approaches. Front Immunol 2021; 12:738762. [PMID: 34707610 PMCID: PMC8542854 DOI: 10.3389/fimmu.2021.738762] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/16/2021] [Indexed: 12/22/2022] Open
Abstract
CD8+ T cells are involved in the pathogenesis of inflammatory bowel disease (IBD), a complex multifactorial chronic disease. Here, we present an overview of the current research with the controversial findings of CD8+ T cell subsets and discuss some possible perspectives on their therapeutic value in IBD. Studies on the role of CD8+ T cells in IBD have contradictory outcomes, which might be related to the heterogeneity of the cells. Recent data suggest that cytotoxic CD8+ T cells (Tc1) and interleukin (IL) 17-producing CD8+ (Tc17) cells contribute to the pathogenesis of IBD. Moreover, subsets of regulatory CD8+ T cells are abundant at sites of inflammation and can exhibit pro-inflammatory features. Some subsets of tissue resident memory CD8+ T cells (Trm) might be immunosuppressant, whereas others might be pro-inflammatory. Lastly, exhausted T cells might indicate a positive outcome for patients. The function and plasticity of different subsets of CD8+ T cells in health and IBD remain to be further investigated in a challenging field due to the limited availability of mucosal samples and adequate controls.
Collapse
Affiliation(s)
- Rosaely Casalegno Garduño
- Mucosal Immunology Group, Department of Pediatrics, Rostock University Medical Center, Rostock, Germany
| | - Jan Däbritz
- Mucosal Immunology Group, Department of Pediatrics, Rostock University Medical Center, Rostock, Germany.,Center for Immunobiology, Blizard Institute, The Barts and the London School of Medicine and Dentistry, Queen Mary University, London, United Kingdom
| |
Collapse
|
58
|
Bandyopadhyay C, Schecterson L, Gumbiner BM. E-cadherin activating antibodies limit barrier dysfunction and inflammation in mouse inflammatory bowel disease. Tissue Barriers 2021; 9:1940741. [PMID: 34402758 PMCID: PMC8794503 DOI: 10.1080/21688370.2021.1940741] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/28/2021] [Accepted: 06/04/2021] [Indexed: 01/21/2023] Open
Abstract
Deficits in gastrointestinal (GI) paracellular permeability has been implicated in etiology of Inflammatory Bowel Disease (IBD), and E-cadherin, a key component of the epithelial junctional complex, has been implicated in both barrier function and IBD. We have previously described antibodies against E-cadherin that activate cell adhesion, and in this study, we show that they increase transepithelial electrical resistance in epithelial cell monolayers in vitro. We therefore tested the hypothesis that adhesion activating E-cadherin mAbs will enhance epithelial barrier function in vivo and limit progression of inflammation in IBD. Activating mAbs to mouse E-cadherin were tested in different mouse models of IBD including the IL10-/- and adoptive T cell transfer models of colitis. Previously established histological and biomarker measures of inflammation were evaluated to monitor disease progression. Mouse E-cadherin activating mAb treatment reduced total colitis score, individual histological measures of inflammation, and other hallmarks of inflammation compared to control treatment. Activating mAbs also reduced the fecal accumulation lipocalin2 and albumin content, consistent with enhanced barrier function. Therefore, E-cadherin activation could be a potential strategy for limiting inflammation in UC.
Collapse
Affiliation(s)
- Chirosree Bandyopadhyay
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, Washington, United States
| | - Leslayann Schecterson
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, Washington, United States
| | - Barry M Gumbiner
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, Washington, United States
- Department of Pediatrics, University of Washington, Seattle, United States
- Department of Biochemistry, University of Washington, Seattle, United States
| |
Collapse
|
59
|
Porphyromonas gingivalis exacerbates ulcerative colitis via Porphyromonas gingivalis peptidylarginine deiminase. Int J Oral Sci 2021; 13:31. [PMID: 34593756 PMCID: PMC8484350 DOI: 10.1038/s41368-021-00136-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 08/18/2021] [Accepted: 08/29/2021] [Indexed: 11/09/2022] Open
Abstract
Ulcerative Colitis (UC) has been reported to be related to Porphyromonas gingivalis (P. gingivalis). Porphyromonas gingivalis peptidylarginine deiminase (PPAD), a virulence factor released by P. gingivalis, is known to induce inflammatory responses. To explore the pathological relationships between PPAD and UC, we used homologous recombination technology to construct a P. gingivalis strain in which the PPAD gene was deleted (Δppad) and a Δppad strain in which the PPAD gene was restored (comΔppad). C57BL/6 mice were orally gavaged with saline, P. gingivalis, Δppad, or comΔppad twice a week for the entire 40 days (days 0-40), and then, UC was induced by dextran sodium sulfate (DSS) solution for 10 days (days 31-40). P. gingivalis and comΔppad exacerbated DDS-induced colitis, which was determined by assessing the parameters of colon length, disease activity index, and histological activity index, but Δppad failed to exacerbate DDS-induced colitis. Flow cytometry and ELISA revealed that compared with Δppad, P. gingivalis, and comΔppad increased T helper 17 (Th17) cell numbers and interleukin (IL)-17 production but decreased regulatory T cells (Tregs) numbers and IL-10 production in the spleens of mice with UC. We also cocultured P. gingivalis, Δppad, or comΔppad with T lymphocytes in vitro and found that P. gingivalis and comΔppad significantly increased Th17 cell numbers and decreased Treg cell numbers. Immunofluorescence staining of colon tissue paraffin sections also confirmed these results. The results suggested that P. gingivalis exacerbated the severity of UC in part via PPAD.
Collapse
|
60
|
Kim JH, Shin CY, Jang SW, Kim DS, Lee W, Kim HG, Kim HR. Anti-inflammatory effects of DA-9601, an extract of Artemisia asiatica, on aceclofenac-induced acute enteritis. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2021; 25:439-448. [PMID: 34448461 PMCID: PMC8405443 DOI: 10.4196/kjpp.2021.25.5.439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/16/2021] [Accepted: 05/06/2021] [Indexed: 11/15/2022]
Abstract
DA-9601 is an extract obtained from Artemisia asiatica, which has been reported to have anti-inflammatory effects on gastrointestinal lesions; however, its possible anti-inflammatory effects on the small intestine have not been studied yet. Therefore, in this study, we investigated the protective effects of DA-9601 against the ACF-induced small intestinal inflammation. Inflammation of the small intestine was confirmed by histological studies and the changes in the CD4+ T cell fraction induced by the inflammation-related cytokines, and the inflammatory reactions were analyzed. Multifocal discrete small necrotic ulcers with intervening normal mucosa were frequently observed after treatment with ACF. The expression of IL-6, IL-17, and TNF-α genes was increased in the ACF group; however, it was found to have been significantly decreased in the DA-9601 treated group. In addition, DA-9601 significantly decreased the levels of proinflammatory mediators such as IL-1β, GM-CSF, IFN-γ, and TNF-α; the anti-inflammatory cytokine IL-10, on the other hand, was observed to have increased. It is known that inflammatory mediators related to T cell imbalance and dysfunction continuously activate the inflammatory response, causing chronic tissue damage. The fractions of IFN-γ+ Th1 cells, IL-4+ Th2 cells, IL-9+ Th9 cells, IL-17+ Th17 cells, and Foxp3+ Treg cells were significantly decreased upon DA-9601 treatment. These data suggest that the inflammatory response induced by ACF is reduced by DA-9601 via lowering of the expression of genes encoding the inflammatory cytokines and the concentration of inflammatory mediators. Furthermore, DA-9601 inhibited the acute inflammatory response mediated by T cells, resulting in an improvement in ACF-induced enteritis.
Collapse
Affiliation(s)
- Ju Hwan Kim
- Department of Pharmacology, College of Medicine, Dankook University, Cheonan 31116, Korea
| | - Chang Yell Shin
- Research Institute of Dong-A ST Co., Ltd., Yongin 17073, Korea
| | - Sun Woo Jang
- Research Institute of Dong-A ST Co., Ltd., Yongin 17073, Korea
| | - Dong-Seok Kim
- Department of Biochemistry, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Wonae Lee
- Department of Pathology, College of Medicine, Dankook University, Cheonan 31116, Korea
| | - Hyung-Gun Kim
- Department of Pharmacology, College of Medicine, Dankook University, Cheonan 31116, Korea.,NeuroVis Inc., Cheonan 31035, Korea
| | - Hak Rim Kim
- Department of Pharmacology, College of Medicine, Dankook University, Cheonan 31116, Korea
| |
Collapse
|
61
|
Marié IJ, Brambilla L, Azzouz D, Chen Z, Baracho GV, Arnett A, Li HS, Liu W, Cimmino L, Chattopadhyay P, Silverman G, Watowich SS, Khor B, Levy DE. Tonic interferon restricts pathogenic IL-17-driven inflammatory disease via balancing the microbiome. eLife 2021; 10:68371. [PMID: 34378531 PMCID: PMC8376249 DOI: 10.7554/elife.68371] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 08/10/2021] [Indexed: 02/07/2023] Open
Abstract
Maintenance of immune homeostasis involves a synergistic relationship between the host and the microbiome. Canonical interferon (IFN) signaling controls responses to acute microbial infection, through engagement of the STAT1 transcription factor. However, the contribution of tonic levels of IFN to immune homeostasis in the absence of acute infection remains largely unexplored. We report that STAT1 KO mice spontaneously developed an inflammatory disease marked by myeloid hyperplasia and splenic accumulation of hematopoietic stem cells. Moreover, these animals developed inflammatory bowel disease. Profiling gut bacteria revealed a profound dysbiosis in the absence of tonic IFN signaling, which triggered expansion of TH17 cells and loss of splenic Treg cells. Reduction of bacterial load by antibiotic treatment averted the TH17 bias and blocking IL17 signaling prevented myeloid expansion and splenic stem cell accumulation. Thus, tonic IFNs regulate gut microbial ecology, which is crucial for maintaining physiologic immune homeostasis and preventing inflammation.
Collapse
Affiliation(s)
| | | | - Doua Azzouz
- NYU School of Medicine, New York, United States
| | - Ze Chen
- NYU School of Medicine, New York, United States
| | | | - Azlann Arnett
- Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, United States
| | - Haiyan S Li
- University of Texas MD Anderson Cancer Center, Houston, United States
| | - Weiguo Liu
- NYU School of Medicine, New York, United States
| | - Luisa Cimmino
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, United States
| | | | | | | | - Bernard Khor
- Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, United States
| | | |
Collapse
|
62
|
Negi S, Saini S, Tandel N, Sahu K, Mishra RP, Tyagi RK. Translating Treg Therapy for Inflammatory Bowel Disease in Humanized Mice. Cells 2021; 10:1847. [PMID: 34440615 PMCID: PMC8393385 DOI: 10.3390/cells10081847] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023] Open
Abstract
Crohn's disease and ulcerative colitis, two major forms of inflammatory bowel disease (IBD) in humans, afflicted in genetically predisposed individuals due to dysregulated immune response directed against constituents of gut flora. The defective immune responses mounted against the regulatory mechanisms amplify and maintain the IBD-induced mucosal inflammation. Therefore, restoring the balance between inflammatory and anti-inflammatory immunepathways in the gut may contribute to halting the IBD-associated tissue-damaging immune response. Phenotypic and functional characterization of various immune-suppressive T cells (regulatory T cells; Tregs) over the last decade has been used to optimize the procedures for in vitro expansion of these cells for developing therapeutic interventional strategies. In this paper, we review the mechanisms of action and functional importance of Tregs during the pathogenesis of IBD and modulating the disease induced inflammation as well as role of mouse models including humanized mice repopulated with the human immune system (HIS) to study the IBD. "Humanized" mouse models provide new tools to analyze human Treg ontogeny, immunobiology, and therapy and the role of Tregs in developing interventional strategies against IBD. Overall, humanized mouse models replicate the human conditions and prove a viable tool to study molecular functions of human Tregs to harness their therapeutic potential.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Colitis, Ulcerative/genetics
- Colitis, Ulcerative/immunology
- Colitis, Ulcerative/metabolism
- Colitis, Ulcerative/therapy
- Crohn Disease/genetics
- Crohn Disease/immunology
- Crohn Disease/metabolism
- Crohn Disease/therapy
- Disease Models, Animal
- Hematopoietic Stem Cell Transplantation
- Humans
- Mice, Transgenic
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/transplantation
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Sushmita Negi
- Biomedical Parasitology and Nano-Immunology Lab, Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India; (S.N.); (S.S.); (K.S.)
