1
|
Liu G, Haw TJ, Starkey MR, Philp AM, Pavlidis S, Nalkurthi C, Nair PM, Gomez HM, Hanish I, Hsu AC, Hortle E, Pickles S, Rojas-Quintero J, Estepar RSJ, Marshall JE, Kim RY, Collison AM, Mattes J, Idrees S, Faiz A, Hansbro NG, Fukui R, Murakami Y, Cheng HS, Tan NS, Chotirmall SH, Horvat JC, Foster PS, Oliver BG, Polverino F, Ieni A, Monaco F, Caramori G, Sohal SS, Bracke KR, Wark PA, Adcock IM, Miyake K, Sin DD, Hansbro PM. TLR7 promotes smoke-induced experimental lung damage through the activity of mast cell tryptase. Nat Commun 2023; 14:7349. [PMID: 37963864 PMCID: PMC10646046 DOI: 10.1038/s41467-023-42913-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/25/2023] [Indexed: 11/16/2023] Open
Abstract
Toll-like receptor 7 (TLR7) is known for eliciting immunity against single-stranded RNA viruses, and is increased in both human and cigarette smoke (CS)-induced, experimental chronic obstructive pulmonary disease (COPD). Here we show that the severity of CS-induced emphysema and COPD is reduced in TLR7-deficient mice, while inhalation of imiquimod, a TLR7-agonist, induces emphysema without CS exposure. This imiquimod-induced emphysema is reduced in mice deficient in mast cell protease-6, or when wild-type mice are treated with the mast cell stabilizer, cromolyn. Furthermore, therapeutic treatment with anti-TLR7 monoclonal antibody suppresses CS-induced emphysema, experimental COPD and accumulation of pulmonary mast cells in mice. Lastly, TLR7 mRNA is increased in pre-existing datasets from patients with COPD, while TLR7+ mast cells are increased in COPD lungs and associated with severity of COPD. Our results thus support roles for TLR7 in mediating emphysema and COPD through mast cell activity, and may implicate TLR7 as a potential therapeutic target.
Collapse
Affiliation(s)
- Gang Liu
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Tatt Jhong Haw
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Malcolm R Starkey
- Depatrment of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Ashleigh M Philp
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Healthcare clinical campus, UNSW, Sydney, Australia
| | - Stelios Pavlidis
- The Airways Disease Section, National Heart & Lung Institute, Imperial College London, London, UK
| | - Christina Nalkurthi
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Prema M Nair
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Henry M Gomez
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Irwan Hanish
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Alan Cy Hsu
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Elinor Hortle
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Sophie Pickles
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | | | - Raul San Jose Estepar
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Jacqueline E Marshall
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Richard Y Kim
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, Australia
| | - Adam M Collison
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Joerg Mattes
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Sobia Idrees
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Alen Faiz
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Nicole G Hansbro
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Ryutaro Fukui
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minatoku, Tokyo, Japan
| | - Yusuke Murakami
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Musashino University, Nishitokyo-shi, Tokyo, Japan
| | - Hong Sheng Cheng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Sanjay H Chotirmall
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | - Jay C Horvat
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Paul S Foster
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Brian Gg Oliver
- Woolcock Institute of Medical Research, University of Sydney & School of Life Sciences, University of Technology, Sydney, Australia
| | | | - Antonio Ieni
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Section of Anatomic Pathology, Università di Messina, Messina, Italy
| | - Francesco Monaco
- Thoracic Surgery, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Gaetano Caramori
- Pneumologia, Dipartimento BIOMORF and Dipartimento di Medicina e Chirurgia, Universities of Messina and Parma, Messina, Italy
| | - Sukhwinder S Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, Australia
| | - Ken R Bracke
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Peter A Wark
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Ian M Adcock
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Healthcare clinical campus, UNSW, Sydney, Australia
| | - Kensuke Miyake
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minatoku, Tokyo, Japan
| | - Don D Sin
- The University of British Columbia Centre for Heart Lung Innovation, St Paul's Hospital & Respiratory Division, Dept of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia.
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia.
| |
Collapse
|
2
|
Cho KA, Cha JE, Kim J, Kim YH, Ryu KH, Woo SY. Mesenchymal Stem Cell-Derived Exosomes Attenuate TLR7-Mediated Mast Cell Activation. Tissue Eng Regen Med 2022; 19:117-129. [PMID: 34792754 PMCID: PMC8782981 DOI: 10.1007/s13770-021-00395-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 10/19/2022] Open
Abstract
BACKGROUND Mast cells are immune sentinels in the skin that respond to a wide range of pathological and environmental stimuli; they owe their function to the expression of Toll-like receptors (TLRs). We previously found that tonsil-derived mesenchymal stem cells (T-MSCs) were able to effectively attenuate TLR7-mediated skin inflammation in mice, which was accompanied by an increase in mast cell number. The present study investigated whether T-MSC extracellular vesicles, such as exosomes, are able to regulate mast cell activation in response to TLR7 stimulation. METHODS The HMC-1 human mast cell line was treated with a TLR7 agonist in the presence or absence of T-MSC exosomes, and the levels of expressed inflammatory cytokines were assessed. Additionally, mice were repeatedly injected with a TLR7 agonist with or without interval treatments with T-MSC exosomes and assessed dermal distribution of mast cells and related immune cells. RESULTS We showed that T-MSC exosomes containing microRNAs that target inflammatory cytokines significantly reduced the expression of inflammatory cytokines in TLR7 agonist-treated HMC-1 cells. In addition, T-MSC exosomes inhibited the increase in the number of both dermal mast cells and CD14-positive cells in TLR7 agonist-treated mice. CONCLUSION Our data suggest that T-MSC exosomes have regulatory effects on mast cell activation under inflammatory conditions, including TLR7 stimulation.
