351
|
Johnson JM, Verkerke ARP, Maschek JA, Ferrara PJ, Lin CT, Kew KA, Neufer PD, Lodhi IJ, Cox JE, Funai K. Alternative splicing of UCP1 by non-cell-autonomous action of PEMT. Mol Metab 2020; 31:55-66. [PMID: 31918922 PMCID: PMC6889607 DOI: 10.1016/j.molmet.2019.10.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/14/2019] [Accepted: 10/30/2019] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Phosphatidylethanolamine methyltransferase (PEMT) generates phosphatidylcholine (PC), the most abundant phospholipid in the mitochondria and an important acyl chain donor for cardiolipin (CL) biosynthesis. Mice lacking PEMT (PEMTKO) are cold-intolerant when fed a high-fat diet (HFD) due to unclear mechanisms. The purpose of this study was to determine whether PEMT-derived phospholipids are important for the function of uncoupling protein 1 (UCP1) and thus for maintenance of core temperature. METHODS To test whether PEMT-derived phospholipids are important for UCP1 function, we examined cold-tolerance and brown adipose (BAT) mitochondria from PEMTKO mice with or without HFD feeding. We complemented these studies with experiments on mice lacking functional CL due to tafazzin knockdown (TAZKD). We generated several conditional mouse models to study the tissue-specific roles of PEMT, including mice with BAT-specific knockout of PEMT (PEMT-BKO). RESULTS Chow- and HFD-fed PEMTKO mice completely lacked UCP1 protein in BAT, despite a lack of difference in mRNA levels, and the mice were accordingly cold-intolerant. While HFD-fed PEMTKO mice exhibited reduced mitochondrial CL content, this was not observed in chow-fed PEMTKO mice or TAZKD mice, indicating that the lack of UCP1 was not attributable to CL deficiency. Surprisingly, the PEMT-BKO mice exhibited normal UCP1 protein levels. Knockout of PEMT in the adipose tissue (PEMT-AKO), liver (PEMT-LKO), or skeletal muscle (PEMT-MKO) also did not affect UCP1 protein levels, suggesting that lack of PEMT in other non-UCP1-expressing cells communicates to BAT to suppress UCP1. Instead, we identified an untranslated UCP1 splice variant that was triggered during the perinatal period in the PEMTKO mice. CONCLUSIONS PEMT is required for UCP1 splicing that yields functional protein. This effect is derived by PEMT in nonadipocytes that communicates to BAT during embryonic development. Future research will focus on identifying the non-cell-autonomous PEMT-dependent mechanism of UCP1 splicing.
Collapse
Affiliation(s)
- Jordan M Johnson
- Diabetes & Metabolism Research Center, University of Utah, 15 N. 2030 E, Salt Lake City, UT, 84112, USA; Department of Nutrition & Integrative Physiology, University of Utah, 250 S. 1850 E., RM 214, Salt Lake City, UT, 84112, USA; Department of Physical Therapy & Athletic Training, University of Utah, 520 Wakara Way, Salt Lake City, UT, 84108, USA; East Carolina Diabetes & Obesity Institute, East Carolina University, 115 Heart Drive, 4101 ECHI, Greenville, NC, 27834, USA
| | - Anthony R P Verkerke
- Diabetes & Metabolism Research Center, University of Utah, 15 N. 2030 E, Salt Lake City, UT, 84112, USA; Department of Nutrition & Integrative Physiology, University of Utah, 250 S. 1850 E., RM 214, Salt Lake City, UT, 84112, USA; Department of Physical Therapy & Athletic Training, University of Utah, 520 Wakara Way, Salt Lake City, UT, 84108, USA; East Carolina Diabetes & Obesity Institute, East Carolina University, 115 Heart Drive, 4101 ECHI, Greenville, NC, 27834, USA
| | - J Alan Maschek
- Diabetes & Metabolism Research Center, University of Utah, 15 N. 2030 E, Salt Lake City, UT, 84112, USA; Metabolomics Core Research Facility, University of Utah, 15 N. Medical Dr. East RM A306, Salt Lake City, UT, 84112, USA; Department of Biochemistry, University of Utah, 15 N. Medical Dr. East RM 4100, Salt Lake City, UT, 84112, USA
| | - Patrick J Ferrara
- Diabetes & Metabolism Research Center, University of Utah, 15 N. 2030 E, Salt Lake City, UT, 84112, USA; Department of Nutrition & Integrative Physiology, University of Utah, 250 S. 1850 E., RM 214, Salt Lake City, UT, 84112, USA; Department of Physical Therapy & Athletic Training, University of Utah, 520 Wakara Way, Salt Lake City, UT, 84108, USA; East Carolina Diabetes & Obesity Institute, East Carolina University, 115 Heart Drive, 4101 ECHI, Greenville, NC, 27834, USA
| | - Chien-Te Lin
- East Carolina Diabetes & Obesity Institute, East Carolina University, 115 Heart Drive, 4101 ECHI, Greenville, NC, 27834, USA
| | - Kimberly A Kew
- East Carolina Diabetes & Obesity Institute, East Carolina University, 115 Heart Drive, 4101 ECHI, Greenville, NC, 27834, USA; Department of Chemistry, East Carolina University, Greenville, NC, 27858, USA
| | - P Darrell Neufer
- East Carolina Diabetes & Obesity Institute, East Carolina University, 115 Heart Drive, 4101 ECHI, Greenville, NC, 27834, USA
| | - Irfan J Lodhi
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO, 63110, USA
| | - James E Cox
- Diabetes & Metabolism Research Center, University of Utah, 15 N. 2030 E, Salt Lake City, UT, 84112, USA; Metabolomics Core Research Facility, University of Utah, 15 N. Medical Dr. East RM A306, Salt Lake City, UT, 84112, USA; Department of Biochemistry, University of Utah, 15 N. Medical Dr. East RM 4100, Salt Lake City, UT, 84112, USA
| | - Katsuhiko Funai
- Diabetes & Metabolism Research Center, University of Utah, 15 N. 2030 E, Salt Lake City, UT, 84112, USA; Department of Nutrition & Integrative Physiology, University of Utah, 250 S. 1850 E., RM 214, Salt Lake City, UT, 84112, USA; Department of Physical Therapy & Athletic Training, University of Utah, 520 Wakara Way, Salt Lake City, UT, 84108, USA; East Carolina Diabetes & Obesity Institute, East Carolina University, 115 Heart Drive, 4101 ECHI, Greenville, NC, 27834, USA; Molecular Medicine Program, University of Utah, 15 N. 2030 E. RM 4145, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
352
|
Shi H, Chi H. Metabolic Control of Treg Cell Stability, Plasticity, and Tissue-Specific Heterogeneity. Front Immunol 2019; 10:2716. [PMID: 31921097 PMCID: PMC6917616 DOI: 10.3389/fimmu.2019.02716] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 11/05/2019] [Indexed: 12/21/2022] Open
Abstract
Regulatory T (Treg) cells are crucial for peripheral immune tolerance and prevention of autoimmunity and tissue damage. Treg cells are inherently defined by the expression of the transcription factor Foxp3, which enforces lineage development and immune suppressive function of these cells. Under various conditions as observed in autoimmunity, cancer and non-lymphoid tissues, a proportion of Treg cells respond to specific environmental signals and display altered stability, plasticity and tissue-specific heterogeneity, which further shape their context-dependent suppressive functions. Recent studies have revealed that metabolic programs play pivotal roles in controlling these processes in Treg cells, thereby considerably expanding our understanding of Treg cell biology. Here we summarize these recent advances that highlight how cell-extrinsic factors, such as nutrients, vitamins and metabolites, and cell-intrinsic metabolic programs, orchestrate Treg cell stability, plasticity, and tissue-specific heterogeneity. Understanding metabolic regulation of Treg cells should provide new insight into immune homeostasis and disease, with important therapeutic implications for autoimmunity, cancer, and other immune-mediated disorders.
Collapse
Affiliation(s)
- Hao Shi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| |
Collapse
|
353
|
Qi L, Zhang Q, Miao Y, Kang W, Tian Z, Xu D, Xiao W, Fang F. Interleukin-33 activates and recruits natural killer cells to inhibit pulmonary metastatic cancer development. Int J Cancer 2019; 146:1421-1434. [PMID: 31709531 DOI: 10.1002/ijc.32779] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 10/28/2019] [Indexed: 12/14/2022]
Abstract
Increasing evidence suggests that IL-33 plays an important role in regulating tumor development. However, conflicting results, obtained from numerous studies, have highlighted the divergent functions of IL-33. The detailed mechanisms by which IL-33 modulates tumor development merit further investigation. Here, we report that IL-33 administration can effectively inhibit the development of pulmonary metastasis of breast cancer in a mouse. In our model, IL-33 promotes the production of TNF-α by macrophages, which increases IL-33 specific receptor (ST2) expression on natural killer (NK) cells and is pivotal in IL-33-induced NK cell activation. IL-33 treatment also facilitates the production of CCL5 in the lung by eosinophils and CD8+ T cells, which mediates the recruitment of NK cells to the tumor microenvironment. The systemic activation and local recruitment of NK cells result in potent tumor rejection in the lung. Our study reports a novel mechanism for the IL-33-meditated suppression of metastatic cancer and provides potential therapeutic strategies for targeting metastatic tumor.
Collapse
Affiliation(s)
- Lu Qi
- Department of Oncology of The First Affiliated Hospital, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China
| | - Qiuyan Zhang
- Department of Oncology of The First Affiliated Hospital, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China
| | - Yuhui Miao
- Department of Oncology of The First Affiliated Hospital, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China
| | - Wenyao Kang
- Department of Oncology of The First Affiliated Hospital, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China
| | - Zhigang Tian
- Department of Oncology of The First Affiliated Hospital, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China
| | - Damo Xu
- School of Medicine, Shenzhen University, Shenzhen, China
| | - Weihua Xiao
- Department of Oncology of The First Affiliated Hospital, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China
| | - Fang Fang
- Department of Oncology of The First Affiliated Hospital, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China
| |
Collapse
|
354
|
Zaaqoq AM, Namas RA, Abdul-Malak O, Almahmoud K, Barclay D, Yin J, Zamora R, Rosengart MR, Billiar TR, Vodovotz Y. Diurnal Variation in Systemic Acute Inflammation and Clinical Outcomes Following Severe Blunt Trauma. Front Immunol 2019; 10:2699. [PMID: 31824494 PMCID: PMC6879654 DOI: 10.3389/fimmu.2019.02699] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 11/04/2019] [Indexed: 12/26/2022] Open
Abstract
Animal studies suggest that the time of day is a determinant of the immunological response to both injury and infection. We hypothesized that due to this diurnal variation, time of injury could affect the systemic inflammatory response and outcomes post-trauma and tested this hypothesis by examining the dynamics of circulating inflammatory mediators in blunt trauma patients injured during daytime vs. nighttime. From a cohort of 472 blunt trauma survivors, two stringently matched sub-cohorts of moderately/severely injured patients [injury severity score (ISS) >20] were identified. Fifteen propensity-matched, daytime-inured (“mDay”) patients (age 43.6 ± 5.2, M/F 11/4, ISS 22.9 ± 0.7) presented during the shortest local annual period (8:00 am−5:00 pm), and 15 propensity-matched “mNight” patients (age 43 ± 4.3, M/F 11/4, ISS 24.5 ± 2.5) presented during the shortest night period (10:00 pm−5:00 am). Serial blood samples were obtained (3 samples within the first 24 h and daily from days 1–7) from all patients. Thirty-two plasma inflammatory mediators were assayed. Two-way Analysis of Variance (ANOVA) was used to compare groups. Dynamic Network Analysis (DyNA) and Dynamic Bayesian Network (DyBN) inference were utilized to infer dynamic interrelationships among inflammatory mediators. Both total hospital and intensive care unit length of stay were significantly prolonged in the mNight group. Circulating IL-17A was elevated significantly in the mNight group from 24 h to 7 days post-injury. Circulating MIP-1α, IL-7, IL-15, GM-CSF, and sST2 were elevated in the mDay group. DyNA demonstrated elevated network complexity in the mNight vs. the mDay group. DyBN suggested that cortisol and sST2 were central nodes upstream of TGF-β1, chemokines, and Th17/protective mediators in both groups, with IL-6 being an additional downstream node in the mNight group only. Our results suggest that time of injury affects clinical outcomes in severely injured patients in a manner associated with an altered systemic inflammation program, possibly implying a role for diurnal or circadian variation in the response to traumatic injury.