- BERPDC Department, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India
| | - Sheetal Saini
- Biomedical Parasitology and Nano-Immunology Lab, Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India; (S.N.); (S.S.); (K.S.)
| | - Nikunj Tandel
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India;
| | - Kiran Sahu
- Biomedical Parasitology and Nano-Immunology Lab, Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India; (S.N.); (S.S.); (K.S.)
| | - Ravi P.N. Mishra
- BERPDC Department, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India
| | - Rajeev K. Tyagi
- Biomedical Parasitology and Nano-Immunology Lab, Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India; (S.N.); (S.S.); (K.S.)
| |
Collapse
|
63
|
Mei C, Wang X, Meng F, Zhang X, Chen L, Yan S, Xue J, Sun X, Wang Y. Aucuboside Inhibits the Generation of Th17 Cells in Mice Colitis. Front Pharmacol 2021; 12:696599. [PMID: 34335262 PMCID: PMC8322701 DOI: 10.3389/fphar.2021.696599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/14/2021] [Indexed: 12/12/2022] Open
Abstract
Aucuboside is an iridoid glycoside extracted from traditional Chinese medicine such as Rehmannia glutinosa, possessing a wide range of biological activities, including antioxidant, anti-aging, anti-inflammatory, and anti-fibrotic effects. The effects of aucuboside on inflammatory bowel disease (IBD) have not been studied. Therefore, the effects of aucuboside on the generation of Foxp3+ regulatory T (Treg) cells and IL-17–producing T helper (Th17) cells in colitis were studied. A mouse colitis model was established by intracolonic administration of 2,4,6-trinitrobenzene sulfonic acid (TNBS) to mimic human IBD. The generation of Treg and Th17 cells was evaluated by flow cytometry. Aucuboside significantly alleviated colitis symptoms, including weight loss, high disease activity index, and inflammatory responses. The generation of Th17 cells in colitis was significantly inhibited by aucuboside and accompanied by the suppression of IL-17 expression. In Raw264.7 cells, the LPS-induced increase in IL-17 expression was also suppressed by aucuboside, which was significantly blocked by the RORγt inhibitor sr2211. In addition, the decrease in the proportion of Treg cells was also partially reversed by aucuboside, which may reflect the aucuboside-induced inhibition of Th17 cells. This previously unrecognized immunoregulatory function of aucuboside may have clinical applications in IBD.
Collapse
Affiliation(s)
- Chenxue Mei
- Department of Immunology, College of Basic Medical Sciences of China Medical University, Shenyang, China.,Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiao Wang
- Department of Immunology, College of Basic Medical Sciences of China Medical University, Shenyang, China
| | - Fanxiang Meng
- Department of Immunology, College of Basic Medical Sciences of China Medical University, Shenyang, China
| | - Xiaoqing Zhang
- Department of Immunology, College of Basic Medical Sciences of China Medical University, Shenyang, China
| | - Ling Chen
- Department of Gastroenterology, Jinqiu Hospital of Liaoning Province, Shenyang, China
| | - Siqi Yan
- Department of Immunology, College of Basic Medical Sciences of China Medical University, Shenyang, China
| | - Junxiu Xue
- Department of Immunology, College of Basic Medical Sciences of China Medical University, Shenyang, China
| | - Xun Sun
- Department of Immunology, College of Basic Medical Sciences of China Medical University, Shenyang, China
| | - Yuanyuan Wang
- Department of Anesthesiology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
64
|
Ngo Thi Phuong N, Palmieri V, Adamczyk A, Klopfleisch R, Langhorst J, Hansen W, Westendorf AM, Pastille E. IL-33 Drives Expansion of Type 2 Innate Lymphoid Cells and Regulatory T Cells and Protects Mice From Severe, Acute Colitis. Front Immunol 2021; 12:669787. [PMID: 34335571 PMCID: PMC8320374 DOI: 10.3389/fimmu.2021.669787] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/25/2021] [Indexed: 12/14/2022] Open
Abstract
The hallmarks of inflammatory bowel disease are mucosal damage and ulceration, which are known to be high-risk conditions for the development of colorectal cancer. Recently, interleukin (IL)-33 and its receptor ST2 have emerged as critical modulators in inflammatory disorders. Even though several studies highlight the IL-33/ST2 pathway as a key factor in colitis, a detailed mode of action remains elusive. Therefore, we investigated the role of IL-33 during intestinal inflammation and its potential as a novel therapeutic target in colitis. Interestingly, the expression of IL-33, but not its receptor ST2, was significantly increased in biopsies from the inflamed colon of IBD patients compared to non-inflamed colonic tissue. Accordingly, in a mouse model of Dextran Sulfate Sodium (DSS) induced colitis, the secretion of IL-33 significantly accelerated in the colon. Induction of DSS colitis in ST2-/- mice displayed an aggravated colon pathology, which suggested a favorable role of the IL 33/ST2 pathway during colitis. Indeed, injecting rmIL-33 into mice suffering from acute DSS colitis, strongly abrogated epithelial damage, pro-inflammatory cytokine secretion, and loss of barrier integrity, while it induced a strong increase of Th2 associated cytokines (IL-13/IL-5) in the colon. This effect was accompanied by the accumulation of regulatory T cells (Tregs) and type 2 innate lymphoid cells (ILC2s) in the colon. Depletion of Foxp3+ Tregs during IL-33 treatment in DSS colitis ameliorated the positive effect on the intestinal pathology. Finally, IL-33 expanded ILC2s, which were adoptively transferred to DSS treated mice, significantly reduced colonic inflammation compared to DSS control mice. In summary, our results emphasize that the IL-33/ST2 pathway plays a crucial protective role in colitis by modulating ILC2 and Treg numbers.
Collapse
Affiliation(s)
- Nhi Ngo Thi Phuong
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Vittoria Palmieri
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Alexandra Adamczyk
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Robert Klopfleisch
- Institute of Veterinary Pathology, Freie Universitaet Berlin, Berlin, Germany
| | - Jost Langhorst
- Department of Internal and Integrative Medicine, Klinikum Bamberg, Chair for Integrative Medicine, University of Duisburg-Essen, Bamberg, Germany
| | - Wiebke Hansen
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Astrid M. Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Eva Pastille
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
65
|
Wiechers C, Zou M, Galvez E, Beckstette M, Ebel M, Strowig T, Huehn J, Pezoldt J. The microbiota is dispensable for the early stages of peripheral regulatory T cell induction within mesenteric lymph nodes. Cell Mol Immunol 2021; 18:1211-1221. [PMID: 33762684 PMCID: PMC8093251 DOI: 10.1038/s41423-021-00647-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 01/22/2021] [Indexed: 02/07/2023] Open
Abstract
Intestinal Foxp3+ regulatory T cell (Treg) subsets are crucial players in tolerance to microbiota-derived and food-borne antigens, and compelling evidence suggests that the intestinal microbiota modulates their generation, functional specialization, and maintenance. Selected bacterial species and microbiota-derived metabolites, such as short-chain fatty acids (SCFAs), have been reported to promote Treg homeostasis in the intestinal lamina propria. Furthermore, gut-draining mesenteric lymph nodes (mLNs) are particularly efficient sites for the generation of peripherally induced Tregs (pTregs). Despite this knowledge, the direct role of the microbiota and their metabolites in the early stages of pTreg induction within mLNs is not fully elucidated. Here, using an adoptive transfer-based pTreg induction system, we demonstrate that neither transfer of a dysbiotic microbiota nor dietary SCFA supplementation modulated the pTreg induction capacity of mLNs. Even mice housed under germ-free (GF) conditions displayed equivalent pTreg induction within mLNs. Further molecular characterization of these de novo induced pTregs from mLNs by dissection of their transcriptomes and accessible chromatin regions revealed that the microbiota indeed has a limited impact and does not contribute to the initialization of the Treg-specific epigenetic landscape. Overall, our data suggest that the microbiota is dispensable for the early stages of pTreg induction within mLNs.
Collapse
Affiliation(s)
- Carolin Wiechers
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Mangge Zou
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Eric Galvez
- Department Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Michael Beckstette
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine, Helmholtz Centre for Infection Research and Hannover Medical School, Hannover, Germany
| | - Maria Ebel
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Till Strowig
- Department Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Jochen Huehn
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany.
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany.
| | - Joern Pezoldt
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany.
- Laboratory of Systems Biology and Genetics, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| |
Collapse
|
66
|
Dong Y, Yang C, Pan F. Post-Translational Regulations of Foxp3 in Treg Cells and Their Therapeutic Applications. Front Immunol 2021; 12:626172. [PMID: 33912156 PMCID: PMC8071870 DOI: 10.3389/fimmu.2021.626172] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/17/2021] [Indexed: 12/15/2022] Open
Abstract
Regulatory T (Treg) cells are indispensable for immune homeostasis due to their roles in peripheral tolerance. As the master transcription factor of Treg cells, Forkhead box P3 (Foxp3) strongly regulates Treg function and plasticity. Because of this, considerable research efforts have been directed at elucidating the mechanisms controlling Foxp3 and its co-regulators. Such work is not only advancing our understanding on Treg cell biology, but also uncovering novel targets for clinical manipulation in autoimmune diseases, organ transplantation, and tumor therapies. Recently, many studies have explored the post-translational regulation of Foxp3, which have shown that acetylation, phosphorylation, glycosylation, methylation, and ubiquitination are important for determining Foxp3 function and plasticity. Additionally, some of these targets have been implicated to have great therapeutic values. In this review, we will discuss emerging evidence of post-translational regulations on Foxp3 in Treg cells and their exciting therapeutic applications.
Collapse
Affiliation(s)
- Yi Dong
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Cuiping Yang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fan Pan
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, China
| |
Collapse
|
67
|
Lee YM, Seo SH, Cho SY, Choi DH, Cheon MW, Kim HY, Youn DH, Pak SC, Son HS, Na CS. Herbal Medicine and Acupuncture Combined Treatment Attenuates Colitis in Rats. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:965-982. [PMID: 33827383 DOI: 10.1142/s0192415x21500464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This study aimed to verify the efficacy of a combined treatment of Jakyakgamcho-tang (JGT) and acupuncture (CV12, ST25, CV4) on colitis induced by dextrane sulfate sodium (DSS). Changes in immuno-mediated factors and metabolites were investigated. Colitis symptoms such as body weight loss and elevated disease activity index were alleviated by the combined treatment. Moreover, treatment with JGT and acupuncture restored the disturbed architecture of colon by suppressing inflammatory cytokine levels of IFN-[Formula: see text] ([Formula: see text] < 0.05), IL-5 ([Formula: see text] < 0.05), and IL-13 ([Formula: see text] < 0.0001) compared with the DSS group. Analysis of metabolic profiles of serum revealed that treatment groups were clearly separated from the DSS group, suggesting that JGT and acupuncture treatment altered serum metabolites. Furthermore, treatments caused opposite metabolite patterns for dimethylbenzimidazole, 1,5-anhydro-D-glucitol, proline, phosphate, glycolic acid, aspartic acid, tryptophan, phthalic acid, ornithine, and glutamic acid compared with the DSS group. The combined treatment group induced more effective metabolite patterns than the JGT group, implying that acupuncture treatment can restore metabolic changes caused by DSS induction. These results indicate that the simultaneous treatment of JGT administration and acupuncture procedure provides better management of the immune function and inflammatory expression of colitis than a single treatment. It is assumed that intestinal microbial control can be achieved by acupuncture stimulation as well as by taking herbal medicine.
Collapse
Affiliation(s)
- Yu-Mi Lee
- College of Korean Medicine, Dongshin University, Naju, Jeollanam-do 58245, Korea
| | - Seung-Ho Seo
- College of Korean Medicine, Dongshin University, Naju, Jeollanam-do 58245, Korea
| | - Seong-Young Cho
- College of Korean Medicine, Dongshin University, Naju, Jeollanam-do 58245, Korea
| | - Dong-Hee Choi
- College of Korean Medicine, Dongshin University, Naju, Jeollanam-do 58245, Korea
| | - Min-Woo Cheon
- Department of Health Administration, Dongshin University, Naju, Jeollanam-do, Korea
| | - Hee-Young Kim
- College of Korean Medicine Daegu Haany University Daegu 42158, Korea
| | - Dae-Hwan Youn
- College of Korean Medicine, Dongshin University, Naju, Jeollanam-do 58245, Korea
| | - Sok Cheon Pak
- School of Biomedical Sciences, Charles Sturt University, Bathurst, NSW 2795, Australia
| | - Hong-Seok Son
- Department of Food Biosciences and Technology, Korea University, Seoul 02841, Korea
| | - Chang-Su Na
- College of Korean Medicine, Dongshin University, Naju, Jeollanam-do 58245, Korea
| |
Collapse
|
68
|
Synthesis of norisoboldine derivatives and bioactivity assay for inducing the generation of regulatory T cells. Bioorg Med Chem Lett 2021; 37:127844. [PMID: 33556569 DOI: 10.1016/j.bmcl.2021.127844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/16/2021] [Accepted: 01/24/2021] [Indexed: 02/07/2023]
Abstract
In this study, we used chemical modification to improve the pharmacological activity of norisoboldine (NOR). A new NOR-benzoic acid derivative, named DC-01, showed more potent induction of Treg cell differentiation than NOR. The in vitro effective concentration of DC-01 (1 μM) is about an order of magnitude lower than that of NOR (10 μM). DC-01 (28, 56 mg/kg) showed better amelioration of dextran sodium sulfate-induced colitis in mice than NOR (20, 40 mg/kg), and DC-01 (28 mg/kg) increased the number of Treg cells slightly better than NOR (20 mg/kg). In summary, DC-01 exerts more potent induction of Treg cell generation, which might be a candidate drug for the treatment of inflammation- and immune-related diseases.
Collapse
|
69
|
Türei D, Valdeolivas A, Gul L, Palacio‐Escat N, Klein M, Ivanova O, Ölbei M, Gábor A, Theis F, Módos D, Korcsmáros T, Saez‐Rodriguez J. Integrated intra- and intercellular signaling knowledge for multicellular omics analysis. Mol Syst Biol 2021; 17:e9923. [PMID: 33749993 PMCID: PMC7983032 DOI: 10.15252/msb.20209923] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 02/11/2021] [Accepted: 02/15/2021] [Indexed: 12/12/2022] Open
Abstract
Molecular knowledge of biological processes is a cornerstone in omics data analysis. Applied to single-cell data, such analyses provide mechanistic insights into individual cells and their interactions. However, knowledge of intercellular communication is scarce, scattered across resources, and not linked to intracellular processes. To address this gap, we combined over 100 resources covering interactions and roles of proteins in inter- and intracellular signaling, as well as transcriptional and post-transcriptional regulation. We added protein complex information and annotations on function, localization, and role in diseases for each protein. The resource is available for human, and via homology translation for mouse and rat. The data are accessible via OmniPath's web service (https://omnipathdb.org/), a Cytoscape plug-in, and packages in R/Bioconductor and Python, providing access options for computational and experimental scientists. We created workflows with tutorials to facilitate the analysis of cell-cell interactions and affected downstream intracellular signaling processes. OmniPath provides a single access point to knowledge spanning intra- and intercellular processes for data analysis, as we demonstrate in applications studying SARS-CoV-2 infection and ulcerative colitis.
Collapse
Affiliation(s)
- Dénes Türei
- Faculty of Medicine and Heidelberg University HospitalInstitute of Computational BiomedicineHeidelberg UniversityHeidelbergGermany
| | - Alberto Valdeolivas
- Faculty of Medicine and Heidelberg University HospitalInstitute of Computational BiomedicineHeidelberg UniversityHeidelbergGermany
| | | | - Nicolàs Palacio‐Escat
- Faculty of Medicine and Heidelberg University HospitalInstitute of Computational BiomedicineHeidelberg UniversityHeidelbergGermany
- Faculty of MedicineJoint Research Centre for Computational Biomedicine (JRC‐COMBINE)RWTH Aachen UniversityAachenGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Michal Klein
- Institute of Computational BiologyHelmholtz Zentrum MünchenNeuherbergGermany
| | - Olga Ivanova
- Faculty of Medicine and Heidelberg University HospitalInstitute of Computational BiomedicineHeidelberg UniversityHeidelbergGermany
| | - Márton Ölbei
- Earlham InstituteNorwichUK
- Quadram Institute BioscienceNorwichUK
| | - Attila Gábor
- Faculty of Medicine and Heidelberg University HospitalInstitute of Computational BiomedicineHeidelberg UniversityHeidelbergGermany
| | - Fabian Theis
- Institute of Computational BiologyHelmholtz Zentrum MünchenNeuherbergGermany
- Department of MathematicsTechnical University of MunichGarchingGermany
| | - Dezső Módos
- Earlham InstituteNorwichUK
- Quadram Institute BioscienceNorwichUK
| | | | - Julio Saez‐Rodriguez
- Faculty of Medicine and Heidelberg University HospitalInstitute of Computational BiomedicineHeidelberg UniversityHeidelbergGermany
- Faculty of MedicineJoint Research Centre for Computational Biomedicine (JRC‐COMBINE)RWTH Aachen UniversityAachenGermany
| |
Collapse
|
70
|
Zhou C, Zheng X, Huang X, Su J, Li M, Chen Z, Li M, Chi H. [ Huangqin decoction alleviates ulcerative colitis by regulating ILC3s-TH cell response]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:256-263. [PMID: 33624600 DOI: 10.12122/j.issn.1673-4254.2021.02.14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To explore the effect of Huangqin decoction (HQD) on group 3 innate lymphoid cells (ILC3s) and helper T cells (Th) for treatment of ulcerative colitis (UC). OBJECTIVE Male Balb/c mice were randomly divided into control group, DSS group, mesalazine group (ME, 400 mg/kg), and 2.275 g/kg, 4.55 g/kg and 9.1 g/kg HQD groups. All the mice were given free access to normal chow. Except for those in the normal control group, all the mice were given 3% DSS solution for 7 days to establish models of UC. The mice in ME group and 3 HQD groups were given mesalazine or HQD via oral gavage at the specified doses once a day. Flow cytometry was performed to analyze the ILC3s, MHC Ⅱ, Th1 and Treg in the lamina propria lymphocytes in the colon. Milliplex was performed to determine cytokine levels of in the colon tissues. OBJECTIVE Compared with those in DSS group, the mice in the 3 HQD groups all showed obviously lessened symptoms of UC with significantl decreased DAI score (P < 0.001) and macroscopic score (P < 0.001). The results of flow cytometry showed that HQD treatment significantly increased the percentage of ILC3s (P < 0.05) and expression of MHCⅡ (P < 0.05), obviously reduced the proportion of Th1 (P < 0.05) but increased Treg cells (P < 0.05) in the colon tissues. Milliplex showed that HQD treatment significantly reduced the expressions of Th-related pro-inflammatory cytokines including IL-2 (P < 0.05), IL-17A (P < 0.05), IL-23 (P < 0.05), TNF-α (P < 0.05), and IFN-γ (P < 0.05). OBJECTIVE HQD alleviates DSS- induced UC in mice by increasing ILC3s and MHC Ⅱ expression to suppress the function of Th17 and Th1 cells and promote Treg and Th2 cells.
Collapse
Affiliation(s)
- C Zhou
- First Clinical Medical College, Guangdong Medical University, Zhanjiang 524023, China
| | - X Zheng
- School Of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Dongguan Mathematical Engineering Academy of Chinese Medicine, Guangzhou University Chinese Medicine, Dongguan 523000, China
| | - X Huang
- School Of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Dongguan Mathematical Engineering Academy of Chinese Medicine, Guangzhou University Chinese Medicine, Dongguan 523000, China
| | - J Su
- Research Laboratory, Foshan Women and Children's Hospital, Affiliated Foshan Women and Children's Hospital of Southern Medical University, Foshan 528000, China
| | - M Li
- School Of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Z Chen
- Dongguan Mathematical Engineering Academy of Chinese Medicine, Guangzhou University Chinese Medicine, Dongguan 523000, China
| | - M Li
- School Of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - H Chi
- Second School of Clinical Medicine, Guangdong Medical University, Dongguan 523000, China
| |
Collapse
|
71
|
Extracellular Vesicles from Thapsigargin-Treated Mesenchymal Stem Cells Ameliorated Experimental Colitis via Enhanced Immunomodulatory Properties. Biomedicines 2021; 9:biomedicines9020209. [PMID: 33670708 PMCID: PMC7922639 DOI: 10.3390/biomedicines9020209] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/15/2021] [Accepted: 02/15/2021] [Indexed: 02/08/2023] Open
Abstract
Therapeutic applications of extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) have attracted considerable attention because of their immunomodulatory properties against immune-mediated, inflammatory diseases. Here, we demonstrated enhanced immunomodulatory properties of EVs secreted from endoplasmic reticulum (ER) stress inducer thapsigargin (TSG)-primed human Wharton's jelly-derived MSCs (WJ-MSCs). EVs from TSG-primed WJ-MSCs (TSG-EV) showed increased yield and expression of immunomodulatory factors, such as transforming growth factor-β1 (TGFβ), cyclooxygenase-2 (COX2), and especially indoleamine 2,3-dioxygenase (IDO), compared to control EVs. TSG-EV showed a significantly enhanced immunosuppressive effect on human peripheral blood-derived T cell proliferation and Th1 and Th17 differentiation, whereas Treg and M2-type macrophage were enriched compared to a control EV-treated group. Furthermore, TSG-EV substantially mitigated mouse experimental colitis by reducing the inflammatory response and maintaining intestinal barrier integrity. A significant increase of Tregs and M2-type macrophages in colitic colons of a TSG-EV-treated mouse suggests an anti-inflammatory effect of TSG-EV in colitis model, possibly mediated by Treg and macrophage polarization. These data indicate that TSG treatment promoted immunomodulatory properties of EVs from WJ-MSCs, and TSG-EV may provide a new therapeutic approach for treatment of colitis.
Collapse
|
72
|
Abstract
OBJECTIVES Villous atrophy (VA) is not pathognomonic of celiac disease (CD). We aimed at reporting distribution, clinical, and immunohistochemical features of seronegative VA (SNVA) in a pediatric population. METHODS We retrospectively collected data from patients who underwent intestinal biopsies between 2010 and 2017 and showed VA without serum CD-associated autoantibodies. Marsh-Oberhuber grading was used. Density of intraepithelial lymphocytes (IELs) expressing CD3 or TCRγδ+ receptor and of lamina propria CD25+ cells was assessed by immunohistochemistry. Intestinal deposits of anti-tissue tranglutaminase2 (anti-TG2) were also investigated by double immunofluorescence. RESULTS Over a 7-year period, 64 out of 1282 patients with VA had negative serum CD serology. Diagnoses were: inflammatory bowel diseases (IBD) (21/64), Gastro-Esophageal Reflux Disease (GERD) (12/64), food allergy (8/64), infections (7/64, of which 3 HIV infections), immune deficiency (3/64), short bowel syndrome (3/64), congenital diarrhea (2/64), other/inconclusive diagnosis (8/64). Forty-four, 15, and 5 showed Marsh 3a, 3b, and 3c lesion, respectively. The latter category included 2 patients with Crohn disease, 2 with immunodeficiencies, 1 with lymphohistiocytosis. In 41/46 (89%) patients, mononuclear CD25+ cells were above the cut-off, indicating mucosal inflammation but only 18/46 (39%) had IELs and TCRγδ + IELs above limits of normality. In 10 of 46 (22%) patients, a positive immunofluorescence indicated the presence of anti-TG2 mucosal antibodies. CONCLUSIONS SNVA is not rare representing up to 5% of the cases of VA. Most patients have a Marsh 3a lesion. Immunohistochemical analysis may be helpful in excluding CD, whereas the finding of mucosal anti-TG2, particularly with a weak staining, shows no absolute specificity for CD.