Collapse
Affiliation(s)
- Akram M Zaaqoq
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Critical Care Medicine, MedStar Washington Hospital Center, Washington, DC, United States
| | - Rami A Namas
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Inflammation and Regeneration Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Othman Abdul-Malak
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Khalid Almahmoud
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Derek Barclay
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jinling Yin
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ruben Zamora
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Inflammation and Regeneration Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Matthew R Rosengart
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Critical Care Medicine, MedStar Washington Hospital Center, Washington, DC, United States
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Inflammation and Regeneration Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Yoram Vodovotz
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Inflammation and Regeneration Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
355
|
Batyrova B, Luwaert F, Maravelia P, Miyabayashi Y, Vashist N, Stark JM, Soori SY, Tibbitt CA, Riese P, Coquet JM, Chambers BJ. PD-1 expression affects cytokine production by ILC2 and is influenced by peroxisome proliferator-activated receptor-γ. IMMUNITY INFLAMMATION AND DISEASE 2019; 8:8-23. [PMID: 31742928 PMCID: PMC7016838 DOI: 10.1002/iid3.279] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 11/03/2019] [Accepted: 11/06/2019] [Indexed: 12/25/2022]
Abstract
Introduction Innate lymphoid cells (ILCs) can provide early cytokine help against a variety of pathogens in the lungs and gastrointestinal tract. Type 2 ILC (ILC2) are comparable to T helper 2 cells found in the adaptive immune system, which secrete cytokines such as interleukin 5 (IL‐5) and IL‐13 and have been found to play roles in host defense against helminth infections and in allergic responses. Recent studies have identified that programmed cell death protein 1 (PD‐1) and peroxisome proliferator activated receptor‐γ (PPAR‐γ) are highly expressed by ILC2. We examined whether PD‐1 plays a role in ILC2 function and whether there was any connection between PD‐1 and PPAR‐γ Methods To ensure that only innate immune cells were present, ILC2 cells were examined from RAG1−/− and PD‐1−/−xRAG1−/− mice under steady‐state or following inoculation with IL‐33. We also tested ILC2 generated from bone marrow of RAG1−/− and PD‐1−/−xRAG1−/− mice for their production of cytokines. These in vitro‐derived ILC2 were also exposed to agonist and antagonist of PPAR‐γ. Results We found that ILC2 from PD‐1−/−xRAG1−/− mice had reduced frequencies of IL‐5 and IL‐13 producing cells both in vitro upon IL‐33 stimulation and in vivo following intraperitoneal administration of IL‐33 when compared with ILC2 from RAG1−/− mice. However, by adding IL‐2, IL‐25, and thymic stromal lymphopoietin to the in vitro cultures, the frequency of IL‐5 and IL‐13 expressing ILC2 from PD‐1−/−xRAG1−/− mice became similar to the frequency observed for ILC2 from RAG1−/− mice. In addition, PPAR‐γ agonists and antagonists were found to increase and decrease PD‐1 expression on ILC2 respectively. Conclusions These findings illustrate that chronic loss of PD‐1 plays a role in ILC2 function and PD‐1 expression can be modulated by PPAR‐γ.
Collapse
Affiliation(s)
- Banu Batyrova
- Department of Medicine, Centre for Infectious Medicine (CIM), Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Fien Luwaert
- Department of Medicine, Centre for Infectious Medicine (CIM), Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Panagiota Maravelia
- Department of Medicine, Centre for Infectious Medicine (CIM), Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Yuria Miyabayashi
- Department of Medicine, Centre for Infectious Medicine (CIM), Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Neha Vashist
- Department of Medicine, Centre for Infectious Medicine (CIM), Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden.,Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Julian M Stark
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institute, Stockholm, Sweden
| | - Sara Y Soori
- Department of Medicine, Centre for Infectious Medicine (CIM), Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Christopher A Tibbitt
- Department of Medicine, Centre for Infectious Medicine (CIM), Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden.,Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institute, Stockholm, Sweden
| | - Peggy Riese
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jonathan M Coquet
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institute, Stockholm, Sweden
| | - Benedict J Chambers
- Department of Medicine, Centre for Infectious Medicine (CIM), Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
356
|
Mannucci C, Calapai G, Gangemi S. Commentary: Circulatory pattern of cytokines, adipokines and bone markers in postmenopausal women with low BMD. Front Immunol 2019; 10:2666. [PMID: 31798591 PMCID: PMC6868060 DOI: 10.3389/fimmu.2019.02666] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 10/28/2019] [Indexed: 12/03/2022] Open
Affiliation(s)
- Carmen Mannucci
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Gioacchino Calapai
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Sebastiano Gangemi
- School and Division of Allergy and Clinical Immunology, Department of Experimental Medicine, University of Messina, Messina, Italy
| |
Collapse
|
357
|
Wang Z, Hua W, Li C, Chang H, Liu R, Ni Y, Sun H, Li Y, Wang X, Hou M, Liu Y, Xu Z, Ji M. Protective Role of Fecal Microbiota Transplantation on Colitis and Colitis-Associated Colon Cancer in Mice Is Associated With Treg Cells. Front Microbiol 2019; 10:2498. [PMID: 31798539 PMCID: PMC6861520 DOI: 10.3389/fmicb.2019.02498] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 10/16/2019] [Indexed: 12/18/2022] Open
Abstract
Colitis-associated cancer (CAC) is the most serious outcome of inflammatory bowel disease, which has an alteration of commensal intestinal microbiota. However, the role of intestinal microbiota on CAC progression is not well-understood. Fecal microbiota transplantation (FMT) was used for treating murine azoxymethane–dextran sodium sulfate (AOM-DSS) model of CAC. Composition of gut microbiota during FMT treatment was analyzed. RT-PCR and ELISA were used to detect the inflammatory factors, and immunofluorescence was applied to examine the phospho-nuclear factor (NF)-κB p65/p100 and Ki67-positive cells in the colons. In addition, flow cytometry was performed to analyze the immune cell after FMT treatment. Rehabilitation of the intestinal microbiota by FMT restored both the ratio and diversity of microbiota during CAC progression. Remarkably, a favorable morphometric outcome characterized by decreased tumor load and size was observed in CAC mice with FMT treatment. In addition, an anti-inflammatory function of FMT was demonstrated by decreasing pro-inflammatory factors but increasing anti-inflammatory factors through inhibiting canonical NF-κB activity and cellular proliferation in colons of CAC mice. The expression of CD4+CD25+Foxp3+ regulatory T cells (Tregs) was significantly increased after FMT treatment in CAC mice, but not T helper (Th)1/2/17 cells. Our study aids in the understanding of CAC pathogenesis and reveals a previously unrecognized role for FMT in the treatment of CAC through restoring the intestinal microbiota and inducing regulatory T cells.
Collapse
Affiliation(s)
- Zitao Wang
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Wenjie Hua
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Chen Li
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Hao Chang
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Ran Liu
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Yangyue Ni
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Hongzhi Sun
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Yangyang Li
- Department of Endocrinology, The Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, China
| | - Xinyue Wang
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Min Hou
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Yu Liu
- Department of Endocrinology, The Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, China
| | - Zhipeng Xu
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Minjun Ji
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China.,Department of Endocrinology, The Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
358
|
Response to Letter to the Editor Analysis of Novel Cardiovascular Biomarkers in Patients With Pulmonary Hypertension (PH). Heart Lung Circ 2019; 28:e149-e150. [DOI: 10.1016/j.hlc.2019.05.169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 05/08/2019] [Indexed: 11/23/2022]
|
359
|
Bawazeer MA, Theoharides TC. IL-33 stimulates human mast cell release of CCL5 and CCL2 via MAPK and NF-κB, inhibited by methoxyluteolin. Eur J Pharmacol 2019; 865:172760. [PMID: 31669588 DOI: 10.1016/j.ejphar.2019.172760] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/18/2019] [Accepted: 10/25/2019] [Indexed: 12/20/2022]
Abstract
Mast Cells (MCs) are critical for allergic reactions but also play important roles in inflammation, following stimulation by non-allergic triggers such as cytokines. Upon stimulation, MCs secrete numerous newly synthesized mediators, but the mechanism of the release of chemokines, which are important in the pathogenesis of allergic and inflammatory diseases, remains unknown. IL-33 is an "alarmin", known to augment allergic stimulation of MCs, but its effect on the release of chemokines is not known. The present work investigated the action of IL-33 on the release of the chemokines CCL5 and CCL2 from human MCs, as well as the inhibitory effect of the flavonoid 3',4',5,7-tetramethoxyflavone (methoxyluteolin). Stimulation of cultured human MCs (LAD2) and primary MCs (hCBMCs) by IL-33 (1-100 ng/ml) increased the gene expression and the release of CCL5 (P < 0.0001) and CCL2 (P < 0.01). Stimulation with IL-33 (10 ng/ml) activated MAPK components, as shown by phosphorylation of p38α MAPK, JNK, and c-Jun using Western blot analysis. Inhibition of these responses by known inhibitors confirmed that CCL5 and CCL2 are stimulated by the activation of p38α MAPK, JNK, and IκB-α. The gene expression and the release of CCL5 and CCL2 stimulated by IL-33 were significantly inhibited by 2 h pre-treatment with methoxyluteolin (10, 50, 100 μM). The inhibition by methoxyluteolin (50 μM) was not mediated via MAPK inhibition as phosphorylated p38α MAPK and JNK expression were not affected. In conclusion, IL-33 plays an important role in chemokine release from human MCs and that is by activation of more than one signaling pathway. The inhibitory effect of methoxyluteolin may indicate that it can be developed as a novel treatment for inflammatory diseases.
Collapse
Affiliation(s)
- Mona Abubakr Bawazeer
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, MA, USA; Graduate Program in Pharmacology and Experimental Therapeutics, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA; College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Theoharis C Theoharides
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, MA, USA; Graduate Program in Pharmacology and Experimental Therapeutics, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA; Department of Internal Medicine, Tufts University School of Medicine and Tufts Medical Center, Boston, MA, USA.
| |
Collapse
|
360
|
The ST2/Interleukin-33 Axis in Hematologic Malignancies: The IL-33 Paradox. Int J Mol Sci 2019; 20:ijms20205226. [PMID: 31652497 PMCID: PMC6834139 DOI: 10.3390/ijms20205226] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 02/07/2023] Open
Abstract
Interleukin (IL)-33 is a chromatin-related nuclear interleukin that is a component of IL-1 family. IL-33 production augments the course of inflammation after cell damage or death. It is discharged into the extracellular space. IL-33 is regarded as an “alarmin” able to stimulate several effectors of the immune system, regulating numerous immune responses comprising cancer immune reactions. IL-33 has been demonstrated to influence tumorigenesis. However, as far as this cytokine is concerned, we are faced with what has sometimes been defined as the IL-33 paradox. Several studies have demonstrated a relevant role of IL-33 to numerous malignancies, where it may have pro- and—less frequently—antitumorigenic actions. In the field of hematological malignancies, the role of IL-33 seems even more complex. Although we can affirm the existence of a negative role of IL-33 in Chronic myelogenos leukemia (CML) and in lymphoproliferative diseases and a positive role in pathologies such as Acute myeloid leukemia (AML), the action of IL-33 seems to be multiple and sometimes contradictory within the same pathology. In the future, we will have to learn to govern the negative aspects of activating the IL-33/ST2 axis and exploit the positive ones.
Collapse
|
361
|
Possible Roles of IL-33 in the Innate-Adaptive Immune Crosstalk of Psoriasis Pathogenesis. Mediators Inflamm 2019; 2019:7158014. [PMID: 31736655 PMCID: PMC6815589 DOI: 10.1155/2019/7158014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 08/26/2019] [Indexed: 01/18/2023] Open
Abstract
Background IL-33 belongs to the IL-1 family, playing a role in several biologic processes as well as in the pathogenesis of different diseases, including skin pathologies. It acts as an alarmin, released by damaged cells. Binding to a ST2 receptor, it stimulates many immune cells such as ILC2 and Th2 cells. IL-33/ST2 axis seems to be involved in Th17 response. According to this, a review was performed to analyze if IL-33 even interplay in the onset of psoriasis, a Th1/Th17 inflammatory disease. Methods Data obtained from the included articles are study author name, publication date, group studied, clinical and biological variables, laboratory tests, and outcome of interest of the study. Results Data are obtained from the 19 studies identified, which assessed the association between IL-33 and psoriasis. Discussion It seems to promote the innate-adaptive immune crosstalk: it could induce mast cells and neutrophil response after being released by injured keratinocytes and after stimulation by some cytokines, in particular TNFα, INFγ, and IL-17A. In addition, it seems to be involved from the onset of disease to the development of comorbidities, as psoriatic arthritis. Conclusion The core of the future research on psoriasis could be to fully understand the role of this complex cytokine, in order also to find a new therapeutic approach.
Collapse
|
362
|
Zhang Y, He J, Zheng H, Huang S, Lu F. Association of TREM-1, IL-1β, IL-33/ST2, and TLR Expressions With the Pathogenesis of Ocular Toxoplasmosis in Mouse Models on Different Genetic Backgrounds. Front Microbiol 2019; 10:2264. [PMID: 31649630 PMCID: PMC6794992 DOI: 10.3389/fmicb.2019.02264] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 09/17/2019] [Indexed: 12/11/2022] Open
Abstract
Ocular toxoplasmosis (OT) is one of the most common causes of posterior uveitis. The signaling of triggering receptor expressed on myeloid cells (TREM)-1 amplifies inflammation, whereas TREM-2 signaling is anti-inflammatory. IL-1β is a major driver of inflammation during infection. Toll-like receptors (TLRs) play important roles in protective immune response during Toxoplasma gondii infection, and interleukin (IL)-33 receptor (T1/ST2) signaling prevents toxoplasmic encephalitis in mice. However, the pathogenic mechanisms of OT are not yet well elucidated. To investigate the role of TREM-1, TREM-2, IL-1β, IL-33/ST2, and TLRs in OT of susceptible C57BL/6 (B6) and resistant BALB/c mice, both strains of mice were intravitreally infected with 500 tachyzoites of the RH strain of T. gondii. Histopathological analysis showed that T. gondii-infected B6 mice had more severe ocular damage observed by light microscopy, higher number of neutrophil elastase-positive cells in the eyes detected by immunohistochemical staining, more T. gondii tachyzoites in the eyes observed by transmission electron microscopy, and higher mRNA expression levels of tachyzoite-specific surface antigen 1 detected by quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR) in comparison of T. gondii-infected BALB/c mice. Detected by using qRT-PCR, the mRNA expression levels of TREM-1, IL-1β, IL-33, ST2, TLR11, TLR12, and TLR13 were significantly higher in the eyes of T. gondii-infected B6 mice than those of T. gondii-infected BALB/c mice, whereas the mRNA expression levels of TLR3 and TLR9 were significantly higher in the eyes of T. gondii-infected BALB/c mice than those of T. gondii-infected B6 mice. Correlation analysis showed that significant positive correlations existed between TREM-1 and IL-1β/IL-33/ST2/TLR9/TLR11 in the eyes of B6 mice and existed between TREM-1 and IL-33/ST2/TLR3/TLR9/TLR13 in the eyes of BALB/c mice after ocular T. gondii infection. Our data revealed that, compared with T. gondii-resistant BALB/c mice, ocular T. gondii infection can stimulate higher production of TREM-1, IL-33, ST2, TLR11, TLR12, and TLR13 in the eyes of T. gondii-susceptible B6 mice, however, whether those lead to more severe ocular pathology in the susceptible B6 mice remain to be further studied.