Collapse
|
73
|
Deng L, Wojciech L, Gascoigne NRJ, Peng G, Tan KSW. New insights into the interactions between Blastocystis, the gut microbiota, and host immunity. PLoS Pathog 2021; 17:e1009253. [PMID: 33630979 PMCID: PMC7906322 DOI: 10.1371/journal.ppat.1009253] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The human gut microbiota is a diverse and complex ecosystem that is involved in beneficial physiological functions as well as disease pathogenesis. Blastocystis is a common protistan parasite and is increasingly recognized as an important component of the gut microbiota. The correlations between Blastocystis and other communities of intestinal microbiota have been investigated, and, to a lesser extent, the role of this parasite in maintaining the host immunological homeostasis. Despite recent studies suggesting that Blastocystis decreases the abundance of beneficial bacteria, most reports indicate that Blastocystis is a common component of the healthy gut microbiome. This review covers recent finding on the potential interactions between Blastocystis and the gut microbiota communities and its roles in regulating host immune responses.
Collapse
Affiliation(s)
- Lei Deng
- Laboratory of Molecular and Cellular Parasitology, Healthy Aging Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Lukasz Wojciech
- Immunology Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Nicholas R. J. Gascoigne
- Immunology Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Guangneng Peng
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Kevin S. W. Tan
- Laboratory of Molecular and Cellular Parasitology, Healthy Aging Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
74
|
The therapeutic efficacy of mesenchymal stromal cells on experimental colitis was improved by the IFN-γ and poly(I:C) priming through promoting the expression of indoleamine 2,3-dioxygenase. Stem Cell Res Ther 2021; 12:37. [PMID: 33413597 PMCID: PMC7791665 DOI: 10.1186/s13287-020-02087-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 12/07/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Inflammatory bowel disease is a chronic and excessive inflammation of the colon and small intestine. We previously reported that priming of mesenchymal stromal cells (MSCs) with poly(I:C) induced them to express indoleamine 2,3-dioxygenase (IDO). We tried to find out whether the IFN-γ and poly(I:C)-primed MSCs have better therapeutic efficacy on the experimental colitis in the IDO1-dependent manner. METHODS To compare the therapeutic effects between the unstimulated MSCs and primed MSCs on murine colitis, mice (C57BL6) were administered with 2.5% dextran sodium sulfate (DSS) in drinking water for 5 days and injected with MSCs intraperitoneally on days 1 and 3 following DSS ingestion. The disease activity index score and body weight loss were assessed daily until day 9. RESULTS Mice receiving the IFN-γ and poly(I:C)-primed MSCs showed a reduced disease activity index and less weight loss. Colon tissue from the same mice presented attenuated pathological damage, increased Paneth cells, increased IDO1-expressing cells, and better proliferation of enterocytes. The primed MSC treatment upregulated the mRNA expression of intestinal stem cell markers (Lgr5, Olfm4, and Bmi1), enterocyte differentiation markers (Muc2, Alpi, Chga, and occludin), and regulatory T (Treg) cells (Foxp3). The same treatment decreased inflammatory cell infiltration to lymphoid organs and the level of pro-inflammatory cytokines (IL-1β, TNF-α, IL-6, and MCP-1) in colon tissue. Notably, in vivo pharmacologic inhibition of the IDO1 activity blocked the Foxp3 upregulation in colon tissue and diminished the protective effects of the primed MSC. CONCLUSIONS The priming of MSCs with the IFN-γ and poly(I:C) is a promising new strategy to improve the therapeutic efficacy of MSC and is worth further research.
Collapse
|
75
|
Liu C, Huang S, Wu Z, Li T, Li N, Zhang B, Han D, Wang S, Zhao J, Wang J. Cohousing-mediated microbiota transfer from milk bioactive components-dosed mice ameliorate colitis by remodeling colonic mucus barrier and lamina propria macrophages. Gut Microbes 2021; 13:1-23. [PMID: 33789528 PMCID: PMC8018355 DOI: 10.1080/19490976.2021.1903826] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 03/07/2021] [Accepted: 03/09/2021] [Indexed: 02/08/2023] Open
Abstract
Human milk oligosaccharides (HMOs) and milk fat globule membrane (MFGM) are highly abundant in breast milk, and have been shown to exhibit potent immunomodulatory effects. Yet, their role in the gut microbiota modulation in relation to colitis remains understudied. Since the mixtures of fructo-oligosaccharides (FOS) and galacto-oligosaccharides (GOS) perfectly mimic the properties and functions of HMOs, the combination of MFGM, FOS, and GOS (CMFG) has therefore been developed and used in this study. Here, CMFG were pre-fed to mice for three weeks to investigate its preventive effect on dextran sodium sulfate (DSS) induced colitis. Moreover, CMFG-treated and vehicle-treated mice were cohoused to further elucidate the preventive role of the gut microbiota transfer in colitis. At the end of the study, 16S rDNA gene amplicon sequencing, short-chain fatty acids (SCFAs) profiling, transcriptome sequencing, histological analysis, immunofluorescence staining and flow cytometry analysis were conducted. Our results showed that CMFG pre-supplementation alleviated DSS-induced colitis as evidenced by decreased disease activity index (DAI) score, reduced body weight loss, increased colon length and mucin secretion, and ameliorated intestinal damage. Moreover, CMFG reduced macrophages in the colon, resulting in decreased levels of IL-1β, IL-6, IL-8, TNF-α, and MPO in the colon and circulation. Furthermore, CMFG altered the gut microbiota composition and promoted SCFAs production in DSS-induced colitis. Markedly, the cohousing study revealed that transfer of gut microbiota from CMFG-treated mice largely improved the DSS-induced colitis as evidenced by reduced intestinal damage and decreased macrophages infiltration in the colon. Moreover, transfer of the gut microbiota from CMFG-treated mice protected against DSS-induced gut microbiota dysbiosis and promotes SCFAs production, which showed to be associated with colitis amelioration. Collectively, these findings demonstrate the beneficial role of CMFG in the gastrointestinal diseases, and further provide evidence for the rational design of effective prophylactic functional diets in both animals and humans.
Collapse
Affiliation(s)
- Cong Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shimeng Huang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhenhua Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Tiantian Li
- Academy of National Food and Strategic Reserves Administration, Beijing, China
| | - Na Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Bing Zhang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shilan Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jiangchao Zhao
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR, USA
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
76
|
Poly(ADP-ribose) polymerase-1 inhibitor ameliorates dextran sulfate sodium-induced colitis in mice by regulating the balance of Th17/Treg cells and inhibiting the NF- κB signaling pathway. Exp Ther Med 2020; 21:134. [PMID: 33376516 PMCID: PMC7751469 DOI: 10.3892/etm.2020.9566] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023] Open
Abstract
Poly(ADP-ribose) polymerase-1 (PARP-1) plays a critical role in inflammatory pathways. The PARP-1 inhibitor, 5-aminoisoquinolinone (5-AIQ), has been demonstrated to exert significant pharmacological effects. The present study aimed to further examine the potential mechanisms of 5-AIQ in a mouse model of dextran sodium sulfate (DSS)-induced colitis. Colitis conditions were assessed by changes in weight, disease activity index, colon length, histopathology and pro-inflammatory mediators. The colonic expression of PARP/NF-κB and STAT3 pathway components was measured by western blot analysis. Flow cytometry was used to analyze the proportion of T helper 17 cells (Th17) and regulatory T cells (Tregs) in the spleen. Western blot analysis and reverse transcription-quantitative PCR were employed to determine the expression of the transcription factors retinoic acid-related orphan receptor and forkhead box protein P3. The results demonstrated that 5-AIQ reduced tissue damage and the inflammatory response in mice with experimental colitis. Moreover, 5-AIQ increased the proportion of Treg cells and decreased the percentage of Th17 cells in the spleen. Furthermore, following 5-AIQ treatment, the main components of the PARP/NF-κB and STAT3 pathways were downregulated. Collectively, these results demonstrate that the PARP-1 inhibitor, 5-AIQ, may suppress intestinal inflammation and protect the colonic mucosa by modulating Treg/Th17 immune balance and inhibiting PARP-1/NF-κB and STAT3 signaling pathways in mice with experimental colitis.
Collapse
|
77
|
Araruna ME, Serafim C, Alves Júnior E, Hiruma-Lima C, Diniz M, Batista L. Intestinal Anti-Inflammatory Activity of Terpenes in Experimental Models (2010-2020): A Review. Molecules 2020; 25:molecules25225430. [PMID: 33233487 PMCID: PMC7699610 DOI: 10.3390/molecules25225430] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 09/26/2020] [Accepted: 09/28/2020] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel diseases (IBDs) refer to a group of disorders characterized by inflammation in the mucosa of the gastrointestinal tract, which mainly comprises Crohn’s disease (CD) and ulcerative colitis (UC). IBDs are characterized by inflammation of the intestinal mucosa, are highly debilitating, and are without a definitive cure. Their pathogenesis has not yet been fully elucidated; however, it is assumed that genetic, immunological, and environmental factors are involved. People affected by IBDs have relapses, and therapeutic regimens are not always able to keep symptoms in remission over the long term. Natural products emerge as an alternative for the development of new drugs; bioactive compounds are promising in the treatment of several disorders, among them those that affect the gastrointestinal tract, due to their wide structural diversity and biological activities. This review compiles 12 terpenes with intestinal anti-inflammatory activity evaluated in animal models and in vitro studies. The therapeutic approach to IBDs using terpenes acts basically to prevent oxidative stress, combat dysbiosis, restore intestinal permeability, and improve the inflammation process in different signaling pathways.
Collapse
Affiliation(s)
- Maria Elaine Araruna
- Postgraduate Program in Natural Products and Bioactive Synthetic, Health Sciences Center, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil; (M.E.A.); (C.S.); (E.A.J.); (M.D.)
| | - Catarina Serafim
- Postgraduate Program in Natural Products and Bioactive Synthetic, Health Sciences Center, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil; (M.E.A.); (C.S.); (E.A.J.); (M.D.)
| | - Edvaldo Alves Júnior
- Postgraduate Program in Natural Products and Bioactive Synthetic, Health Sciences Center, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil; (M.E.A.); (C.S.); (E.A.J.); (M.D.)
| | - Clelia Hiruma-Lima
- Department of Structural and Functional Biology (Physiology), Institute of Biosciences, São Paulo State University, Botucatu 18618-970, SP, Brazil;
| | - Margareth Diniz
- Postgraduate Program in Natural Products and Bioactive Synthetic, Health Sciences Center, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil; (M.E.A.); (C.S.); (E.A.J.); (M.D.)
- Department of Pharmacy, Health Sciences Center, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
| | - Leônia Batista
- Postgraduate Program in Natural Products and Bioactive Synthetic, Health Sciences Center, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil; (M.E.A.); (C.S.); (E.A.J.); (M.D.)
- Department of Pharmacy, Health Sciences Center, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
- Correspondence: ; Tel.: +55-83-32167003; Fax: +55-83-32167502
| |
Collapse
|
78
|
Abbasi-Kenarsari H, Heidari N, Baghaei K, Amani D, Zali MR, Gaffari Khaligh S, Shafiee A, Hashemi SM. Synergistic therapeutic effect of mesenchymal stem cells and tolerogenic dendritic cells in an acute colitis mouse model. Int Immunopharmacol 2020; 88:107006. [PMID: 33182049 DOI: 10.1016/j.intimp.2020.107006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 08/24/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023]
Abstract
Cell-based therapy with tolerizing cells has been applied for the treatment of inflammatory bowel disease (IBD) in previous experimental and clinical studies with promising results. In the current study, we utilized the dextran sulfate sodium (DSS)-induced colitis model, to investigate if tolerogenic dendritic cell-mesenchymal stem cell (tDC-MSC) combination therapy can augment the therapeutic effects of single transplantation of each cell type. The effect of MSC and tDC co-transplantation on the severity of colitis was assessed by daily monitoring of body weight, stool consistency, and rectal bleeding, and compared with control groups. Moreover, the colon length, colon weight, myeloperoxidase (MPO) activity were measured and evaluated with histological analysis of colon tissues. The Treg cell percentage and cytokine levels in spleens and mesenteric lymph nodes (MLNs) were measured by flow cytometry and ELISA, respectively. The results showed co-transplantation of MSCs and tDCs was more effective in alleviating the clinical and histological manifestations of colitis than monotherapy, especially when compared with MSC alone. The protective effects of tDC-MSC were accompanied by the induction of Treg cells and increased the production of anti-inflammatory cytokines in spleens and mesenteric lymph nodes. Together, co-transplantation of MSCs and tDCs could be a promising and effective therapeutic approach in the treatment of IBD.
Collapse
Affiliation(s)
- Hajar Abbasi-Kenarsari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Heidari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorder Research Center, Research Institute for Gastroenterology and Liver Disease, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Davar Amani
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Disease Research Center, Research Institute for Gastroenterology and Liver Disease, Shahid Beheshti University of Medical Science, Tehran, Iran
| | | | - Abbas Shafiee
- UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
79
|
Liao F, Lu X, Dong W. Exosomes derived from T regulatory cells relieve inflammatory bowel disease by transferring miR-195a-3p. IUBMB Life 2020; 72:2591-2600. [PMID: 33108032 DOI: 10.1002/iub.2385] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/27/2020] [Accepted: 08/29/2020] [Indexed: 12/14/2022]
Abstract
Previous studies have demonstrated the therapeutic effects of regulatory T (Treg) cells on inflammatory bowel disease (IBD), but the mechanism is not well-understood. Exosomes have been proposed as a novel mechanism underlying the action of Tregs. This study aimed to investigate the therapeutic effects of exosomes secreted by Treg cells (Treg-Exo) on IBD and to explore the underlying mechanism. Treg-Exo was isolated from BALB/c mouse spleen mononuclear cells and then injected into a murine model of IBD induced by dextran sodium sulfate (DSS) exposure. A co-culture model of Treg-Exo and colonic epithelial YAMC cells in the presence of TNF-α was used to investigate the communication between Tregs and intestinal epithelial cells. in vitro results showed that Treg-Exo could be transferred to YAMC cells where Treg-Exo promoted cell proliferation and inhibited cell apoptosis. Animal experiments showed that Treg-Exo administration alleviated the DSS-induced IBD in mice. The therapeutic effects of Treg-Exo both in vitro and in vivo were eliminated when miR-195a-3p expression was inhibited in Treg-Exo. The pro-apoptotic Caspase 12 was identified as a direct target of miR-195a-3p. In conclusion, Treg-Exo alleviated the DSS-induced IBD through transferring miR-195a-3p.
Collapse
Affiliation(s)
- Fei Liao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaohong Lu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
80
|
Vent-Schmidt J, Goldsmith LJ, Steiner TS. Patients' Willingness and Perspectives Toward Chimeric Antigen Receptor T-Regulatory Cell Therapy for Inflammatory Bowel Diseases. CROHN'S & COLITIS 360 2020; 2:otaa085. [PMID: 36777762 PMCID: PMC9802168 DOI: 10.1093/crocol/otaa085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Indexed: 11/14/2022] Open
Abstract
Background Inflammatory bowel disease is a life-changing disease resulting from recurrent intestinal inflammation. Current therapies (eg, steroids and biologics) are associated with mild to severe side effects, and none provide a cure. Recent research has focused on genetically engineering gut-specific anti-inflammatory T-regulatory cells (CAR-Tregs) to control intestinal inflammation, a logistically and conceptually complex approach. The purpose of our study was to understand patients' willingness to try CAR-Treg given 2 hypothetical scenarios-in a clinical trial or as a new treatment. Methods We surveyed people living with inflammatory bowel disease about their willingness to try CAR-Treg. The online survey was developed using patient focus groups and associated literature. We recruited participants through email and social media. We used descriptive and inferential statistics to analyze closed-ended questions and inductive thematic analysis to analyze open-ended follow-up questions. Results Survey participants indicated high willingness to try CAR-Treg therapy in both a clinical trial and as a new treatment. Willingness to try was not correlated with disease state or medication history. Women were less likely than men to indicate willingness to participate in a clinical trial. Participants' reasons for being willing to try CAR-Treg therapy included the wish to change their current treatment and the calling to participate in research. Participants that were not willing to try CAR-Treg mentioned the lack of long-term data and the success of their current therapy. Conclusions This is the first study to our knowledge to investigate patient willingness to try CAR-Treg therapy. Our results demonstrate the promise of moving this therapy into clinical practice as most patients indicated willingness to try.
Collapse
Affiliation(s)
- Jens Vent-Schmidt
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Laurie J Goldsmith
- GoldQual Consulting, Richmond Hill, Ontario, Canada,Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Theodore S Steiner
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada,BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada,Address correspondence to: Theodore S. Steiner, Dipl-MolMed, 950 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada ()
| |
Collapse
|
81
|
Olson KE, Namminga KL, Schwab AD, Thurston MJ, Lu Y, Woods A, Lei L, Shen W, Wang F, Joseph SB, Gendelman HE, Mosley RL. Neuroprotective Activities of Long-Acting Granulocyte-Macrophage Colony-Stimulating Factor (mPDM608) in 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine-Intoxicated Mice. Neurotherapeutics 2020; 17:1861-1877. [PMID: 32638217 PMCID: PMC7851309 DOI: 10.1007/s13311-020-00877-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Loss of dopaminergic neurons along the nigrostriatal axis, neuroinflammation, and peripheral immune dysfunction are the pathobiological hallmarks of Parkinson's disease (PD). Granulocyte-macrophage colony-stimulating factor (GM-CSF) has been successfully tested for PD treatment. GM-CSF is a known immune modulator that induces regulatory T cells (Tregs) and serves as a neuronal protectant in a broad range of neurodegenerative diseases. Due to its short half-life, limited biodistribution, and potential adverse effects, alternative long-acting treatment schemes are of immediate need. A long-acting mouse GM-CSF (mPDM608) was developed through Calibr, a Division of Scripps Research. Following mPDM608 treatment, complete hematologic and chemistry profiles and T-cell phenotypes and functions were determined. Neuroprotective and anti-inflammatory capacities of mPDM608 were assessed in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-intoxicated mice that included transcriptomic immune profiles. Treatment with a single dose of mPDM608 resulted in dose-dependent spleen and white blood cell increases with parallel enhancements in Treg numbers and immunosuppressive function. A shift in CD4+ T-cell gene expression towards an anti-inflammatory phenotype corresponded with decreased microgliosis and increased dopaminergic neuronal cell survival. mPDM608 elicited a neuroprotective peripheral immune transformation. The observed phenotypic shift and neuroprotective response was greater than observed with recombinant GM-CSF (rGM-CSF) suggesting human PDM608 as a candidate for PD treatment.
Collapse
Affiliation(s)
- Katherine E. Olson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Krista L. Namminga
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Aaron D. Schwab
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Mackenzie J. Thurston
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Yaman Lu
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Ashley Woods
- Calibr, a Division of Scripps Research, La Jolla, CA 92037 USA
| | - Lei Lei
- Calibr, a Division of Scripps Research, La Jolla, CA 92037 USA
| | - Weijun Shen
- Calibr, a Division of Scripps Research, La Jolla, CA 92037 USA
| | - Feng Wang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China
| | - Sean B. Joseph
- Calibr, a Division of Scripps Research, La Jolla, CA 92037 USA
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198 USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 6898-5880 USA
| | - R. Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198 USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 6898-5880 USA
| |
Collapse
|
82
|
An immune therapy model for effective treatment on inflammatory bowel disease. PLoS One 2020; 15:e0238918. [PMID: 32970698 PMCID: PMC7514012 DOI: 10.1371/journal.pone.0238918] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 08/26/2020] [Indexed: 01/12/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a disease that causes inflammation throughout the digestive tract. Repeated inflammation and frequent relapses cause intestinal damage and expose the patient to a higher risk. In this work, we proposed an immune therapy model for effective treatment strategy through mathematical modeling for patients with IBD. We evaluated the ability of the patient's immune system to recover during treatment. For this, we defined the interval of healthy individual, and examined the frequency of compartments such as T cells and cytokines considered in the model maintain the normal state. Based on the fact that each patient has a unique immune system, we have shown at the same drug works differently, depending on the individual immune system characteristics for every patient. It is known that IBD is related to an imbalance between pro- and anti- inflammatory cytokines as the cause of the disease. So the ratios of pro- to anti- inflammatory cytokines are used as an indicator of patient's condition and inflammation status in various diseases. We compared the ratios of pro- to anti- inflammatory cytokine according to patient's individual immune system and drugs. Since the effects of biological drugs are highly dependent on the patient's own immune system, it is essential to define the immune system status before selecting and using a biological drug.
Collapse
|
83
|
Tryptophan Metabolism, Regulatory T Cells, and Inflammatory Bowel Disease: A Mini Review. Mediators Inflamm 2020; 2020:9706140. [PMID: 32617076 PMCID: PMC7306093 DOI: 10.1155/2020/9706140] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disorder of the gastrointestinal tract resulting from the homeostasis imbalance of intestinal microenvironment, immune dysfunction, environmental and genetic factors, and so on. This disease is associated with multiple immune cells including regulatory T cells (Tregs). Tregs are a subset of T cells regulating the function of various immune cells to induce immune tolerance and maintain intestinal immune homeostasis. Tregs are correlated with the initiation and progression of IBD; therefore, strategies that affect the differentiation and function of Tregs may be promising for the prevention of IBD-associated pathology. It is worth noting that tryptophan (Trp) metabolism is effective in inducing the differentiation of Tregs through microbiota-mediated degradation and kynurenine pathway (KP), which is important for maintaining the function of Tregs. Interestingly, patients with IBD show Trp metabolism disorder in the pathological process, including changes in the concentrations of Trp and its metabolites and alteration in the activities of related catalytic enzymes. Thus, manipulation of Treg differentiation through Trp metabolism may provide a potential target for prevention of IBD. The purpose of this review is to highlight the relationship between Trp metabolism and Treg differentiation and the role of this interaction in the pathogenesis of IBD.
Collapse
|
84
|
Paeoniflorin ameliorates ulcerative colitis by modulating the dendritic cell-mediated T H17/T reg balance. Inflammopharmacology 2020; 28:1705-1716. [PMID: 32472435 PMCID: PMC7572351 DOI: 10.1007/s10787-020-00722-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
Abstract
Immunological tolerance is critical for maintaining gut homeostasis. An imbalance between interleukin-17 (IL-17)-producing T helper 17 (TH17) cells and regulatory T cells (Treg cells) is involved in ulcerative colitis (UC) pathogenesis. Dendritic cells (DCs) are able to induce T cell differentiation. Paeoniflorin (PF) is a monoterpene glucoside that is commonly used for treatment of autoimmune disease. However, the immunological mechanism of PF involvement in UC treatment is unclear. The present study aimed to explore whether PF can restore the TH17/Treg balance by modulating DCs. The effects of PF on DCs, TH17 cells and Treg cells were measured. Furthermore, PF-treated DCs were injected into mice with 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis. PF inhibited MHC-II and CD86 expression on the DC surface (P < 0.05), decreased interleukin (IL)-12 secretion in vitro and in vivo (P < 0.05), and restored the TH17/Treg ratio in the mouse model of colitis (P < 0.05). PF-treated DCs diminished TH17 differentiation (4.26% in vitro and 1.64% in vivo) and decreased IL-17 expression (P < 0.05) while inducing CD4+CD25+Foxp3+ Treg differentiation (7.82% in vitro and 6.85% in vivo) and increasing Foxp3 and IL-10 production (P < 0.05). Additionally, both PF and PF-treated DCs improved colonic histopathology in the mouse model of colitis (P < 0.05). In conclusion this study suggested that PF can ameliorate TNBS-induced colitis by modulating the DC-mediated TH17/Treg balance.