Collapse
Affiliation(s)
- Yanxia Zhang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Jian He
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Public Experimental Teaching Center, Sun Yat-sen University, Guangzhou, China
| | - Huanqin Zheng
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Shiguang Huang
- School of Stomatology, Jinan University, Guangzhou, China
| | - Fangli Lu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
363
|
Circulatory factors associated with function and prognosis in patients with severe heart failure. Clin Res Cardiol 2019; 109:655-672. [PMID: 31562542 PMCID: PMC7239817 DOI: 10.1007/s00392-019-01554-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 09/13/2019] [Indexed: 02/07/2023]
Abstract
Background Multiple circulatory factors are increased in heart failure (HF). Many have been linked to cardiac and/or skeletal muscle tissue processes, which in turn might influence physical activity and/or capacity during HF. This study aimed to provide a better understanding of the mechanisms linking HF with the loss of peripheral function. Methods and results Physical capacity measured by maximum oxygen uptake, myocardial function (measured by echocardiography), physical activity (measured by accelerometry), and mortality data was collected for patients with severe symptomatic heart failure an ejection fraction < 35% (n = 66) and controls (n = 28). Plasma circulatory factors were quantified using a multiplex immunoassay. Multivariate (orthogonal projections to latent structures discriminant analysis) and univariate analyses identified many factors that differed significantly between HF and control subjects, mainly involving biological functions related to cell growth and cell adhesion, extracellular matrix organization, angiogenesis, and inflammation. Then, using principal component analysis, links between circulatory factors and physical capacity, daily physical activity, and myocardial function were identified. A subset of ten biomarkers differentially expressed in patients with HF vs controls covaried with physical capacity, daily physical activity, and myocardial function; eight of these also carried prognostic value. These included established plasma biomarkers of HF, such as NT-proBNP and ST2 along with recently identified factors such as GDF15, IGFBP7, and TfR, as well as a new factor, galectin-4. Conclusions These findings reinforce the importance of systemic circulatory factors linked to hemodynamic stress responses and inflammation in the pathogenesis and progress of HF disease. They also support established biomarkers for HF and suggest new plausible markers. Graphic abstract ![]()
Electronic supplementary material The online version of this article (10.1007/s00392-019-01554-3) contains supplementary material, which is available to authorized users.
Collapse
|
364
|
Pleural Effusion IL-33/sST2 Levels and Effects of Low and High IL-33/sST2 Levels on Human Mesothelial Cell Adhesion and Migration. Inflammation 2019; 42:2072-2085. [DOI: 10.1007/s10753-019-01070-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
365
|
Tsai YG, Liou JH, Hung SI, Chen CB, Chiu TM, Wang CW, Chung WH. Increased Type 2 Innate Lymphoid Cells in Patients with Drug Reaction with Eosinophilia and Systemic Symptoms Syndrome. J Invest Dermatol 2019; 139:1722-1731. [PMID: 30735685 DOI: 10.1016/j.jid.2018.10.048] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 09/14/2018] [Accepted: 10/16/2018] [Indexed: 12/17/2022]
|
366
|
Diedrichs F, Stolk M, Jürchott K, Haag M, Sittinger M, Seifert M. Enhanced Immunomodulation in Inflammatory Environments Favors Human Cardiac Mesenchymal Stromal-Like Cells for Allogeneic Cell Therapies. Front Immunol 2019; 10:1716. [PMID: 31396228 PMCID: PMC6665953 DOI: 10.3389/fimmu.2019.01716] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 07/09/2019] [Indexed: 12/29/2022] Open
Abstract
Rising numbers of patients with cardiovascular diseases and limited availability of donor hearts require new and improved therapy strategies. Human atrial appendage-derived cells (hAACs) are promising candidates for an allogeneic cell-based treatment. In this study, we evaluated their inductive and modulatory capacity regarding immune responses and underlying key mechanisms in vitro. For this, cryopreserved hAACs were either cultured in the presence of interferon-gamma (IFNγ) or left unstimulated. The expression of characteristic mesenchymal stromal cell markers (CD29, CD44, CD73, CD105, CD166) was revealed by flow cytometry that also highlighted a predominant negativity for CD90. A low immunogeneic phenotype in an inflammatory milieu was shown by lacking expression of co-stimulatory molecules and upregulation of the inhibitory ligands PD-L1 and PD-L2, despite de novo expression of HLA-DR. Co-cultures of hAACs with allogeneic peripheral blood mononuclear cells, proved their low immunogeneic state by absence of induced T cell proliferation and activation. Additionally, elevated levels of IL-1β, IL-33, and IL-10 were detectable in those cell culture supernatants. Furthermore, the immunomodulatory potential of hAACs was assessed in co-cultures with αCD3/αCD28-activated peripheral blood mononuclear cells. Here, a strong inhibition of T cell proliferation and reduction of pro-inflammatory cytokines (IFNγ, TNFα, TNFβ, IL-17A, IL-2) were observable after pre-stimulation of hAACs with IFNγ. Transwell experiments confirmed that mostly soluble factors are responsible for these suppressive effects. We were able to identify indolamin-2,3-dioxygenase (IDO) as a potential key player through a genome-wide gene expression analysis and could demonstrate its involvement in the observed immunological responses. While the application of blocking antibodies against both PD-1 ligands did not affect the immunomodulation by hAACs, 1-methyl-L-tryptophan as specific inhibitor of IDO was able to restore proliferation and to lower apoptosis of T cells. In conclusion, hAACs represent a cardiac-derived mesenchymal stromal-like cell type with a high potential for the application in an allogeneic setting, since they do not trigger T cell responses and even increase their immunomodulatory potential in inflammatory environments.
Collapse
Affiliation(s)
- Falk Diedrichs
- Berlin Institute of Health (BIH), Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Meaghan Stolk
- BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Karsten Jürchott
- BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Institute of Medical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Marion Haag
- BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Tissue Engineering Laboratory, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Michael Sittinger
- BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Tissue Engineering Laboratory, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Martina Seifert
- BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Institute of Medical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
367
|
Vianello E, Dozio E, Bandera F, Schmitz G, Nebuloni M, Longhi E, Tacchini L, Guazzi M, Corsi Romanelli MM. Dysfunctional EAT thickness may promote maladaptive heart remodeling in CVD patients through the ST2-IL33 system, directly related to EPAC protein expression. Sci Rep 2019; 9:10331. [PMID: 31316160 PMCID: PMC6637132 DOI: 10.1038/s41598-019-46676-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 07/03/2019] [Indexed: 01/11/2023] Open
Abstract
Dysfunctional epicardial adipose tissue (EAT) secretome can influence the heart’s stretch response. However, the molecular mechanisms are still poorly understood. The aim of this study was to clarify how dysfunctional EAT promotes maladaptive heart remodeling in cardiovascular disease (CVD) through ST2 production associated with exchange protein directly activated by cAMP (EPAC) proteins. A series of 55 CVD males were enrolled and their EAT thickness, LV mass and volumes were measured by echocardiography. Blood, plasma and EAT biopsies were collected for molecular and proteomic assays. Taking EAT thickness as a continuous variable there was a direct correlation between the ST2 cardiac stretch mediator and EAT thickness (r = 0.54, p < 0.01) and an inverse relation between the ST2 gene and IL-33 expression (r −0.50, p < 0.01). In the CVD population EPAC2 expression directly correlated with the ST2 gene (r = 0.74, p < 0.0001) causing an ST2/IL-33 system local (p < 0.001) and systemic (sST2 = 57.33 ± 3.22 and IL-33 = 0.53 ± 017 pg/mL; p < 0.0001) protein imbalance associated with maladaptive remodeling. This indicated that dysfunctional EAT is a source of both EPAC and ST2 protein and an EPAC2 isoform seems involved in ST2 production in adipose tissue. Both EPAC2 and ST2 expression were directly related to maladaptive heart remodeling indices, suggesting EAT measurements could be useful in the early assessment of CVD complications.
Collapse
Affiliation(s)
- Elena Vianello
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy.
| | - Elena Dozio
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Francesco Bandera
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy.,Cardiology University Department, Heart Failure Unit, IRCCS Policlinico San Donato, San Donato Milanese, Milano, Italy
| | - Gerd Schmitz
- Department of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Manuela Nebuloni
- U.O.C. of Surgical Pathology, Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, Milan, Italy
| | - Erika Longhi
- U.O.C. of Surgical Pathology, Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, Milan, Italy
| | - Lorenza Tacchini
- Cardiology University Department, Heart Failure Unit, IRCCS Policlinico San Donato, San Donato Milanese, Milano, Italy
| | - Marco Guazzi
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy.,Cardiology University Department, Heart Failure Unit, IRCCS Policlinico San Donato, San Donato Milanese, Milano, Italy
| | - Massimiliano Marco Corsi Romanelli
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy.,U.O.C. SMEL-1 of Clinical Pathology, IRCCS Policlinico San Donato, San Donato Milanese, Milano, Italy
| |
Collapse
|
368
|
Nie Y, Hu Y, Yu K, Zhang D, Shi Y, Li Y, Sun L, Qian F. Akt1 regulates pulmonary fibrosis via modulating IL-13 expression in macrophages. Innate Immun 2019; 25:451-461. [PMID: 31299858 PMCID: PMC6900639 DOI: 10.1177/1753425919861774] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Idiopathic pulmonary fibrosis is a progressive interstitial pneumonia characterised by fibroblast accumulation, collagen deposition and extracellular matrix (ECM) remodelling. It was reported that Akt1 mediated idiopathic pulmonary fibrosis progression through regulating the apoptosis of alveolar macrophage, while its effect on macrophage-produced cytokines remains largely unknown. In the present study, we first examined the phosphorylation of Akt1 in lung sections from idiopathic pulmonary fibrosis patients by immunohistochemistry before applying a bleomycin-induced idiopathic pulmonary fibrosis model using Akt1−/− mice and Akt1+/+ littermates. The results showed that Akt1 was remarkably up-regulated in idiopathic pulmonary fibrosis patients, while in vivo studies revealed that Akt1-deficient mice had well-preserved alveolar structure and fewer collagens, secreted fewer matrix components, including alpha smooth-muscle actin and fibronectin and survived significantly longer than Akt1+/+ littermates. Additionally, the pro-fibrogenic cytokine IL-13 was down-regulated at least twofold in Akt1−/−mice compared to the Akt1+/+group on d 3 and 7 after bleomycin treatment. Furthermore, it was found that Akt1–/– macrophages displayed down-regulation of IL-13 compared to Akt1+/+ macrophages in which Akt1 was phosphorylated in response to IL-33 stimulation. These findings indicate that Akt1 modulates pulmonary fibrosis through inducing IL-13 production by macrophages, suggesting that targeting Akt1 may simultaneously block the fibrogenic processes of idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Yunjuan Nie
- 1 Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, PR China
| | - Yudong Hu
- 2 Engineering Research Center of Cell and Therapeutic Ab, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, PR China
| | - Kaikai Yu
- 2 Engineering Research Center of Cell and Therapeutic Ab, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, PR China
| | - Dan Zhang
- 3 Research Center for Cancer Precision Medicine, Bengbu Medical College, PR China
| | - Yinze Shi
- 1 Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, PR China
| | - Yaolin Li
- 1 Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, PR China
| | - Lei Sun
- 2 Engineering Research Center of Cell and Therapeutic Ab, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, PR China
| | - Feng Qian
- 2 Engineering Research Center of Cell and Therapeutic Ab, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, PR China.,3 Research Center for Cancer Precision Medicine, Bengbu Medical College, PR China
| |
Collapse
|
369
|
Landskron G, De la Fuente López M, Dubois-Camacho K, Díaz-Jiménez D, Orellana-Serradell O, Romero D, Sepúlveda SA, Salazar C, Parada-Venegas D, Quera R, Simian D, González MJ, López-Köstner F, Kronberg U, Abedrapo M, Gallegos I, Contreras HR, Peña C, Díaz-Araya G, Roa JC, Hermoso MA. Interleukin 33/ST2 Axis Components Are Associated to Desmoplasia, a Metastasis-Related Factor in Colorectal Cancer. Front Immunol 2019; 10:1394. [PMID: 31281317 PMCID: PMC6598075 DOI: 10.3389/fimmu.2019.01394] [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: 09/22/2018] [Accepted: 06/03/2019] [Indexed: 12/24/2022] Open
Abstract
In colorectal cancer (CRC), cancer-associated fibroblasts (CAFs) are the most abundant component from the tumor microenvironment (TM). CAFs facilitate tumor progression by inducing angiogenesis, immune suppression and invasion, thus altering the organization/composition of the extracellular matrix (i.e., desmoplasia) and/or activating epithelial-mesenchymal transition (EMT). Soluble factors from the TM can also contribute to cell invasion through secretion of cytokines and recently, IL-33/ST2 pathway has gained huge interest as a protumor alarmin, promoting progression to metastasis by inducing changes in TM. Hence, we analyzed IL-33 and ST2 content in tumor and healthy tissue lysates and plasma from CRC patients. Tissue localization and distribution of these molecules was evaluated by immunohistochemistry (using localization reference markers α-smooth muscle actin or α-SMA and E-cadherin), and clinical/histopathological information was obtained from CRC patients. In vitro experiments were conducted in primary cultures of CAFs and normal fibroblasts (NFs) isolated from tumor and healthy tissue taken from CRC patients. Additionally, migration and proliferation analysis were performed in HT29 and HCT116 cell lines. It was found that IL-33 content increases in left-sided CRC patients with lymphatic metastasis, with localization in tumor epithelia associated with abundant desmoplasia. Although ST2 content showed similarities between tumor and healthy tissue, a decreased immunoreactivity was observed in left-sided tumor stroma, associated to metastasis related factors (advanced stages, abundant desmoplasia, and presence of tumor budding). A principal component analysis (including stromal and epithelial IL-33/ST2 and α-SMA immunoreactivity with extent of desmoplasia) allowed us to distinguish clusters of low, intermediate and abundant desmoplasia, with potential to develop a diagnostic signature with benefits for further therapeutic targets. IL-33 transcript levels from CAFs directly correlated with CRC cell line migration induced by CAFs conditioned media, with rhIL-33 inducing a mesenchymal phenotype in HT29 cells. These results indicate a role of IL-33/ST2 in tumor microenvironment, specifically in the interaction between CAFs and epithelial tumor cells, thus contributing to invasion and metastasis in left-sided CRC, most likely by activating desmoplasia.
Collapse
Affiliation(s)
- Glauben Landskron
- Immunology Program, Innate Immunity Laboratory, Faculty of Medicine, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile
| | - Marjorie De la Fuente López
- Immunology Program, Innate Immunity Laboratory, Faculty of Medicine, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile.,Research Sub-direction, Academic Direction, Clinica Las Condes, Santiago, Chile
| | - Karen Dubois-Camacho
- Immunology Program, Innate Immunity Laboratory, Faculty of Medicine, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile
| | - David Díaz-Jiménez
- Immunology Program, Innate Immunity Laboratory, Faculty of Medicine, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile
| | - Octavio Orellana-Serradell
- Immunology Program, Innate Immunity Laboratory, Faculty of Medicine, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile
| | - Diego Romero
- Pathology Department, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Santiago A Sepúlveda
- Pathology Department, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Christian Salazar
- Immunology Program, Innate Immunity Laboratory, Faculty of Medicine, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile
| | - Daniela Parada-Venegas
- Immunology Program, Innate Immunity Laboratory, Faculty of Medicine, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile
| | - Rodrigo Quera
- Inflammatory Bowel Disease Program, Gastroenterology Department, Clinica Las Condes, Santiago, Chile
| | - Daniela Simian
- Research Sub-direction, Academic Direction, Clinica Las Condes, Santiago, Chile
| | - María-Julieta González
- Cell and Molecular Biology Program, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | | | - Udo Kronberg
- Coloproctology Department, Clinica Las Condes, Santiago, Chile
| | - Mario Abedrapo
- Coloproctology Department, Clinica Las Condes, Santiago, Chile.,Coloproctology Surgery Department, Hospital Clinico Universidad de Chile, Santiago, Chile
| | - Iván Gallegos
- Pathology Department, Hospital Clinico Universidad de Chile, Santiago, Chile
| | - Héctor R Contreras
- Department of Basic and Clinic Oncology, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Cristina Peña
- Medical Oncology Department, Ramon y Cajal University Hospital, IRYCIS, CIBERONC, Madrid, Spain
| | - Guillermo Díaz-Araya
- Molecular Pharmacology Laboratory, Faculty of Chemical Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
| | - Juan Carlos Roa
- Pathology Department, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Marcela A Hermoso
- Immunology Program, Innate Immunity Laboratory, Faculty of Medicine, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile
| |
Collapse
|
370
|
Copsel S, Wolf D, Komanduri KV, Levy RB. The promise of CD4 +FoxP3 + regulatory T-cell manipulation in vivo: applications for allogeneic hematopoietic stem cell transplantation. Haematologica 2019; 104:1309-1321. [PMID: 31221786 PMCID: PMC6601084 DOI: 10.3324/haematol.2018.198838] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/07/2019] [Indexed: 12/12/2022] Open
Abstract
CD4+FoxP3+ regulatory T cells (Tregs) are a non-redundant population critical for the maintenance of self-tolerance. Over the past decade, the use of these cells for therapeutic purposes in transplantation and autoimmune disease has emerged based on their capacity to inhibit immune activation. Basic science discoveries have led to identifying key receptors on Tregs that can regulate their proliferation and function. Notably, the understanding that IL-2 signaling is crucial for Treg homeostasis promoted the hypothesis that in vivo IL-2 treatment could provide a strategy to control the compartment. The use of low-dose IL-2 in vivo was shown to selectively expand Tregs versus other immune cells. Interestingly, a number of other Treg cell surface proteins, including CD28, CD45, IL-33R and TNFRSF members, have been identified which can also induce activation and proliferation of this population. Pre-clinical studies have exploited these observations to prevent and treat mice developing autoimmune diseases and graft-versus-host disease post-allogeneic hematopoietic stem cell transplantation. These findings support the development of translational strategies to expand Tregs in patients. Excitingly, the use of low-dose IL-2 for patients suffering from graft-versus-host disease and autoimmune disease has demonstrated increased Treg levels together with beneficial outcomes. To date, promising pre-clinical and clinical studies have directly targeted Tregs and clearly established the ability to increase their levels and augment their function in vivo. Here we review the evolving field of in vivo Treg manipulation and its application to allogeneic hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
| | | | - Krishna V Komanduri
- Department of Microbiology and Immunology.,Sylvester Comprehensive Cancer Center.,Division of Transplantation and Cellular Therapy, Department of Medicine
| | - Robert B Levy
- Department of Microbiology and Immunology .,Division of Transplantation and Cellular Therapy, Department of Medicine.,Department of Ophthalmology, Miller School of Medicine, University of Miami, FL, USA
| |
Collapse
|
371
|
Interleukin 1 Receptor-Like 1 (IL1RL1) Promotes Airway Bacterial and Viral Infection and Inflammation. Infect Immun 2019; 87:IAI.00340-19. [PMID: 31061143 DOI: 10.1128/iai.00340-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 04/30/2019] [Indexed: 01/14/2023] Open
Abstract
Interleukin 1 receptor-like 1 (IL1RL1), also known as suppression of tumorigenicity 2 (ST2), is the receptor for interleukin 33 (IL-33) and has been increasingly studied in type 2 inflammation. An increase in airway IL-33/ST2 signaling in asthma has been associated with eosinophilic inflammation, but little is known about the role of ST2 in neutrophilic inflammation. Airway Mycoplasma pneumoniae and human rhinovirus (HRV) infections are linked to neutrophilic inflammation during acute exacerbations of asthma. However, whether ST2 contributes to M. pneumoniae- and HRV-mediated airway inflammation is poorly understood. The current study sought to determine the functions of ST2 during airway M. pneumoniae or HRV infection. In cultured normal human primary airway epithelial cells, ST2 overexpression (OE) increased the production of neutrophilic chemoattractant IL-8 in the absence or presence of M. pneumoniae or HRV1B infection. ST2 OE also enhanced HRV1B-induced IP-10, a chemokine involved in asthma exacerbations. In the M. pneumoniae-infected mouse model, ST2 deficiency, in contrast to sufficiency, significantly reduced the levels of neutrophils following acute (≤24 h) infection, while in the HRV1B-infected mouse model, ST2 deficiency significantly reduced the levels of proinflammatory cytokines KC, IP-10, and IL-33 in bronchoalveolar lavage (BAL) fluid. Overall, ST2 overexpression in human epithelial cells and ST2 sufficiency in mice increased the M. pneumoniae and HRV loads in cell supernatants and BAL fluid. After pathogen infection, ST2-deficient mice showed a higher level of the host defense protein lactotransferrin in BAL fluid. Our data suggest that ST2 promotes proinflammatory responses (e.g., neutrophils) to airway bacterial and viral infection and that blocking ST2 signaling may broadly attenuate airway infection and inflammation.
Collapse
|
372
|
Rönnberg E, Ghaib A, Ceriol C, Enoksson M, Arock M, Säfholm J, Ekoff M, Nilsson G. Divergent Effects of Acute and Prolonged Interleukin 33 Exposure on Mast Cell IgE-Mediated Functions. Front Immunol 2019; 10:1361. [PMID: 31275312 PMCID: PMC6593472 DOI: 10.3389/fimmu.2019.01361] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 05/29/2019] [Indexed: 12/12/2022] Open
Abstract
Background: Epithelial cytokines, including IL-33 and Thymic stromal lymphopoietin (TSLP), have attracted interest because of their roles in chronic allergic inflammation-related conditions such as asthma. Mast cells are one of the major targets of IL-33, to which they respond by secreting cytokines. Most studies performed thus far have investigated the acute effects of IL-33 on mast cells. In the current study, we investigated how acute vs. prolonged exposure of mast cells to IL-33 and TSLP affects mediator synthesis and IgE-mediated activation. Methods: Human lung mast cells (HLMCs), cord blood-derived mast cells (CBMCs), and the ROSA mast cell line were used for this study. Receptor expression and the levels of mediators were measured after treatment with IL-33 and/or TSLP. Results: IL-33 induced the release of cytokines. Prolonged exposure to IL-33 increased while TSLP reduced intracellular levels of tryptase. Acute IL-33 treatment strongly potentiated IgE-mediated activation. In contrast, 4 days of exposure to IL-33 decreased IgE-mediated activation, an effect that was accompanied by a reduction in FcεRI expression. Conclusion: We show that IL-33 plays dual roles in mast cells, in which its acute effects include cytokine release and the potentiation of IgE-mediated degranulation, whereas prolonged exposure to IL-33 reduces IgE-mediated activation. We conclude that mast cells act quickly in response to the alarmin IL-33 to initiate an acute inflammatory response, whereas extended exposure to IL-33 during prolonged inflammation reduces IgE-mediated responses. This negative feedback effect suggests the presence of a novel regulatory pathway that modulates IgE-mediated human mast cell responses.
Collapse
Affiliation(s)
- Elin Rönnberg
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Avan Ghaib
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
- Department of Microbiology, College of Medicine, University of Sulaimani, Sulaimani, Iraq
| | - Carlos Ceriol
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Mattias Enoksson
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Michel Arock
- Molecular and Cellular Oncology, LBPA CNRS UMR 8113, Ecole Normale Supérieure de Cachan, Cachan, France
- Laboratoire Central d'Hématologie, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Jesper Säfholm
- The Unit for Asthma and Allergy Research, The Institute of Environmental Medicine, Karolinska Institutet, Solna, Sweden
| | - Maria Ekoff
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Gunnar Nilsson
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
373
|
Filippone RT, Sahakian L, Apostolopoulos V, Nurgali K. Eosinophils in Inflammatory Bowel Disease. Inflamm Bowel Dis 2019; 25:1140-1151. [PMID: 30856253 DOI: 10.1093/ibd/izz024] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Indexed: 12/16/2022]
Abstract
Clinical investigations in inflammatory bowel disease (IBD) patients have provided increasing evidence that eosinophils contribute to chronic intestinal inflammation. Accumulation of eosinophils in the gastrointestinal tract correlates with the variations of eosinophil regulatory molecules; however, their role in gastrointestinal dysfunction in IBD has not been fully elucidated. This review will describe the development and characterization of gastrointestinal eosinophils, mechanisms of eosinophil recruitment to the gastrointestinal tract. Moreover, the eosinophil-induced changes to the enteric nervous system associated with disease severity and gastrointestinal dysfunction will be analyzed with suggestive molecular pathways for enteric neuronal injury. Current and potential therapeutic interventions targeting eosinophils will be discussed.