Collapse
|
85
|
Abstract
The disease course of autoimmune diseases such as rheumatoid arthritis is altered during pregnancy, and a similar modulatory role of pregnancy on inflammatory bowel disease (IBD) has been proposed. Hormonal, immunological, and microbial changes occurring during normal pregnancy may interact with the pathophysiology of IBD. IBD consists of Crohn's disease and ulcerative colitis, and because of genetic, immunological, and microbial differences between these disease entities, they may react differently during pregnancy and should be described separately. This review will address the pregnancy-induced physiological changes and their potential effect on the disease course of ulcerative colitis and Crohn's disease, with emphasis on the modulation of epithelial barrier function and immune profiles by pregnancy hormones, microbial changes, and microchimerism.
Collapse
|
86
|
Amoroso C, Perillo F, Strati F, Fantini M, Caprioli F, Facciotti F. The Role of Gut Microbiota Biomodulators on Mucosal Immunity and Intestinal Inflammation. Cells 2020; 9:cells9051234. [PMID: 32429359 PMCID: PMC7291275 DOI: 10.3390/cells9051234] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/12/2022] Open
Abstract
Alterations of the gut microbiota may cause dysregulated mucosal immune responses leading to the onset of inflammatory bowel diseases (IBD) in genetically susceptible hosts. Restoring immune homeostasis through the normalization of the gut microbiota is now considered a valuable therapeutic approach to treat IBD patients. The customization of microbe-targeted therapies, including antibiotics, prebiotics, live biotherapeutics and faecal microbiota transplantation, is therefore considered to support current therapies in IBD management. In this review, we will discuss recent advancements in the understanding of host−microbe interactions in IBD and the basis to promote homeostatic immune responses through microbe-targeted therapies. By considering gut microbiota dysbiosis as a key feature for the establishment of chronic inflammatory events, in the near future it will be suitable to design new cost-effective, physiologic, and patient-oriented therapeutic strategies for the treatment of IBD that can be applied in a personalized manner.
Collapse
Affiliation(s)
- Chiara Amoroso
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (C.A.); (F.P.); (F.S.)
| | - Federica Perillo
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (C.A.); (F.P.); (F.S.)
| | - Francesco Strati
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (C.A.); (F.P.); (F.S.)
| | - Massimo Fantini
- Gastroenterology Unit, Duilio Casula Hospital, AOU Cagliari, 09042 Cagliari, Italy;
- Department of Medical Science and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Flavio Caprioli
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20135 Milan, Italy;
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, 20135 Milan, Italy
| | - Federica Facciotti
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (C.A.); (F.P.); (F.S.)
- Correspondence:
| |
Collapse
|
87
|
Dulic S, Toldi G, Sava F, Kovács L, Molnár T, Milassin Á, Farkas K, Rutka M, Balog A. Specific T-Cell Subsets Can Predict the Efficacy of Anti-TNF Treatment in Inflammatory Bowel Diseases. Arch Immunol Ther Exp (Warsz) 2020; 68:12. [PMID: 32248339 PMCID: PMC7128008 DOI: 10.1007/s00005-020-00575-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 03/03/2020] [Indexed: 12/19/2022]
Abstract
The effect of TNF-blockers on T-lymphocyte subsets is largely unknown in inflammatory bowel diseases (IBDs). The aim of the present study was to analyze the prevalence of T-cell subtypes and their correlation to therapeutic response. Sixty-eight patients with Crohn’s disease (CD), 46 with ulcerative colitis (UC) were enrolled. (1) The clinical course was followed after the initiation of TNF-blockers (prospective study). (2) The immunophenotype was also compared between long-term anti-TNF treated-responders and non-responders (cross-sectional study). The results were compared with those of therapy-naïve patients with active disease and those in remission with non-biological immunosuppressive therapy, and with healthy controls. Fourteen subtypes of peripheral blood T cells were measured with flow cytometry. The prevalence of Th2 and Th17 cells, of HLA-DR- and CD69-positive CD4 and CD8 cells, was higher, whereas the percentage of CD45RA-positive CD4 and CD8 cells was lower in both IBDs than in controls. CD8CD69 cell frequency was lower in remission, and decreased during anti-TNF therapy in CD responders. CD8CD45RO memory cells had higher prevalence in UC non-responders than in those starting anti-TNF. CD4CD45RO percentage < 49.05 at the initiation of TNF-blockers was predictive of a subsequent therapeutic response in CD, and Th2 and Th17 prevalence correlated with the duration of remission on TNF-blockers in UC. This study provided a detailed description of the T-cell composition in IBDs. CD8CD69 prevalence may be an activity marker in CD, and CD4CD45RO, Th2 and Th17 levels could be predictive for a therapeutic response to anti-TNF.
Collapse
Affiliation(s)
- Sonja Dulic
- Department of Rheumatology and Immunology, Faculty of Medicine, Albert Szent-Györgyi Health Center, University of Szeged, Kálvária sgt. 57, Szeged, 6725, Hungary
| | - Gergely Toldi
- First Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
| | - Florentina Sava
- First Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
| | - László Kovács
- Department of Rheumatology and Immunology, Faculty of Medicine, Albert Szent-Györgyi Health Center, University of Szeged, Kálvária sgt. 57, Szeged, 6725, Hungary
| | - Tamás Molnár
- First Department of Medicine, Faculty of Medicine, Albert Szent-Györgyi Health Center, University of Szeged, Szeged, Hungary
| | - Ágnes Milassin
- First Department of Medicine, Faculty of Medicine, Albert Szent-Györgyi Health Center, University of Szeged, Szeged, Hungary
| | - Klaudia Farkas
- First Department of Medicine, Faculty of Medicine, Albert Szent-Györgyi Health Center, University of Szeged, Szeged, Hungary
| | - Mariann Rutka
- First Department of Medicine, Faculty of Medicine, Albert Szent-Györgyi Health Center, University of Szeged, Szeged, Hungary
| | - Attila Balog
- Department of Rheumatology and Immunology, Faculty of Medicine, Albert Szent-Györgyi Health Center, University of Szeged, Kálvária sgt. 57, Szeged, 6725, Hungary.
| |
Collapse
|
88
|
Jia Y, Anwaar S, Li L, Yin Z, Ye Z, Huang Z. A new target for the treatment of inflammatory bowel disease: Interleukin-37. Int Immunopharmacol 2020; 83:106391. [PMID: 32208166 DOI: 10.1016/j.intimp.2020.106391] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/22/2020] [Accepted: 03/08/2020] [Indexed: 12/19/2022]
Abstract
Interleukin (IL)-37 belongs to the IL-1 cytokine family. It has anti-inflammatory effects on numerous autoimmune diseases such as asthma, psoriasis, inflammatory bowel disease (IBD), systemic lupus erythematosus (SLE), multiple sclerosis (MS) and rheumatoid arthritis (RA). Mechanistically, IL-37 plays an anti-inflammatory role by regulating the expression of inflammatory factors in two ways: binding extracellular receptors IL-18R or transferring into the nucleus with Smad3. IBD is a kind of idiopathic intestinal inflammatory disease with unknown etiology and pathogenesis. Recent researches had proved that IL-37 is negatively involved in the pathogenesis and development of IBD. Among various inflammatory diseases, IL-37 has been shown to regulate inflammatory development by acting on various immune cells such as neutrophils, macrophages (Mϕ), dendritic cells (DCs), T cells and intestinal epithelial cells. This review summarizes the biological role of IL-37, and its immunoregulatory effects on the immune cells, especially anti-inflammatory function in both human and experimental models of IBD.
Collapse
Affiliation(s)
- Yuning Jia
- Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Health Science Center, Shenzhen University, Shenzhen 518055, China
| | - Shoaib Anwaar
- Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Health Science Center, Shenzhen University, Shenzhen 518055, China
| | - Linyun Li
- Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Health Science Center, Shenzhen University, Shenzhen 518055, China
| | - Zhihua Yin
- Shenzhen City Futian Qu Rheumatology Specialist Hospital, Shenzhen 518089, China
| | - Zhizhon Ye
- Shenzhen City Futian Qu Rheumatology Specialist Hospital, Shenzhen 518089, China.
| | - Zhong Huang
- Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Health Science Center, Shenzhen University, Shenzhen 518055, China.
| |
Collapse
|
89
|
Meyer F, Seibert FS, Nienen M, Welzel M, Beisser D, Bauer F, Rohn B, Westhoff TH, Stervbo U, Babel N. Propionate supplementation promotes the expansion of peripheral regulatory T-Cells in patients with end-stage renal disease. J Nephrol 2020; 33:817-827. [PMID: 32144645 PMCID: PMC7381474 DOI: 10.1007/s40620-019-00694-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/26/2019] [Indexed: 12/14/2022]
Abstract
Patients with end-stage renal disease (ESRD) suffer from a progressively increasing low-grade systemic inflammation, which is associated with higher morbidity and mortality. Regulatory T cells (Tregs) play an important role in regulation of the inflammatory process. Previously, it has been demonstrated that short-chain fatty acids reduce inflammation in the central nervous system in a murine model of multiple sclerosis through an increase in tissue infiltrating Tregs. Here, we evaluated the effect of the short-chain fatty acid propionate on the chronic inflammatory state and T-cell composition in ESRD patients. Analyzing ESRD patients and healthy blood donors before, during, and 60 days after the propionate supplementation by multiparametric flow cytometry we observed a gradual and significant expansion in the frequencies of CD25highCD127- Tregs in both groups. Phenotypic characterization suggests that polarization of naïve T cells towards Tregs is responsible for the observed expansion. In line with this, we observed a significant reduction of inflammatory marker CRP under propionate supplementation. Of interest, the observed anti-inflammatory surroundings did not affect the protective pathogen-specific immunity as demonstrated by the stable frequencies of effector/memory T cells specific for tetanus/diphtheria recall antigens. Collectively, our data suggest that dietary supplements with propionate have a beneficial effect on the elevated systemic inflammation of ESRD patients. The effect can be achieved through an expansion of circulating Tregs without affecting the protective pathogen-reactive immunity.
Collapse
Affiliation(s)
- Fabian Meyer
- Medical Department I , Centre for Translational Medicine, Marienhospital Herne, Universitätsklinikum Der Ruhr-Universität Bochum, Ruhr-Universität Bochum, Hölkeskampring 40, 44623, Herne, Germany
| | - Felix S Seibert
- Medical Department I , Centre for Translational Medicine, Marienhospital Herne, Universitätsklinikum Der Ruhr-Universität Bochum, Ruhr-Universität Bochum, Hölkeskampring 40, 44623, Herne, Germany
| | - Mikalai Nienen
- Medical Department I , Centre for Translational Medicine, Marienhospital Herne, Universitätsklinikum Der Ruhr-Universität Bochum, Ruhr-Universität Bochum, Hölkeskampring 40, 44623, Herne, Germany
| | - Marius Welzel
- Biodiversity, University of Duisburg-Essen, Essen, Germany
| | | | - Frederic Bauer
- Medical Department I , Centre for Translational Medicine, Marienhospital Herne, Universitätsklinikum Der Ruhr-Universität Bochum, Ruhr-Universität Bochum, Hölkeskampring 40, 44623, Herne, Germany
| | - Benjamin Rohn
- Medical Department I , Centre for Translational Medicine, Marienhospital Herne, Universitätsklinikum Der Ruhr-Universität Bochum, Ruhr-Universität Bochum, Hölkeskampring 40, 44623, Herne, Germany
| | - Timm H Westhoff
- Medical Department I , Centre for Translational Medicine, Marienhospital Herne, Universitätsklinikum Der Ruhr-Universität Bochum, Ruhr-Universität Bochum, Hölkeskampring 40, 44623, Herne, Germany
| | - Ulrik Stervbo
- Medical Department I , Centre for Translational Medicine, Marienhospital Herne, Universitätsklinikum Der Ruhr-Universität Bochum, Ruhr-Universität Bochum, Hölkeskampring 40, 44623, Herne, Germany.
| | - Nina Babel
- Medical Department I , Centre for Translational Medicine, Marienhospital Herne, Universitätsklinikum Der Ruhr-Universität Bochum, Ruhr-Universität Bochum, Hölkeskampring 40, 44623, Herne, Germany.
- Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany.
| |
Collapse
|
90
|
Immunohistochemical staining with chemokine panel of non-specific colitis predicts future IBD diagnosis. Cytokine 2020; 127:154935. [PMID: 31770615 DOI: 10.1016/j.cyto.2019.154935] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/15/2019] [Accepted: 11/18/2019] [Indexed: 12/16/2022]
|
91
|
Younis N, Zarif R, Mahfouz R. Inflammatory bowel disease: between genetics and microbiota. Mol Biol Rep 2020; 47:3053-3063. [PMID: 32086718 DOI: 10.1007/s11033-020-05318-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/11/2020] [Indexed: 02/07/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic relapsing inflammatory disease that can involve any part of the gastrointestinal tract. It includes two main disorders: Crohn's disease (CD) and Ulcerative colitis (UC). CD and UC often share a similar clinical presentation; however, they affect distinct parts of the GI Tract with a different gut wall inflammatory extent. Ultimately, IBD seems to emanate from an uncontrollably continuous inflammatory process arising against the intestinal microbiome in a genetically susceptible individual. It is a multifactorial disease stemming from the impact of both environmental and genetic components on the intestinal microbiome. Furthermore, IBD genetics has gained a lot of attention. Around 200 loci were identified as imparting an increased risk for IBD. Few of them were heavily investigated and determined as highly linked to IBD. These genes, as discussed below, include NOD2, ATG16L1, IRGM, LRRK2, PTPN2, IL23R, Il10, Il10RA, Il10RB, CDH1 and HNF4α among others. Consequently, the incorporation of a genetic panel covering these key genes would markedly enhance the diagnosis and evaluation of IBD.
Collapse
Affiliation(s)
- Nour Younis
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Cairo Street, Beirut, Lebanon
| | - Rana Zarif
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Cairo Street, Beirut, Lebanon
| | - Rami Mahfouz
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Cairo Street, Beirut, Lebanon.
| |
Collapse
|
92
|
TSG-6 in extracellular vesicles from canine mesenchymal stem/stromal is a major factor in relieving DSS-induced colitis. PLoS One 2020; 15:e0220756. [PMID: 32040478 PMCID: PMC7010233 DOI: 10.1371/journal.pone.0220756] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue derived mesenchymal stem/stromal cell (ASC)-derived extracellular vesicles (EV) have been reported to be beneficial against dextran sulfate sodium (DSS)-induced colitis in mice. However, the underlying mechanisms have not been fully elucidated. We hypothesize that the tumor necrosis factor-α-stimulated gene/protein 6 (TSG-6) in EVs is a key factor influencing the alleviation of colitis symptoms. DSS-induced colitis mice (C57BL/6, male, Naïve = 6, Sham = 8, PBS = 8 EV = 8, CTL-EV = 8, TSG-6 depleted EV = 8) were intraperitoneally administered EVs (100 ug/mice) on day 1, 3, and 5; colon tissues were collected on day 10 for histopathological, RT-qPCR, western blot and immunofluorescence analyses. In mice injected with EV, inflammation was alleviated. Indeed, EVs regulated the levels of pro- and anti-inflammatory cytokines, such as TNF-α, IL-1β, IFN-γ, IL-6, and IL-10 in inflamed colons. However, when injected with TSG-6 depleted EV, the degree of inflammatory relief was reduced. Furthermore, TSG-6 in EVs plays a key role in increasing regulatory T cells (Tregs) and polarizing macrophage from M1 to M2 in the colon. In conclusion, this study shows that TSG-6 in EVs is a major factor in the relief of DSS-induced colitis, by increasing the number of Tregs and macrophage polarization from M1 to M2 in the colon.
Collapse
|
93
|
TNF-α and INF-γ primed canine stem cell-derived extracellular vesicles alleviate experimental murine colitis. Sci Rep 2020; 10:2115. [PMID: 32034203 PMCID: PMC7005871 DOI: 10.1038/s41598-020-58909-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 01/13/2020] [Indexed: 12/11/2022] Open
Abstract
The inflammatory bowel diseases (IBD) are characterized by relapsing inflammation and immune activation diseases of the gastrointestinal tract. Extracellular vesicles, which elicit similar biological activity to the stem cell themselves, have been used experimentally to treat dextran sulfate sodium (DSS)-induced colitis in murine models though immunosuppressive potential. In this study, we investigated whether the Extracellular vesicles (EVs) obtained by stimulating inflammatory cytokine on canine adipose mesenchymal stem cells (cASC) improved anti-inflammatory and/or immunosuppressive potential of EVs, and/or their ability to alleviate inflammation in colitis. We also explored the correlation between immune cells and the inflammatory repressive effect of primed EVs. Pro-inflammatory cytokines such as TNF-α and IFN-γ increased immunosuppressive protein such as HGF, TSG-6, PGE2 and TGF-β in EVs. Moreover, the anti-inflammatory effect of EVs was improved through pretreatment with inflammatory cytokines. Importantly, EVs obtained from primed stem cells effectively induced macrophage polarization toward an anti-inflammatory M2 phenotype and suppressed activated immunity by enhancing regulatory T cells in inflamed colon in mice. Our results provide a new and effective therapy for the EVs obtained from ASC stimulated with TNF-α and IFN-γ against not only IBD, but also immune-mediated disease.
Collapse
|
94
|
Jamwal DR, Marati RV, Harrison CA, Midura-Kiela MT, Figliuolo Paz VR, Besselsen DG, Ghishan FK, Kiela PR. Total CD3 T Cells Are Necessary and Sufficient to Induce Colitis in Immunodeficient Mice With Dendritic Cell-Specific Deletion of TGFbR2: A Novel IBD Model to Study CD4 and CD8 T-Cell Interaction. Inflamm Bowel Dis 2020; 26:229-241. [PMID: 31559420 PMCID: PMC7185689 DOI: 10.1093/ibd/izz191] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/06/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a multifactorial disorder, with the innate and adaptive immune cells contributing to disease initiation and progression. However, the intricate cross-talk between immune cell lineages remains incompletely understood. The role of CD8+ T cells in IBD pathogenesis has been understudied, largely due to the lack of appropriate models. METHODS We previously reported spontaneous colitis in mice with impaired TGFβ signaling due to dendritic cell-specific knockout of TGFbR2 (TGFβR2ΔDC). Here, we demonstrate that crossing TGFβR2ΔDC mice with a Rag1-/- background eliminates all symptoms of colitis and that adoptive transfer of unfractionated CD3+ splenocytes is sufficient to induce progressive colitis in Rag1-/-TGFβR2ΔDC mice. RESULTS Both CD4+ and CD8+ T cells are required for the induction of colitis accompanied by activation of both T-cell lineages and DCs, increased expression of mucosal IFNγ, TNFα, IL6, IL1β, and IL12, and decreased frequencies of CD4+FoxP3+ regulatory T cells. Development of colitis required CD40L expression in CD4+ T cells, and the disease was partially ameliorated by IFNγ neutralization. CONCLUSIONS This novel model provides an important tool for studying IBD pathogenesis, in particular the complex interactions among innate and adaptive immune cells in a controlled fashion, and represents a valuable tool for preclinical evaluation of novel therapeutics.
Collapse
Affiliation(s)
| | - Raji V Marati
- Department of Pediatrics, Tucson, Arizona, USA,Present affiliation: Roche Tissue Diagnostics, Tucson, Arizona, USA
| | - Christy A Harrison
- Department of Pediatrics, Tucson, Arizona, USA,Present affiliation: New York City Department of Health & Mental Hygiene, Long Island City, New York, USA
| | | | | | | | | | - Pawel R Kiela
- Department of Pediatrics, Tucson, Arizona, USA,Department of Immunobiology, University of Arizona, Tucson, Arizona, USA,Address correspondence to: Pawel R. Kiela, DVM, PhD, University of Arizona, Steele Children’s Research Center, 1501 N. Campbell Ave, Rm. 6351, Tucson, AZ 85724 ()
| |
Collapse
|
95
|
Sabel R, Fronza AS, Carrenho LZB, Maes A, Barros ML, Pollo LAE, Biavatti MW, D'Herde K, Vandenabeele P, Kreuger MRO. Anti-inflammatory activity of the sesquiterpene lactone diacethylpiptocarphol in dextransulfate sodium-induced colitis in mice. JOURNAL OF ETHNOPHARMACOLOGY 2019; 245:112186. [PMID: 31472273 DOI: 10.1016/j.jep.2019.112186] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 08/22/2019] [Accepted: 08/23/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sesquiterpene lactones are organic compounds derived mainly from plants that exhibit anti-inflammatory and antitumor activities being one of the key mechanism of action of NF-kB pathway and synthesis of cytokines such as IL-1 and TNF- α. AIM OF THE STUDY The overall objective of the present study was to evaluate the anti-inflammatory action of a sesquiterpene lactone diacethylpiptocarphol (DPC) from Vernonia scorpioides (Lam.) Pers. and parthenolide (PTH) in Balb-c mice with DSS-induced colitis. MATERIALS AND METHODS The anti-inflammatory effects of Intraperitonial administration of DPC (5 mg/kg/day) were evaluated in Balb/c mice with DSS-induced colitis, and further the body weight measurement, TNF-α and TGF-β level was determined. RESULTS After intraperitoneal treatment for one week, DSS-induced colitis was significantly reduced in mice treated with either of both sesquiterpenes lactones, as witnessed by reduced cellular infiltration, tissue damage, TNF-α production, and enhanced production of TGF-β. CONCLUSIONS Sesquiterpene lactone DPC, isolated from Vernonia scorpioides showed anti-inflammatory activity, in this experimental model of colitis the sesquiterpene lactones DPC and PTH exhibit equal anti-inflammatory activity.
Collapse
Affiliation(s)
- R Sabel
- Universidade do Vale do Itajaí, Brazil
| | | | | | - A Maes
- Universidade do Vale do Itajaí, Brazil
| | | | - L A E Pollo
- Universidade Federal de Santa Catarin, Brazil
| | | | - K D'Herde
- Anatomy and Embryology Group, Ghent University, Ghent, Belgium
| | - P Vandenabeele
- Inflammation Research Center (IRC), VIB, Ghent, Belgium; Department Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Methusalem Programm, Ghent University, Ghent, Belgium
| | - M R O Kreuger
- Universidade do Vale do Itajaí, Brazil; Centro Universitário Avantis, Brazil
| |
Collapse
|
96
|
Wen J, Khan I, Li A, Chen X, Yang P, Song P, Jing Y, Wei J, Che T, Zhang C. Alpha-linolenic acid given as an anti-inflammatory agent in a mouse model of colonic inflammation. Food Sci Nutr 2019; 7:3873-3882. [PMID: 31890165 PMCID: PMC6924294 DOI: 10.1002/fsn3.1225] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/08/2019] [Accepted: 04/13/2019] [Indexed: 01/12/2023] Open
Abstract
This study examined the relationship between the high-fat, high-sugar diet (HFHSD) and trinitrobenzene sulfonic acid (TNBS) induced mouse colitis, the therapeutic effect of alpha-linolenic acid (ALA) on mouse colitis, and the relationship between HFHSD and hyperlipidemia. We also examined the possible underlying mechanisms behind their interactions. Female BABL/c mice were fed with HFHSD for the 9 weeks. At the same time, ALA treatment (150 or 300 mg/kg) was administered on a daily basis. At the end of the 9 weeks, experimental colitis was induced by the intra-colonic administration of TNBS. Body weight, spleen weight, disease activity index (DAI), histological changes, T-cell-related cytokine level, and lipid profiles were measured after treatment. TNBS induced severe clinical manifestations of colitis and histological damage. Low-ALA (150 mg/kg) administration profoundly ameliorated TNBS-induced clinical manifestations, body weight loss, spleen weight loss, and histological damage. On the contrary, the high-ALA (300 mg/kg) administration did not ameliorate colitis and even exacerbated the symptoms. HFHSD consumption assisted TNBS in changing IL-12, IFN-γ, IL-2, and IL-17A in the liver. As expected, these changes were recovered through low-ALA. In addition, HFHSD had a significant impact on the total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), and triglyceride (TG), which related to the increased risk of hyperlipidemia. In summation, HFHSD exacerbated the TNBS-induced colitis via the Th1/Th17 pathway. The Low-ALA (150 mg/kg) exhibited protective effects against the TNBS-induced colitis via the Th1/Th2/Th17 pathway.