Collapse
Affiliation(s)
- Rhiannon T Filippone
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Australia
| | - Lauren Sahakian
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Australia
| | - Vasso Apostolopoulos
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Australia
| | - Kulmira Nurgali
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Australia.,Department of Medicine Western Health, Melbourne University, Melbourne, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, Australia
| |
Collapse
|
374
|
McEntee CP, Finlay CM, Lavelle EC. Divergent Roles for the IL-1 Family in Gastrointestinal Homeostasis and Inflammation. Front Immunol 2019; 10:1266. [PMID: 31231388 PMCID: PMC6568214 DOI: 10.3389/fimmu.2019.01266] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/17/2019] [Indexed: 12/11/2022] Open
Abstract
Inflammatory disorders of the gastro-intestinal tract are a major cause of morbidity and significant burden from a health and economic perspective in industrialized countries. While the incidence of such conditions has a strong environmental component, in particular dietary composition, epidemiological studies have identified specific hereditary mutations which result in disequilibrium between pro- and anti-inflammatory factors. The IL-1 super-family of cytokines and receptors is highly pleiotropic and plays a fundamental role in the pathogenesis of several auto-inflammatory conditions including rheumatoid arthritis, multiple sclerosis and psoriasis. However, the role of this super-family in the etiology of inflammatory bowel diseases remains incompletely resolved despite extensive research. Herein, we highlight the currently accepted paradigms as they pertain to specific IL-1 family members and focus on some recently described non-classical roles for these pathways in the gastrointestinal tract. Finally, we address some of the shortcomings and sources of variance in the field which to date have yielded several conflicting results from similar studies and discuss the potential effect of these factors on data interpretation.
Collapse
Affiliation(s)
- Craig P McEntee
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom.,Faculty of Biology, Medicine and Health, Manchester Collaborative Centre for Inflammation Research, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Conor M Finlay
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom.,Faculty of Biology, Medicine and Health, Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Ed C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Advanced Materials and BioEngineering Research (AMBER), Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
375
|
Abstract
Since the pro-inflammatory cytokine IL-33 and its receptor (ST2) are closely involved in regulating both innate and adaptive immune responses, it is conceivable that they may play an important role in organ transplantation. IL-33 is broadly expressed by multiple cell types such as fibroblasts, epithelial cells, and endothelial cells. As a strong inducer of type 2 helper T (Th2) cellular immune responses, IL-33 can significantly prolong allograft survival in organ transplantation partially via altering gene expression profiles and increasing frequency of regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). Nevertheless, the IL-33 signaling pathway and its underlying mechanisms remain largely undefined in transplant biology. This present mini-review summarizes recent advances in the studies concerning the IL-33/ST2 signaling pathway and the analysis of its biological function in the field transplantation. The literature points to a deleterious role of activation of the IL-33/ST2 signaling pathway, giving rise to ischemia/reperfusion, acute kidney injury and failure, acute heart rejection, as well as liver fibrosis. Under pro-inflammatory conditions, IL-33 expression is upregulated. Alteration of IL-33 levels has been suggested as a biomarker for predicting organ injury and ongoing allogeneic transplant outcome. These studies have deepened our understanding of immunobiological role of IL-33 and its receptor in organ transplantation. Modulation of the IL-33/ST2 signaling pathway might be utilized as a therapeutic target in the clinic.
Collapse
Affiliation(s)
- Ying Jin
- Zhejiang University, Second Affiliated Hospital of School of Medicine, Department of Traditional Chinese Medicine & Rehabilitation, Hangzhou City, People's Republic of China
| | - Deqiang Kong
- Zhejiang University, Second Affiliated Hospital of School of Medicine, Department of General Surgery, Hangzhou City, People's Republic of China
| | - Chen Liu
- Zhejiang University, Second Affiliated Hospital of School of Medicine, Department of General Surgery, Hangzhou City, People's Republic of China
| | - Weihua Gong
- Zhejiang University, Second Affiliated Hospital of School of Medicine, Department of General Surgery, Hangzhou City, People's Republic of China
| |
Collapse
|
376
|
Olver TD, Edwards JC, Jurrissen TJ, Veteto AB, Jones JL, Gao C, Rau C, Warren CM, Klutho PJ, Alex L, Ferreira-Nichols SC, Ivey JR, Thorne PK, McDonald KS, Krenz M, Baines CP, Solaro RJ, Wang Y, Ford DA, Domeier TL, Padilla J, Rector RS, Emter CA. Western Diet-Fed, Aortic-Banded Ossabaw Swine: A Preclinical Model of Cardio-Metabolic Heart Failure. JACC Basic Transl Sci 2019; 4:404-421. [PMID: 31312763 PMCID: PMC6610000 DOI: 10.1016/j.jacbts.2019.02.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/13/2019] [Accepted: 02/21/2019] [Indexed: 12/12/2022]
Abstract
The development of new treatments for heart failure lack animal models that encompass the increasingly heterogeneous disease profile of this patient population. This report provides evidence supporting the hypothesis that Western Diet-fed, aortic-banded Ossabaw swine display an integrated physiological, morphological, and genetic phenotype evocative of cardio-metabolic heart failure. This new preclinical animal model displays a distinctive constellation of findings that are conceivably useful to extending the understanding of how pre-existing cardio-metabolic syndrome can contribute to developing HF.
Collapse
Key Words
- AB, aortic-banded
- CON, control
- EDPVR, end-diastolic pressure−volume relationship
- EF, ejection fraction
- HF, heart failure
- HFpEF, heart failure with preserved ejection fraction
- HFrEF, heart failure with reduced ejection fraction
- IL1RL1, interleukin 1 receptor-like 1
- LV, left ventricle
- NF, nuclear factor
- PTX3, pentraxin-3
- WD, Western Diet
- cardio-metabolic disease
- heart failure
- integrative pathophysiology
- preclinical model of cardiovascular disease
Collapse
Affiliation(s)
- T. Dylan Olver
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| | - Jenna C. Edwards
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| | - Thomas J. Jurrissen
- Department of Nutrition and Exercise Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Adam B. Veteto
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - John L. Jones
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Chen Gao
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Christoph Rau
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Chad M. Warren
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois
| | - Paula J. Klutho
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - Linda Alex
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | | | - Jan R. Ivey
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| | - Pamela K. Thorne
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| | - Kerry S. McDonald
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Maike Krenz
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - Christopher P. Baines
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - R. John Solaro
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois
| | - Yibin Wang
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - David A. Ford
- Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University- School of Medicine, St. Louis, Missouri
| | - Timothy L. Domeier
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri-Columbia, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
- Department of Child Health, University of Missouri-Columbia, Columbia, Missouri
| | - R. Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri-Columbia, Columbia, Missouri
- Department of Medicine – University of Missouri-Columbia, Columbia, Missouri
- Research Service, Harry S Truman Memorial VA Hospital, University of Missouri-Columbia, Columbia, Missouri
| | - Craig A. Emter
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| |
Collapse
|
377
|
Kalinkovich A, Livshits G. A cross talk between dysbiosis and gut-associated immune system governs the development of inflammatory arthropathies. Semin Arthritis Rheum 2019; 49:474-484. [PMID: 31208713 DOI: 10.1016/j.semarthrit.2019.05.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/09/2019] [Accepted: 05/28/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Emerging evidence suggests that dysbiosis, imbalanced gut microbial community, might be a key player in the development of various diseases, including inflammatory arthropathies, such as rheumatoid arthritis, spondyloarthritis (mainly, ankylosing spondylitis and psoriatic arthritis), and osteoarthritis. Yet, the underlying mechanisms and corresponding interactions remain poorly understood. METHODS We conducted a critical and extensive literature review to explore the association between dysbiosis and the development of inflammatory arthropathies. We also reviewed the literature to assess the perspectives that ameliorate inflammatory arthropathies by manipulating the microbiota with probiotics, prebiotics or fecal microbiota transplantation. RESULTS Some bacterial species (e.g. Prevotella, Citrobacter rodentium, Collinsella aerofaciens, Segmented filamentous bacteria) participate in the creation of the pro-inflammatory immune status, presumably via epitope mimicry, modification of self-antigens, enhanced cell apoptosis mechanisms, and destruction of tight junction proteins and intestinal barrier integrity, all leading to the development and maintainance of inflammatory arthropathies. Whether dysbiosis is an epiphenomenon or is an active driver of these disorders remains unclear, yet, recent observations clearly suggest that dysbiosis precedes and triggers their development implying a causative relationship between dysbiosis and inflammatory arthropathies. The underlying mechanisms include dysbiosis-mediated changes in the functional activity of the intestinal immune cell subsets, such as innate lymphoid cells, mucosa-associated invariant T cells, invariant natural killer T cells, T-follicular helper and T-regulatory cells. In turn, disturbed functionality of the gut-associated immune system is shown to promote the overgrowth of many bacteria, thus establishing a detrimental vicious circle of actively maintaining arthritis. CONCLUSIONS Analysis of the data described in the review supports the notion that a close, dynamic and tightly regulated cross talk between dysbiosis and the gut-associated immune system governs the development of inflammatory arthropathies.
Collapse
Affiliation(s)
- Alexander Kalinkovich
- Human Population Biology Research Unit, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Gregory Livshits
- Human Population Biology Research Unit, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
378
|
Pusceddu I, Dieplinger B, Mueller T. ST2 and the ST2/IL-33 signalling pathway-biochemistry and pathophysiology in animal models and humans. Clin Chim Acta 2019; 495:493-500. [PMID: 31136737 DOI: 10.1016/j.cca.2019.05.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/26/2019] [Accepted: 05/24/2019] [Indexed: 12/15/2022]
Abstract
ST2 is an interleukin (IL)-1 receptor family member with transmembrane (ST2L) and soluble (sST2) isoforms. Structurally, the ST2 gene products are very similar in mice and humans. In humans and in mice, alternative promoter activation and splicing produce ST2L and sST2. ST2L represents the longest transcript, whereas sST2 is the truncated, soluble isoform. ST2L is the biological receptor for IL-33, a member of the IL-1 family. IL-33 is the functional ligand of ST2L and signals the presence of tissue damage to local immune cells. IL-33/ST2L signalling leads to the production of inflammatory cytokines/chemokines and to the induction of the immune response. Conversely, sST2 functions as a decoy receptor for IL-33, inhibiting the effects of IL-33/ST2L signalling. Animal studies have allowed the investigation of ST2 and the IL-33/ST2L signalling pathway at multiple levels. However, clinical studies have mainly focused on the determination of sST2 in the circulation. In humans, plasma concentrations of sST2 increase in several diseases, such as heart disease, pulmonary disease, burn injury and graft-versus-host disease. Consequently, increased plasma concentrations of sST2 are not specific for a single disorder in humans and are thus of limited value for diagnostic purposes. However, increased plasma concentrations of sST2 have been linked to a worse prognosis in numerous diseases. Nevertheless, the major source of circulating sST2 in healthy and diseased humans is currently not fully established. In addition, whether the downregulation of sST2 can improve the outcome of patients in the clinical setting has not been elucidated. The aim of the present review was to provide an update on the findings regarding the biochemistry and pathophysiology of ST2 and the sST2 signalling pathway in humans and experimental models.
Collapse
Affiliation(s)
- Irene Pusceddu
- Department of Clinical Pathology, Hospital of Bolzano, Bolzano, Italy
| | - Benjamin Dieplinger
- Department of Laboratory Medicine, Konventhospital Barmherzige Brueder Linz, Linz, Austria
| | - Thomas Mueller
- Department of Clinical Pathology, Hospital of Bolzano, Bolzano, Italy.
| |
Collapse
|
379
|
Williams MA, O'Callaghan A, Corr SC. IL-33 and IL-18 in Inflammatory Bowel Disease Etiology and Microbial Interactions. Front Immunol 2019; 10:1091. [PMID: 31139196 PMCID: PMC6527769 DOI: 10.3389/fimmu.2019.01091] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/29/2019] [Indexed: 12/19/2022] Open
Abstract
The IL-1 cytokines are a newly expanded family, with each of its 11 members playing an important role in health and disease. Typically acting as pro- or anti-inflammatory mediators of first-line innate immunity, their production is particularly important in the context of mucosal defenses, through handling breach of the delicate epithelial barrier and mediating a local immune response to invading pathogens. Mucosal immunity is often aberrantly orchestrated in intestinal diseases, such as Inflammatory Bowel Disease (IBD). Various studies have pointed to IL-1 cytokines as being important players in IBD with context-dependent roles, either through promoting auto-inflammatory mechanisms, or alleviating disease through protection against breach of pathogens across the epithelial barrier. This mini-review will succinctly examine the role of IL-1 family members in IBD, with a special focus on the recently described IL-33 as well as IL-18, and will explore the disease models within which these cytokines have been studied. Furthermore, we will examine the evidence of interplay of these cytokines with the gut microbiota, with hopes of summarizing our current knowledge of these family members and their potential for unraveling novel molecular mechanisms of IBD pathology.