Collapse
Affiliation(s)
- Juan Wen
- School of Life SciencesLanzhou UniversityLanzhouChina
- Key Laboratory of Cell Activities and Stress AdaptationsMinistry of EducationLanzhou UniversityLanzhouChina
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental PollutionLanzhou UniversityLanzhouChina
| | - Israr Khan
- School of Life SciencesLanzhou UniversityLanzhouChina
- Key Laboratory of Cell Activities and Stress AdaptationsMinistry of EducationLanzhou UniversityLanzhouChina
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental PollutionLanzhou UniversityLanzhouChina
| | - Anping Li
- School of Life SciencesLanzhou UniversityLanzhouChina
| | - Xinjun Chen
- Laboratory of Pathogenic Biology and ImmunologyHainan Medical UniversityHaikouChina
| | - Pingrong Yang
- School of Life SciencesLanzhou UniversityLanzhouChina
- Gansu Institute of Drug ControlLanzhouChina
| | - Pingshun Song
- School of Life SciencesLanzhou UniversityLanzhouChina
- Gansu Institute of Drug ControlLanzhouChina
| | - Yaping Jing
- School of Life SciencesLanzhou UniversityLanzhouChina
- Key Laboratory of Cell Activities and Stress AdaptationsMinistry of EducationLanzhou UniversityLanzhouChina
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental PollutionLanzhou UniversityLanzhouChina
| | - Junshu Wei
- School of Life SciencesLanzhou UniversityLanzhouChina
- Key Laboratory of Cell Activities and Stress AdaptationsMinistry of EducationLanzhou UniversityLanzhouChina
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental PollutionLanzhou UniversityLanzhouChina
| | - Tuanjie Che
- Gansu Key Laboratory of Functional Genomics and Molecular DiagnosisLanzhouChina
| | - Chunjiang Zhang
- School of Life SciencesLanzhou UniversityLanzhouChina
- Key Laboratory of Cell Activities and Stress AdaptationsMinistry of EducationLanzhou UniversityLanzhouChina
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental PollutionLanzhou UniversityLanzhouChina
| |
Collapse
|
97
|
Zachariassen LF, Hansen AK, Krych L, Nielsen DS, Holm TL, Tougaard P, Hansen CHF. Cesarean section increases sensitivity to oxazolone-induced colitis in C57BL/6 mice. Mucosal Immunol 2019; 12:1348-1357. [PMID: 31554900 DOI: 10.1038/s41385-019-0207-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 08/30/2019] [Accepted: 09/07/2019] [Indexed: 02/07/2023]
Abstract
Children born by cesarean section (CS) have an increased risk of developing inflammatory bowel disease (IBD), possibly due to skewed microbial colonization during birth and consequently impaired bacterial stimulation of the developing immune system. The aim of this study was to investigate the association between CS and experimental colitis in a murine model of IBD. It was hypothesized that CS aggravates colonic inflammation due to a change in gut microbiota (GM) composition. C57BL/6 mice, delivered by CS or vaginal delivery (VD), were intra-rectally challenged with oxazolone at 8 weeks of age and monitored for colitis symptoms. The results showed that CS delivered mice experienced an increased body weight loss and colon weight, together with higher colonic concentrations of TNF-α and MPO compared with VD mice. Increased infiltration of inflammatory cells was present in CS delivered mice, as well as a downregulation in expression of the gut integrity genes occludin and tight junction protein 1 indicative of an impaired barrier function. The GM from CS delivered mice without colitis partly contributed to the increase in colitis symptoms when inoculated into germ-free recipient mice. In conclusion, CS increased sensitivity to oxazolone induced colitis in mice.
Collapse
Affiliation(s)
- Line Fisker Zachariassen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Axel Kornerup Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lukasz Krych
- Department of Food Science, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Dennis Sandris Nielsen
- Department of Food Science, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | | | - Peter Tougaard
- Molecular Signaling and Cell Death Unit, VIB-Ugent Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium.,Department for Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Camilla Hartmann Friis Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
98
|
Bertani L, Antonioli L, Fornai M, Tapete G, Baiano Svizzero G, Marchi S, Blandizzi C, Costa F. Evaluation of cytokine levels as putative biomarkers to predict the pharmacological response to biologic therapy in inflammatory bowel diseases. MINERVA GASTROENTERO 2019; 65:298-308. [PMID: 31646851 DOI: 10.23736/s1121-421x.19.02621-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cytokines play a central role in the pathogenesis of inflammatory bowel diseases. For this reason, the vast majority of biological therapies are aimed to block pro-inflammatory cytokines or their receptors. Although these drugs have modified the course of the disease due to their efficacy, a high rate of non-response or loss of response over time is still an important issue for clinicians. In this perspective, many studies have been conducted in recent years to individuate a reliable biomarker of therapeutic response. In this review, we discuss the role of cytokines involved in the pathogenesis and in the therapy of inflammatory bowel diseases, and their putative use as pharmacological biomarkers of therapy responsiveness.
Collapse
Affiliation(s)
- Lorenzo Bertani
- Unit of Gastroenterology, Department of Translational Research and New Technologies in Medicine and Surgery, Pisa University Hospital, Pisa, Italy -
| | - Luca Antonioli
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Matteo Fornai
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gherardo Tapete
- Unit of Gastroenterology, Department of Translational Research and New Technologies in Medicine and Surgery, Pisa University Hospital, Pisa, Italy
| | - Giovanni Baiano Svizzero
- Unit of Gastroenterology, Department of Translational Research and New Technologies in Medicine and Surgery, Pisa University Hospital, Pisa, Italy
| | - Santino Marchi
- Unit of Gastroenterology, Department of Translational Research and New Technologies in Medicine and Surgery, Pisa University Hospital, Pisa, Italy
| | - Corrado Blandizzi
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesco Costa
- Unit of Inflammatory Bowel Diseases, Department of General Surgery and Gastroenterology, Pisa University Hospital, Pisa, Italy
| |
Collapse
|
99
|
Shakurnia A, Sheikhi A, Mirzapour M, Baharifar V, Baharifar N, Aghamohammadi N, Sheikhi M, Matinrad M, Mousavinasab SN, Sheikhi S, Sheikhi R. Sugarcane molasses enhances TGF-β secretion and FOXP3 gene expression by Bifidobacterium Animalis Subsp. Lactis stimulated PBMCs of Ulcerative Colitis patients. Complement Ther Med 2019; 47:102210. [PMID: 31780030 DOI: 10.1016/j.ctim.2019.102210] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 10/03/2019] [Accepted: 10/04/2019] [Indexed: 02/07/2023] Open
Abstract
Ulcerative colitis (UC) is one of the inflammatory diseases of the gut with frequent bloody diarrhea leads to increased rates of anemia. Evidences indicate the immunomodulation disorders in the response to intestinal microbiota in UC. Although sugarcane molasses, rich in necessary minerals and vitamins, could be a good support nutrient but its effect on immune system of UC patients is unknown. To determine how the immune system of UC patients responds to molasses this study was planned. Bifidobacterium lactis were cultivated on MRS broth. PBMCs of 12 UC patients were separated by Ficoll-Hypaque centrifugation and co-cultured with different concentrations of UV killed bacteria and/or molasses in RPMI-1640 plus 10 % FCS. The gene expression of FoxP3 was measured by real-time PCR. TGF-β and TNF-α were measured in supernatant of PBMCs by ELISA. Sugarcane molasses and B. lactis significantly augmented TGF-β compared to control (p < 0.01 and p < 0.001 respectively). The secretion levels of TGF-β by B. lactis plus molasses compared to B. lactis stimulated PBMCs was significantly higher (p < 0.05) but the level of TNF-α by PBMCs after 2/4/12 h incubation with B. lactis plus molasses compared to B. lactis alone was not changed (p > 0.2). The level of FOXP3 expression after treatment with molasses was increased significantly (p < 0.05). These data show that if sugarcane molasses added to B. lactis, not only do not increase the pro-inflammatory cytokine, TNF-α, but also augments the anti-inflammatory cytokine, TGF-β by PBMCs. Therefore, these results pave the way for further investigation to show sugarcane molasses as a safe support to compensate the lost nutrients in UC patients.
Collapse
Affiliation(s)
- Abdolhussein Shakurnia
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Abdolkarim Sheikhi
- Department of Immunology, Faculty of Medicine, Dezful University of Medical Sciences, Dezful, Iran.
| | | | - Vahid Baharifar
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Narges Baharifar
- Department of Immunology, Faculty of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Nima Aghamohammadi
- Department of Internal Medicine, Faculty of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Mehdi Sheikhi
- Faculty of Medicine, Kazeroon Azad University, Kazeroon, Iran
| | | | - S Nouraddin Mousavinasab
- Department of Social Medicine, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sepideh Sheikhi
- Faculty of Nursing and Midwifery, Islamic Azad University of Dezful, Dezful, Iran
| | - Razieh Sheikhi
- Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
100
|
Tacconi C, Schwager S, Cousin N, Bajic D, Sesartic M, Sundberg JP, Neri D, Detmar M. Antibody-Mediated Delivery of VEGFC Ameliorates Experimental Chronic Colitis. ACS Pharmacol Transl Sci 2019; 2:342-352. [PMID: 32259068 DOI: 10.1021/acsptsci.9b00037] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Indexed: 12/13/2022]
Abstract
Crohn's disease (CD) and ulcerative colitis (UC) are two distinct forms of inflammatory bowel disease (IBD) characterized by an expanded lymphatic network with impaired functionality both in mouse models and in human patients. In this study, we investigated whether targeted delivery of the pro-lymphangiogenic vascular endothelial growth factor C (VEGFC) to the site of inflammation may represent a new, clinically feasible strategy for treating IBD. To achieve targeting of inflamed tissue, we developed a fusion protein consisting of human VEGFC fused to the F8 antibody (F8-VEGFC), which specifically binds to the extradomain A (EDA) of fibronectin, a spliced isoform almost exclusively expressed in inflamed tissues. The therapeutic activity of intravenously administered F8-VEGFC, compared to a targeted construct lacking VEGFC (F8-SIP), was investigated in a mouse model of dextran sodium sulfate (DSS)-induced colitis. The presence of EDA fibronectin was detected in both human and mouse inflamed colon tissue. Biodistribution studies of radiolabeled F8-VEGFC revealed a specific accumulation of the antibody in the colon of DSS-administered mice, as compared to an untargeted VEGFC fusion protein (KSF-VEGFC) (binding the irrelevant hen egg lysozyme antigen). Systemic treatment with F8-VEGFC significantly reduced the clinical and histological signs of inflammation, expanded the lymphatic vascular network, reduced the density of immune cells, and also decreased the expression of inflammatory cytokines in the inflamed colon. Overall, these results reveal that administration of F8-VEGFC represents a novel and promising approach for the treatment of IBD.
Collapse
Affiliation(s)
- Carlotta Tacconi
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, 8093 Zurich, Switzerland
| | - Simon Schwager
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, 8093 Zurich, Switzerland
| | - Nikola Cousin
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, 8093 Zurich, Switzerland
| | - Davor Bajic
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, 8093 Zurich, Switzerland
| | - Marko Sesartic
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, 8093 Zurich, Switzerland
| | - John P Sundberg
- The Jackson Laboratory, Bar Harbor, Maine 04609, United States
| | - Dario Neri
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, 8093 Zurich, Switzerland
| | - Michael Detmar
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|