Collapse
Affiliation(s)
- Michelle A Williams
- Department of Microbiology, School of Genetics and Microbiology, Moyne Institute of Preventative Medicine, Trinity College Dublin, Dublin, Ireland
| | - Amy O'Callaghan
- Department of Microbiology, School of Genetics and Microbiology, Moyne Institute of Preventative Medicine, Trinity College Dublin, Dublin, Ireland
| | - Sinéad C Corr
- Department of Microbiology, School of Genetics and Microbiology, Moyne Institute of Preventative Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
380
|
Cui G, Li Z, Ren J, Yuan A. IL-33 in the tumor microenvironment is associated with the accumulation of FoxP3-positive regulatory T cells in human esophageal carcinomas. Virchows Arch 2019; 475:579-586. [DOI: 10.1007/s00428-019-02579-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/07/2019] [Accepted: 04/18/2019] [Indexed: 12/13/2022]
|
381
|
You J, Lin J, Zhou YF, Peng XD, He H, Li C, Zhu GQ, Zhao XQ, Zhao GQ. Role of the IL-33/ST2/p38 signaling pathway in the immune response of corneal epithelial cells to Aspergillus fumigatus infection. Int J Ophthalmol 2019; 12:549-556. [PMID: 31024805 DOI: 10.18240/ijo.2019.04.04] [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: 12/05/2018] [Accepted: 03/05/2019] [Indexed: 12/11/2022] Open
Abstract
AIM To investigate the expression of interleukin (IL)-33 in the cornea and human corneal epithelial cells (HCECs) exposed to Aspergillus fumigatus (A. fumigatus), and to determine the function of IL-33/ST2/p38 signaling pathway in the immune response of corneal epithelial cells to A. fumigatus infection. METHODS The mRNA and protein expression of IL-33 in HCECs and mice corneas were examined by quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) and Western blot analysis, respectively. IL-33 expression was also detected in cornea samples from healthy donors and patients with fungal keratitis with immunohistochemistry. The cultured HCECs were treated with inactive A. fumigatus hyphae at various concentrations with or without recombinant human IL-33 protein, soluble recombinant ST2 protein, specific ST2 neutralizing antibody, or the mitogen-activated protein kinase (MAPK) p38 inhibitor SB203580 for evaluation of the expression and activation of IL-33/ST2/p38 signaling in the regulation of proinflammatory cytokines. The production levels of IL-6 and IL-1β were determined by qRT-PCR and enzyme-linked immunosorbent assay (ELISA). The proliferation of HCECs was determined by a Cell Counting Kit-8 (CCK8) assay and cell count. RESULTS IL-33 expression levels increased in the corneal tissues of patients with fungal keratitis and in mice corneas of experimental A. fumigatus infection, as well as in HCECs with infection of A. fumigatus. A. fumigatus strongly stimulated HCECs-generated proinflammatory cytokine (IL-6 and IL-1β) production at both the mRNA and protein levels. This production of pro-inflammatory mediators stimulated by A. fumigatus was further stimulated by IL-33 and was prevented by soluble ST2 protein or ST2 neutralizing antibody. Moreover, IL-33 naturally promoted the p38 phosphorylation induced by A. fumigatus, which was suppressed by soluble ST2 protein. The MAPK p38 inhibitor SB203580 also inhibited the A. fumigatus-induced proinflammatory cytokine production. IL-33 administration for 48h and 72h promoted the proliferation of HCECs, which was attenuated by treatment with soluble recombinant human ST2 protein. CONCLUSION A. fumigatus elevates IL-33 expression in human and mice corneas and HCECs. Thus, IL-33/ST2/p38 signaling may play an important role in amplifying the immune response of corneal epithelial cells to A. fumigatus infection. Besides, IL-33 promotes the cell proliferation of HCECs via its receptor ST2. These findings suggest a novel autocrine mechanism of amplification of the fungal-induced inflammatory response in the corneal epithelium, highlighting a potential therapeutic target for fungal keratitis.
Collapse
Affiliation(s)
- Jia You
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Jing Lin
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Yi-Fan Zhou
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Xu-Dong Peng
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Hong He
- Department of Clinical Laboratory, the Affiliated Hospital of Qingdao University, Qingdao266003, Shandong Province, China
| | - Cui Li
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Guo-Qiang Zhu
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Xue-Qi Zhao
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Gui-Qiu Zhao
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| |
Collapse
|
382
|
Gabryelska A, Kuna P, Antczak A, Białasiewicz P, Panek M. IL-33 Mediated Inflammation in Chronic Respiratory Diseases-Understanding the Role of the Member of IL-1 Superfamily. Front Immunol 2019; 10:692. [PMID: 31057533 PMCID: PMC6477074 DOI: 10.3389/fimmu.2019.00692] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 03/13/2019] [Indexed: 12/19/2022] Open
Abstract
Interleukin 33 (IL-33) is an alarmin cytokine from the IL-1 family. IL-33 is localized in the nucleus and acts there as a gene regulator. Following injury, stress or cell death, it is released from the nucleus, and exerts its pro-inflammatory biological functions via the transmembrane form of the ST2 receptor, which is present mainly as attached to immune cells. In recent years, IL-33 became a focus of many studies due to its possible role in inflammatory disorders. Among respiratory disorders, the contribution of IL-33 to the development of asthma, in particular, has been most identified. Increased level of IL-33 in lung epithelial cells and blood serum has been observed in asthma patients. The IL-33/ST2 interaction activated the Th2 mediated immune response and further production of many pro-inflammatory cytokines. Single nucleotide polymorphisms in the IL-33 gene cause a predisposition to the development of asthma. Similarly, in chronic pulmonary obstructive disease (COPD), both increased expression of IL-33 and the ST2 receptor has been observed. Interestingly, cigarette smoke, a key inducer of COPD, not only activates IL-33 production by epithelial and endothelial cells, but also induces the expression of IL-33 in peripheral blood mononuclear cells. Knowledge regarding its contribution in other respiratory disorders, such as obstructive sleep apnea, remains greatly limited. Recently it was shown that IL-33 is one of the inflammatory mediators by which levels in blood serum are increased in OSA patients, compared to healthy control patients. This mini review summarizes current knowledge on IL-33 involvement in chosen chronic respiratory disorders and proposes this interleukin as a possible link in the pathogenesis of these diseases.
Collapse
Affiliation(s)
- Agata Gabryelska
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, Łódz, Poland.,Department of Internal Medicine, Asthma and Allergy, Medical University of Lodz, Łódz, Poland
| | - Piotr Kuna
- Department of Internal Medicine, Asthma and Allergy, Medical University of Lodz, Łódz, Poland
| | - Adam Antczak
- Department of General and Oncological Pulmonology, Medical University of Lodz, Łódz, Poland
| | - Piotr Białasiewicz
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, Łódz, Poland
| | - Michał Panek
- Department of Internal Medicine, Asthma and Allergy, Medical University of Lodz, Łódz, Poland
| |
Collapse
|
383
|
Yin-Yang 1 transcription factor modulates ST2 expression during adverse cardiac remodeling post-myocardial infarction. J Mol Cell Cardiol 2019; 130:216-233. [PMID: 30998979 DOI: 10.1016/j.yjmcc.2019.04.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 03/19/2019] [Accepted: 04/11/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND The cardioprotective effects of metformin remain poorly defined. Interleukin (IL)-33/ST2L signaling is a novel cardioprotective pathway, which is antagonized by the soluble isoform sST2. No data exist about the regulation of ST2 expression. This study aimed to evaluate the pathophysiological implication of Yin-Yang 1 (Yy1) transcription factor in cardiac remodeling and the expression of the soluble ST2 isoform. METHODS AND RESULTS Myocardial infarction (MI) was induced in Wistar rats randomly receiving metformin or saline solution by permanent ligation of the left anterior coronary artery. In addition, a model of cardiomyocyte "biochemical strain" was used. Metformin administration improved post-MI cardiac remodeling, an effect that was associated with increased IL-33 and reduced sST2 levels in the myocardium. The anti-remodeling effects of metformin were also associated with a decrease in the transcription factor Yy1 intranuclear level and lower levels of phosphorylated HDAC4 within the cytoplasmic space. These effects were also observed in a cardiomyocyte biochemical strain model, where Yy1 silencing or HDAC4 inhibition blocked sST2 production in cardiomyocytes. Metformin blocked the HDAC4 phosphorylation induced by MI, preventing its export from the nucleus to the cytosol. The presence of dephosphorylated HDAC4 in the nucleus acted as a co-repressor of Yy1, repressing sST2 expression. CONCLUSION The transcription factor Yy1 regulates sST2 expression, and repression of Yy1 by metformin results in lower levels of sST2 that are associated with favorable myocardial remodeling. The manipulation of YY1 or its co-repressor HDAC4 emerge as new targets to modulate ST2/IL33 signaling and prevent adverse cardiac remodeling.
Collapse
|
384
|
Griesenauer B, Jiang H, Yang J, Zhang J, Ramadan AM, Egbosiuba J, Campa K, Paczesny S. ST2/MyD88 Deficiency Protects Mice against Acute Graft-versus-Host Disease and Spares Regulatory T Cells. THE JOURNAL OF IMMUNOLOGY 2019; 202:3053-3064. [PMID: 30979817 DOI: 10.4049/jimmunol.1800447] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 03/18/2019] [Indexed: 12/11/2022]
Abstract
Acute graft-versus-host disease (aGVHD) hinders the efficacy of allogeneic hematopoietic cell transplantation (HCT). Plasma levels of soluble membrane-bound ST2 (ST2) are elevated in human and murine aGVHD and correlated to type 1 T cells response. ST2 signals through the adapter protein MyD88. The role of MyD88 in T cells during aGVHD has yet to be elucidated. We found that knocking out MyD88 in the donor T cells protected against aGVHD independent of IL-1R and TLR4 signaling in two murine HCT models. This protection was entirely driven by MyD88-/- CD4 T cells. Transplanting donor MyD88-/- conventional T cells (Tcons) with wild-type (WT) or MyD88-/- regulatory T cells (Tregs) lowered aGVHD severity and mortality. Transcriptome analysis of sorted MyD88-/- CD4 T cells from the intestine 10 d post-HCT showed lower levels of Il1rl1 (gene of ST2), Ifng, Csf2, Stat5, Batf, and Jak2 Transplanting donor ST2-/- Tcons with WT or ST2-/- Tregs showed a similar phenotype with what we observed when using donor MyD88-/- Tcons. Decreased ST2 was confirmed at the protein level with less secretion of soluble ST2 and more expression of ST2 compared with WT T cells. Our data suggest that Treg suppression from lack of MyD88 signaling in donor Tcons during alloreactivity uses the ST2 but not the IL-1R or TLR4 pathways, and ST2 represents a potential aGVHD therapeutic target sparing Tregs.
Collapse
Affiliation(s)
| | - Hua Jiang
- Indiana University School of Medicine, Indianapolis, IN 46202
| | - Jinfeng Yang
- Indiana University School of Medicine, Indianapolis, IN 46202
| | - Jilu Zhang
- Indiana University School of Medicine, Indianapolis, IN 46202
| | | | - Jane Egbosiuba
- Indiana University School of Medicine, Indianapolis, IN 46202
| | - Khaled Campa
- Indiana University School of Medicine, Indianapolis, IN 46202
| | - Sophie Paczesny
- Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
385
|
Wang Z, Guhl S, Franke K, Artuc M, Zuberbier T, Babina M. IL-33 and MRGPRX2-Triggered Activation of Human Skin Mast Cells-Elimination of Receptor Expression on Chronic Exposure, but Reinforced Degranulation on Acute Priming. Cells 2019; 8:cells8040341. [PMID: 30979016 PMCID: PMC6523246 DOI: 10.3390/cells8040341] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/08/2019] [Accepted: 04/10/2019] [Indexed: 12/27/2022] Open
Abstract
Clinically relevant exocytosis of mast cell (MC) mediators can be triggered by high-affinity IgE receptor (FcεRI)-aggregation (allergic route) or by the so-called pseudo-allergic pathway elicited via MAS-related G protein-coupled receptor-X2 (MRGPRX2). The latter is activated by drugs and endogenous neuropeptides. We recently reported that FcεRI-triggered degranulation is attenuated when human skin mast cells are chronically exposed to IL-33. Here, we were interested in the regulation of the MRGPRX2-route. Chronic exposure of skin MCs to IL-33 basically eliminated the pseudo-allergic/neurogenic route as a result of massive MRGPRX2 reduction. This downregulation seemed to partially require c-Jun N-terminal Kinase (JNK), but not p38, the two kinases activated by IL-33 in skin MCs. Surprisingly, however, JNK had a positive effect on MRGPRX2 expression in the absence of IL-33. This was evidenced by Accell®-mediated JNK knockdown and JNK inhibition. In stark contrast to the dampening effect upon prolonged exposure, IL-33 was able to prime for increased degranulation by MRGPRX2 ligands when administered directly before stimulation. This supportive effect depended on p38, but not on JNK activity. Our data reinforce the concept that exposure length dictates whether IL-33 will enhance or attenuate secretion. IL-33 is, thus, the first factor to acutely enhance MRGPRX2-triggered degranulation. Finally, we reveal that p38, rarely associated with MC degranulation, can positively affect exocytosis in a context-dependent manner.
Collapse
Affiliation(s)
- Zhao Wang
- Department of Dermatology, Venerology and Allergy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany.
| | - Sven Guhl
- Department of Dermatology, Venerology and Allergy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany.
| | - Kristin Franke
- Department of Dermatology, Venerology and Allergy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany.
| | - Metin Artuc
- Department of Dermatology, Venerology and Allergy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany.
| | - Torsten Zuberbier
- Department of Dermatology, Venerology and Allergy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany.
| | - Magda Babina
- Department of Dermatology, Venerology and Allergy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
386
|
Abstract
Severe asthma defined as uncontrolled or refractory asthma despite adequate use of high dose of inhaled steroid and additional long acting bronchodilators is associated with a high risk of comorbidities, exacerbations and persistent asthma-related symptoms. It remains a significant health care problem and represents the majority of health costs due to asthma. A better understanding of the basic mechanisms of the disease has allowed identification of new phenotypes and endotypes and of some predictive biomarkers. In the meantime an increasing number of promising biologicals are commercialized or on development providing new hopes to achieve asthma control and decrease exacerbation rate without the use of systemic corticosteroid. The increasing number of highly expensive available molecules poses physicians a new challenge: the identification of "the good treatment for the good patient". This article discuss the different biological available or in development in the field of severe asthma based on their mechanism of action and target. One of the aims is to help clarify the clinical decision-making process taking in account both the phenotype/endotype of the patient and the characteristics of these new drugs.
Collapse
|
387
|
Yang J, Ramadan A, Reichenbach DK, Loschi M, Zhang J, Griesenauer B, Liu H, Hippen KL, Blazar BR, Paczesny S. Rorc restrains the potency of ST2+ regulatory T cells in ameliorating intestinal graft-versus-host disease. JCI Insight 2019; 4:122014. [PMID: 30694220 DOI: 10.1172/jci.insight.122014] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 01/25/2019] [Indexed: 01/19/2023] Open
Abstract
Soluble stimulation-2 (ST2) is increased during graft-versus-host disease (GVHD), while Tregs that express ST2 prevent GVHD through unknown mechanisms. Transplantation of Foxp3- T cells and Tregs that were collected and sorted from different Foxp3 reporter mice indicated that in mice that developed GVHD, ST2+ Tregs were thymus derived and predominantly localized to the intestine. ST2-/- Treg transplantation was associated with reduced total intestinal Treg frequency and activation. ST2-/- versus WT intestinal Treg transcriptomes showed decreased Treg functional markers and, reciprocally, increased Rorc expression. Rorc-/- T cells transplantation enhanced the frequency and function of intestinal ST2+ Tregs and reduced GVHD through decreased gut-infiltrating soluble ST2-producing type 1 and increased IL-4/IL-10-producing type 2 T cells. Cotransfer of ST2+ Tregs sorted from Rorc-/- mice with WT CD25-depleted T cells decreased GVHD severity and mortality, increased intestinal ST2+KLRG1+ Tregs, and decreased type 1 T cells after transplantation, indicating an intrinsic mechanism. Ex vivo IL-33-stimulated Tregs (TregIL-33) expressed higher amphiregulin and displayed better immunosuppression, and adoptive transfer prevented GVHD better than control Tregs or TregIL-33 cultured with IL-23/IL-17. Amphiregulin blockade by neutralizing antibody in vivo abolished the protective effect of TregIL-33. Our data show that inverse expression of ST2 and RORγt in intestinal Tregs determines GVHD and that TregIL-33 has potential as a cellular therapy avenue for preventing GVHD.
Collapse
Affiliation(s)
- Jinfeng Yang
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Abdulraouf Ramadan
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Dawn K Reichenbach
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Michael Loschi
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jilu Zhang
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Brad Griesenauer
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Hong Liu
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Keli L Hippen
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Bruce R Blazar
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sophie Paczesny
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
388
|
Chan BCL, Lam CWK, Tam LS, Wong CK. IL33: Roles in Allergic Inflammation and Therapeutic Perspectives. Front Immunol 2019; 10:364. [PMID: 30886621 PMCID: PMC6409346 DOI: 10.3389/fimmu.2019.00364] [Citation(s) in RCA: 202] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 02/12/2019] [Indexed: 12/29/2022] Open
Abstract
Interleukin (IL)-33 belongs to IL-1 cytokine family which is constitutively produced from the structural and lining cells including fibroblasts, endothelial cells, and epithelial cells of skin, gastrointestinal tract, and lungs that are exposed to the environment. Different from most cytokines that are actively secreted from cells, nuclear cytokine IL-33 is passively released during cell necrosis or when tissues are damaged, suggesting that it may function as an alarmin that alerts the immune system after endothelial or epithelial cell damage during infection, physical stress, or trauma. IL-33 plays important roles in type-2 innate immunity via activation of allergic inflammation-related eosinophils, basophils, mast cells, macrophages, and group 2 innate lymphoid cells (ILC2s) through its receptor ST2. In this review, we focus on the recent advances of the underlying intercellular and intracellular mechanisms by which IL-33 can regulate the allergic inflammation in various allergic diseases including allergic asthma and atopic dermatitis. The future pharmacological strategy and application of traditional Chinese medicines targeting the IL-33/ST2 axis for anti-inflammatory therapy of allergic diseases were also discussed.
Collapse
Affiliation(s)
- Ben C L Chan
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Christopher W K Lam
- State Key Laboratory of Quality Research in Chinese Medicines, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau
| | - Lai-Shan Tam
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Chun K Wong
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.,Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
389
|
Paczesny S, Metzger J. Clinical Proteomics for Post-Hematopoeitic Stem Cell Transplantation Outcomes. Proteomics Clin Appl 2019; 13:e1800145. [PMID: 30307119 PMCID: PMC6440827 DOI: 10.1002/prca.201800145] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 09/28/2018] [Indexed: 12/20/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is the most effective form of tumor immunotherapy available to date. However, while HSCT can induce beneficial graft-versus-leukemia (GVL) effect, the adverse effect of graft-versus-host disease (GVHD), which is closely linked to GVL, is the major source of morbidity and mortality following HSCT. Until recently, available diagnostic and staging tools frequently fail to identify those at higher risk of disease progression or death. Furthermore, there are shortcomings in the prediction of the need for therapeutic interventions or the response rates to different forms of therapy. The past decade has been characterized by an explosive evolution of proteomics technologies, largely due to important advances in high-throughput MS instruments and bioinformatics. Building on these opportunities, blood biomarkers have been identified and validated both as promising diagnostic tools, prognostic tools that risk-stratify patients before future occurrence of GVHD and as predictive tools for responsiveness to GVHD therapy and non-relapse mortality. These biomarkers might facilitate timely and selective therapeutic intervention. This review summarizes current information on clinical proteomics for GVHD as well as other complications following HSCT. Finally, it proposes future directions for the translation of clinical proteomics to discovery of new potential therapeutic targets to the development of drugs.
Collapse
Affiliation(s)
- Sophie Paczesny
- Department of Pediatrics, Department of Microbiology Immunology, and Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | |
Collapse
|
390
|
Abe K, Takahashi A, Fujita M, Hayashi M, Okai K, Nozawa Y, Ohira H. Interleukin-33/ST2-Mediated Inflammation Plays a Critical Role in the Pathogenesis and Severity of Type I Autoimmune Hepatitis. Hepatol Commun 2019; 3:670-684. [PMID: 31061955 PMCID: PMC6492473 DOI: 10.1002/hep4.1326] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 01/22/2019] [Indexed: 12/11/2022] Open
Abstract
Interleukin (IL)‐33 was recently described as a new member of the IL‐1 family; members of this family have proinflammatory activity. IL‐33 and its soluble receptor ST2 (sST2) have been implicated in the pathogenesis of autoimmune diseases. This study investigated serum IL‐33 and sST2 in type I autoimmune hepatitis (AIH) and the relationship of these molecules with clinical and pathologic parameters. Subjects included 65 patients with AIH who were diagnosed in our hospital. The control population included 17 healthy individuals and 36 patients with primary biliary cholangitis (PBC). Mean age at AIH diagnosis was 55.5 years, and the male‐to‐female ratio was 6:59. Serum IL‐33 and sST2 levels were significantly higher in patients with AIH than in those with PBC or controls. Importantly, immunohistochemistry revealed high IL‐33 expression in liver sections from patients with AIH. In particular, serum IL‐33 and sST2 levels were significantly higher in acute‐onset AIH than in chronic‐onset AIH. Serum IL‐33 levels were positively correlated with serum total bilirubin (TB), alanine aminotransferase (ALT), and necroinflammatory activity in AIH. We performed multivariate logistic regression analysis and found serum IL‐33 levels to be independent factors for severe activity. Serum sST2 levels were positively correlated with serum TB and ALT and negatively correlated with serum albumin and prothrombin time in AIH. In particular, serum sST2 levels were significantly higher in severe symptoms of AIH. Serum IL‐33 and sST2 levels in patients with AIH responsive to treatment with prednisolone were significantly decreased after treatment. Interestingly, serum IL‐33 level was associated with a significantly increased risk of relapse. Conclusion: IL‐33/ST2 may play an important role in the pathogenesis and severity of AIH and may be a promising target for AIH therapy. This study aimed to investigate serum IL‐33 and sST2 in type I autoimmune hepatitis (AIH) patients and its relationship with clinical and pathological parameters. Multivariate analysis was performed, and serum IL‐33 levels were independent factors for severe activity and relapse. Our findings suggest that IL‐33/ST2 may play an important role in the pathogenesis and severity of AIH and may present a promising target for AIH therapy.
Collapse
Affiliation(s)
- Kazumichi Abe
- Department of Gastroenterology Fukushima Medical University School of Medicine Fukushima Japan
| | - Atsushi Takahashi
- Department of Gastroenterology Fukushima Medical University School of Medicine Fukushima Japan
| | - Masashi Fujita
- Department of Gastroenterology Fukushima Medical University School of Medicine Fukushima Japan
| | - Manabu Hayashi
- Department of Gastroenterology Fukushima Medical University School of Medicine Fukushima Japan
| | - Ken Okai
- Department of Gastroenterology Fukushima Medical University School of Medicine Fukushima Japan
| | - Yoshihiro Nozawa
- Department of Pathology Shirakawa Kousei General Hospital Shirakawa Japan
| | - Hiromasa Ohira
- Department of Gastroenterology Fukushima Medical University School of Medicine Fukushima Japan
| |
Collapse
|
391
|
Piper C, Drobyski WR. Inflammatory Cytokine Networks in Gastrointestinal Tract Graft vs. Host Disease. Front Immunol 2019; 10:163. [PMID: 30853956 PMCID: PMC6395399 DOI: 10.3389/fimmu.2019.00163] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 01/18/2019] [Indexed: 11/23/2022] Open
Abstract
Graft vs. host disease (GVHD) is the major non-relapse complication associated with allogeneic hematopoietic stem cell transplantation (HSCT). Damage to the gastrointestinal (GI) tract from acute GVHD is a particularly serious event that can result in significant morbidity and mortality. Proinflammatory cytokines play a critical role in the pathophysiology of intestinal GVHD, in part by activating donor T cell populations which subsequently induce tissue damage. In this review, we summarize pre-clinical data derived from experimental murine models that have examined the role of inflammatory cytokine pathways that play critical roles in the pathophysiology of GVHD of the GI tract. Specific areas of focus are on STAT 3-dependent cytokines (e.g., IL-6, IL-23, and IL-21), and members of the IL-1 cytokine family, both of which have been shown to induce pathological damage within the GI tract during this disease. We also review established and ongoing efforts to translate these pre-clinical findings into the clinic in an effort to reduce morbidity and mortality due to this complication.
Collapse
Affiliation(s)
- Clint Piper
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - William R Drobyski
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Medicine, Bone Marrow Transplant Program, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
392
|
Inhibition of the dipeptidyl peptidase DPP4 (CD26) reveals IL-33-dependent eosinophil-mediated control of tumor growth. Nat Immunol 2019; 20:257-264. [PMID: 30778250 DOI: 10.1038/s41590-019-0321-5] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 01/14/2019] [Indexed: 12/30/2022]
Abstract
Post-translational modification of chemokines mediated by the dipeptidyl peptidase DPP4 (CD26) has been shown to negatively regulate lymphocyte trafficking, and its inhibition enhances T cell migration and tumor immunity by preserving functional chemokine CXCL10. By extending those initial findings to pre-clinical models of hepatocellular carcinoma and breast cancer, we discovered a distinct mechanism by which inhibition of DPP4 improves anti-tumor responses. Administration of the DPP4 inhibitor sitagliptin resulted in higher concentrations of the chemokine CCL11 and increased migration of eosinophils into solid tumors. Enhanced tumor control was preserved in mice lacking lymphocytes and was ablated after depletion of eosinophils or treatment with degranulation inhibitors. We further demonstrated that tumor-cell expression of the alarmin IL-33 was necessary and sufficient for eosinophil-mediated anti-tumor responses and that this mechanism contributed to the efficacy of checkpoint-inhibitor therapy. These findings provide insight into IL-33- and eosinophil-mediated tumor control, revealed when endogenous mechanisms of DPP4 immunoregulation are inhibited.
Collapse
|
393
|
Wang Y, Richter L, Becker M, Amador C, Hyde RK. IL1RL1 is dynamically expressed on Cbfb-MYH11 + leukemia stem cells and promotes cell survival. Sci Rep 2019; 9:1729. [PMID: 30742053 PMCID: PMC6370767 DOI: 10.1038/s41598-018-38408-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 12/21/2018] [Indexed: 11/25/2022] Open
Abstract
Acute myeloid leukemia (AML) is often characterized by the presence of specific, recurrent chromosomal abnormalities. One of the most common aberrations, inversion of chromosome 16 [inv(16)], generates the fusion oncogene CBFB-MYH11. Previously, we used a mouse knock-in model to show that Cbfb-MYH11 induces changes in gene expression and results in the accumulation of abnormal myeloid cells, a subset of which are enriched for leukemia stem cell (LSC) activity. One gene upregulated by Cbfb-MYH11 encodes the cytokine receptor IL1RL1 (ST2). IL1RL1 and its ligand IL-33 are known regulators of mature myeloid cells, but their roles in AML are not known. Here, we use Cbfb-MYH11 knock-in mice to show that IL1RL1 is expressed by cell populations with high LSC activity, and that the cell surface expression of IL1RL1 is dynamic, implying that the expression of IL1RL1 is not restricted to a specific stage of differentiation. We also show that treatment with IL-33 increased serial replating ability and expression of pro-survival proteins in vitro. Finally, we show that IL1RL1+ cells can survive chemotherapy better than IL1RL1− cells in vivo. Collectively, our results indicate that IL1RL1 is dynamically expressed in Cbfb-MYH11+ leukemia cells and promotes their survival.
Collapse
Affiliation(s)
- Yiqian Wang
- Department of Biochemistry and Molecular Biology, and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| | - Lisa Richter
- Department of Biochemistry and Molecular Biology, and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| | - Michelle Becker
- Department of Biochemistry and Molecular Biology, and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| | - Catalina Amador
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - R Katherine Hyde
- Department of Biochemistry and Molecular Biology, and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States.
| |
Collapse
|
394
|
Lindahl G, Abrahamsson A, Dabrosin C. Dietary flaxseed and tamoxifen affect the inflammatory microenvironment in vivo in normal human breast tissue of postmenopausal women. Eur J Clin Nutr 2019; 73:1250-1259. [DOI: 10.1038/s41430-019-0396-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/18/2018] [Accepted: 12/23/2018] [Indexed: 12/27/2022]
|
395
|
Cui G, Ren J, Xu G, Li Z, Zheng W, Yuan A. Cellular and clinicopathological features of the IL-33/ST2 axis in human esophageal squamous cell carcinomas. Cancer Cell Int 2018; 18:203. [PMID: 30559604 PMCID: PMC6290492 DOI: 10.1186/s12935-018-0700-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 12/06/2018] [Indexed: 12/14/2022] Open
Abstract
Background Emerging evidence has suggested that interleukin (IL)-33 and its primary functional receptor ST2 are involved in the pathogenesis of tumorigenesis. Methods Using immunohistochemistry (IHC) and double immunofluorescence staining, we characterized the cellular and clinicopathological features of the IL-33/ST2 axis in different compartments in human esophageal squamous cell carcinoma (ESCC) surgical specimens. Results IHC data revealed an increased expression of IL-33-immunoreactivity (IR) and ST2-IR located in both ESCC cells and tumor stromal cells; which were associated with advanced clinicopathological features such as TNM stages and node involvement. However, the Kaplan–Meier analysis showed that densities of neither IL-33 positive nor ST2 positive cells in both the ESCC mass and stroma were associated with the overall survival rate in patients with ESCC. Double immunofluorescence staining for cellular feature analysis demonstrated that these IL-33 positive and ST2 positive cells in ESCCs were with a high proliferation rate, and IL-33-IR was frequently co-expressed with ST2-IR in both ESCC and stromal cells. Conclusion Significant altered cellular features of the IL-33/ST2 axis in ESCCs were associated with advanced clinicopathological variables. The data suggest that the IL-33/ST2 axis might be involved in the progression of human ESCCs.
Collapse
Affiliation(s)
- Guanglin Cui
- 1Research Group of Gastrointestinal Diseases, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan China.,2Faculty of Health Science, Nord University, Campus Levanger, Levanger, Norway
| | - Jingli Ren
- 3Department of Pathology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan China
| | - Gang Xu
- 1Research Group of Gastrointestinal Diseases, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan China
| | - Zhenfeng Li
- 1Research Group of Gastrointestinal Diseases, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan China
| | - Wei Zheng
- 1Research Group of Gastrointestinal Diseases, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan China
| | - Aping Yuan
- 1Research Group of Gastrointestinal Diseases, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan China.,4Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| |
Collapse
|
396
|
Abstract
Sepsis was known to ancient Greeks since the time of great physician Hippocrates (460-377 BC) without exact information regarding its pathogenesis. With time and medical advances, it is now considered as a condition associated with organ dysfunction occurring in the presence of systemic infection as a result of dysregulation of the immune response. Still with this advancement, we are struggling for the development of target-based therapeutic approach for the management of sepsis. The advancement in understanding the immune system and its working has led to novel discoveries in the last 50 years, including different pattern recognition receptors. Inflammasomes are also part of these novel discoveries in the field of immunology which are <20 years old in terms of their first identification. They serve as important cytosolic pattern recognition receptors required for recognizing cytosolic pathogens, and their pathogen-associated molecular patterns play an important role in the pathogenesis of sepsis. The activation of both canonical and non-canonical inflammasome signaling pathways is involved in mounting a proinflammatory immune response via regulating the generation of IL-1β, IL-18, IL-33 cytokines and pyroptosis. In addition to pathogens and their pathogen-associated molecular patterns, death/damage-associated molecular patterns and other proinflammatory molecules involved in the pathogenesis of sepsis affect inflammasomes and vice versa. Thus, the present review is mainly focused on the inflammasomes, their role in the regulation of immune response associated with sepsis, and their targeting as a novel therapeutic approach.
Collapse
Affiliation(s)
- Vijay Kumar
- Children's Health Queensland Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, Brisbane, Australia,
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Australia,
| |
Collapse
|
397
|
Cui G, Yuan A, Pang Z, Zheng W, Li Z, Goll R. Contribution of IL-33 to the Pathogenesis of Colorectal Cancer. Front Oncol 2018; 8:561. [PMID: 30547011 PMCID: PMC6279916 DOI: 10.3389/fonc.2018.00561] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 11/12/2018] [Indexed: 12/13/2022] Open
Abstract
The development of colorectal cancer (CRC) is not only determined by transformed cells per se, but also by factors existing in their immune microenvironment. Accumulating scientific evidence has revealed that interleukin (IL)-33, an IL-1 family member, plays an essential role in the regulation of immune response and is relevant in CRC pathogenesis. Data from both human and experimental studies demonstrated that IL-33 inhibits host anti-tumor immunity, remodels tumor stroma and enhances angiogenesis, thereby promoting the development of CRC. These pro-tumor effects of IL-33 are mainly mediated by IL-33 receptor ST2 (also known as IL-1RL1). Based on those findings, it is currently hypothesized that the IL-33/ST2 pathway is a potential biomarker and therapeutic target for colorectal tumorigenesis. Herein, we summarize the recent discoveries in understanding the critical role of the IL-33/ST2 pathway in contributing to the pathogenesis of colorectal tumorigenesis and discuss its potential implications for the future development of effective anti-tumor strategies.
Collapse
Affiliation(s)
- Guanglin Cui
- Research Group of Gastrointestinal Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Faculty of Health Science, Nord University, Levanger, Norway
| | - Aping Yuan
- Research Group of Gastrointestinal Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhigang Pang
- Research Group of Gastrointestinal Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Zheng
- Research Group of Gastrointestinal Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenfeng Li
- Research Group of Gastrointestinal Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rasmus Goll
- Department of Gastroenterology, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
398
|
Larouche M, Gagné-Ouellet V, Boucher-Lafleur AM, Larose MC, Plante S, Madore AM, Laviolette M, Flamand N, Chakir J, Laprise C. Methylation profiles of IL33 and CCL26 in bronchial epithelial cells are associated with asthma. Epigenomics 2018; 10:1555-1568. [PMID: 30468398 DOI: 10.2217/epi-2018-0044] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
AIM This study aimed to characterize DNA methylation (DNA-me) in promoter region of IL33, IL1RL1 and CCL26 in asthma and their impacts on transcriptional activity in bronchial epithelial cells (BECs). PATIENTS & METHODS We performed bis-pyrosequencing, quantitative real-time PCR and sequencing in BECs from ten asthmatic and ten control individuals. RESULTS We detected lower DNA-me levels of IL33 and CCL26 in asthmatic than control BECs. No correlation was found between methylation and expression levels. Interestingly, carriers of a mutative allele in a haplotype within the promoter of IL33 had a lower IL33 DNA-me level and CCL26 gene expression correlated with eosinophil count. CONCLUSION These findings highlight the importance of investigating both epigenetic and genetic mechanisms in understanding the epithelial immune response in asthma.
Collapse
Affiliation(s)
- Miriam Larouche
- Département des sciences fondamentales, Université du Québec à Chicoutimi, Saguenay, QC G7H 2B1, Canada
| | - Valérie Gagné-Ouellet
- Département des sciences fondamentales, Université du Québec à Chicoutimi, Saguenay, QC G7H 2B1, Canada
| | | | - Marie-Chantal Larose
- Centre de recherche, Institut universitaire de cardiologie et de pneumologie de Québec, Département de médecine, Faculté de médecine, Université Laval, Québec City, QC G1V 4G5, Canada
| | - Sophie Plante
- Centre de recherche, Institut universitaire de cardiologie et de pneumologie de Québec, Département de médecine, Faculté de médecine, Université Laval, Québec City, QC G1V 4G5, Canada
| | - Anne-Marie Madore
- Département des sciences fondamentales, Université du Québec à Chicoutimi, Saguenay, QC G7H 2B1, Canada
| | - Michel Laviolette
- Centre de recherche, Institut universitaire de cardiologie et de pneumologie de Québec, Département de médecine, Faculté de médecine, Université Laval, Québec City, QC G1V 4G5, Canada
| | - Nicolas Flamand
- Centre de recherche, Institut universitaire de cardiologie et de pneumologie de Québec, Département de médecine, Faculté de médecine, Université Laval, Québec City, QC G1V 4G5, Canada
| | - Jamila Chakir
- Centre de recherche, Institut universitaire de cardiologie et de pneumologie de Québec, Département de médecine, Faculté de médecine, Université Laval, Québec City, QC G1V 4G5, Canada
| | - Catherine Laprise
- Département des sciences fondamentales, Université du Québec à Chicoutimi, Saguenay, QC G7H 2B1, Canada.,Centre intersectoriel en santé durable, Université du Québec à Chicoutimi, Saguenay, QC G7H 2B1, Canada
| |
Collapse
|
399
|
Niedzielska M, Israelsson E, Angermann B, Sidders BS, Clausen M, Catley M, Malhotra R, Dumont C. Differential gene expression in human tissue resident regulatory T cells from lung, colon, and blood. Oncotarget 2018; 9:36166-36184. [PMID: 30546835 PMCID: PMC6281418 DOI: 10.18632/oncotarget.26322] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 10/24/2018] [Indexed: 12/22/2022] Open
Abstract
As we learn more about how immune responses occur in situ, it is becoming clear that each organ/tissue is characterized with its own anatomy and microenvironment which may affect and even determine the outcome of the immune responses. With emerging data from animal studies showing that regulatory T cells infiltrating non-lymphoid tissues exhibit unique phenotypes and transcriptional signatures and display functions beyond their well-established suppressive roles, there is an urgent need to explore the function of tissue Treg cells in humans. Here we characterized the transcriptome of Treg residing at the human mucosal tissue obtained from the normal area of cancer resections and their peripheral blood counterparts, identifying human lung and colon tissue Treg signature genes and their upstream regulators. Pathway analysis highlighted potential differences in the cross-talk between tissue Treg cells and other non-immune tissue-specific cell types. For example, genes associated with wnt pathway were differentially regulated in lung Treg cells compared to blood or colon indicating a potential role for lung Treg cells in epithelium repair and regeneration. Moreover, we identified several non-coding RNAs specifically expressed by tissue-resident Tregs. These results provide a comprehensive view of lung and colon tissue Treg transcriptional landscape.
Collapse
Affiliation(s)
- Magdalena Niedzielska
- Bioscience, Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Elisabeth Israelsson
- Bioscience, Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Bastian Angermann
- Bioscience, Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Benjamin S Sidders
- Bioscience, Oncology, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Cambridge, UK
| | - Maryam Clausen
- Translational Genomics, Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Matthew Catley
- Bioscience, Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Rajneesh Malhotra
- Bioscience, Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Céline Dumont
- Bioscience, Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
400
|
Afferni C, Buccione C, Andreone S, Galdiero MR, Varricchi G, Marone G, Mattei F, Schiavoni G. The Pleiotropic Immunomodulatory Functions of IL-33 and Its Implications in Tumor Immunity. Front Immunol 2018; 9:2601. [PMID: 30483263 PMCID: PMC6242976 DOI: 10.3389/fimmu.2018.02601] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 10/22/2018] [Indexed: 12/14/2022] Open
Abstract
Interleukin-33 (IL-33) is a IL-1 family member of cytokines exerting pleiotropic activities. In the steady-state, IL-33 is expressed in the nucleus of epithelial, endothelial, and fibroblast-like cells acting as a nuclear protein. In response to tissue damage, infections or necrosis IL-33 is released in the extracellular space, where it functions as an alarmin for the immune system. Its specific receptor ST2 is expressed by a variety of immune cell types, resulting in the stimulation of a wide range of immune reactions. Recent evidences suggest that different IL-33 isoforms exist, in virtue of proteolytic cleavage or alternative mRNA splicing, with potentially different biological activity and functions. Although initially studied in the context of allergy, infection, and inflammation, over the past decade IL-33 has gained much attention in cancer immunology. Increasing evidences indicate that IL-33 may have opposing functions, promoting, or dampening tumor immunity, depending on the tumor type, site of expression, and local concentration. In this review we will cover the biological functions of IL-33 on various immune cell subsets (e.g., T cells, NK, Treg cells, ILC2, eosinophils, neutrophils, basophils, mast cells, DCs, and macrophages) that affect anti-tumor immune responses in experimental and clinical cancers. We will also discuss the possible implications of diverse IL-33 mutations and isoforms in the anti-tumor activity of the cytokine and as possible clinical biomarkers.
Collapse
Affiliation(s)
- Claudia Afferni
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Carla Buccione
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Sara Andreone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Maria Rosaria Galdiero
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, Naples, Italy.,Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore", National Research Council, Naples, Italy
| | - Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|