Collapse
Affiliation(s)
- Kyung-Ah Cho
- Department of Microbiology, College of Medicine, Ewha Womans University, 25, Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07804, Republic of Korea
| | - Je-Eun Cha
- Department of Microbiology, College of Medicine, Ewha Womans University, 25, Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07804, Republic of Korea
| | - Jungwoo Kim
- Department of Microbiology, College of Medicine, Ewha Womans University, 25, Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07804, Republic of Korea
| | - Yu-Hee Kim
- Advanced Biomedical Research Institute, Ewha Womans University Seoul Hospital, Gangseo-Gu, Seoul, 07804, Republic of Korea
| | - Kyung-Ha Ryu
- Department of Pediatrics, College of Medicine, Ewha Womans University, Seoul, 07804, Republic of Korea
| | - So-Youn Woo
- Department of Microbiology, College of Medicine, Ewha Womans University, 25, Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07804, Republic of Korea.
| |
Collapse
|
3
|
Ogawa Y, Kinoshita M, Kawamura T, Shimada S. Intracellular TLRs of Mast Cells in Innate and Acquired Immunity. Handb Exp Pharmacol 2022; 276:133-159. [PMID: 34505203 DOI: 10.1007/164_2021_540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Mast cells (MCs) distribute to interface tissues with environment, such as skin, airway, and gut mucosa, thereby functioning as the sentinel against invading allergens and pathogens. To respond to and exclude these external substances promptly, MCs possess granules containing inflammatory mediators, including heparin, proteases, tumor necrosis factor, and histamine, and produce these mediators as a consequence of degranulation within minutes of activation. As a delayed response to external substances, MCs de novo synthesize inflammatory mediators, such as cytokines and chemokines, by sensing pathogen- and damage-associated molecular patterns through their pattern recognition receptors, including Toll-like receptors (TLRs). A substantial number of studies have reported immune responses by MCs through surface TLR signaling, particularly TLR2 and TLR4. However, less attention has been paid to immune responses through nucleic acid-recognizing intracellular TLRs. Among intracellular TLRs, human and rodent MCs express TLR3, TLR7, and TLR9, but not TLR8. Some virus infections modulate intracellular TLR expression in MCs. MC-derived mediators, such as histamine, cysteinyl leukotrienes, LL-37, and the granulocyte-macrophage colony-stimulating factor, have also been reported to modulate intracellular TLR expression in an autocrine and/or paracrine fashion. Synthetic ligands for intracellular TLRs and some viruses are sensed by intracellular TLRs of MCs, leading to the production of inflammatory cytokines and chemokines including type I interferons. These MC responses initiate and facilitate innate responses and the subsequent recruitment of additional innate effector cells. MCs also associate with the regulation of adaptive immunity. In this overview, the expression of intracellular TLRs in MCs and the recognition of pathogens, including viruses, by intracellular TLRs in MCs were critically evaluated.
Collapse
Affiliation(s)
- Youichi Ogawa
- Department of Dermatology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan.
| | - Manao Kinoshita
- Department of Dermatology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Tatsuyoshi Kawamura
- Department of Dermatology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Shinji Shimada
- Department of Dermatology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|
4
|
Agier J, Brzezińska-Błaszczyk E, Witczak P, Kozłowska E, Żelechowska P. The impact of TLR7 agonist R848 treatment on mast cell phenotype and activity. Cell Immunol 2021; 359:104241. [PMID: 33158544 DOI: 10.1016/j.cellimm.2020.104241] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 01/21/2023]
Abstract
Bearing in mind that mast cell contribution to viral clearance is still not fully understood, in this study, we evaluated the effect of Toll-like receptor (TLR)7 viral single-stranded ribonucleic acid (ssRNA) mimic ligand, namely resiquimod (R)848, on mast cell phenotype and activity. We demonstrated that rat peritoneal mast cells exhibit surface and intracellular expression of ssRNA-specific TLR7 molecule, and that mimic ligand switches the self-expression of this receptor. We also detected other proteins associated with the cellular antiviral response: interferon-alpha receptor 1 (IFNAR1), interferon-gamma receptor 1 (IFNGR1), and major histocompatibility complex I (MHC I). Moreover, we showed that R848 caused the decrease of all molecule's expression after prolonged incubation. Interestingly, we found that R848 induced the increase of high-affinity IgE receptor (FcεRI) expression. Finally, we documented that TLR7 ligand-stimulated mast cells synthesize/release interferon (IFN)-α and -β, tumor necrosis factor (TNF), and chemokines CCL3, CXCL8, as well as pro-inflammatory lipid mediators. Our findings confirm that mast cells may respond to TLR7 ligand by altering their phenotype and synthesizing mediators and could serve as active participants in the antiviral immune response.
Collapse
Affiliation(s)
- Justyna Agier
- Department of Experimental Immunology, Medical University of Lodz, Pomorska 251, 92-213 Lodz, Poland.
| | - Ewa Brzezińska-Błaszczyk
- Department of Experimental Immunology, Medical University of Lodz, Pomorska 251, 92-213 Lodz, Poland
| | - Piotr Witczak
- Department of Experimental Immunology, Medical University of Lodz, Pomorska 251, 92-213 Lodz, Poland
| | - Elżbieta Kozłowska
- Department of Experimental Immunology, Medical University of Lodz, Pomorska 251, 92-213 Lodz, Poland
| | - Paulina Żelechowska
- Department of Experimental Immunology, Medical University of Lodz, Pomorska 251, 92-213 Lodz, Poland
| |
Collapse
|
5
|
Abstract
Mast cells (MCs) are well known for their role in allergic conditions. This cell can be activated by various types of secretagogues, ranging from a small chemical to a huge protein. Mast cell activation by secretagogues triggers the increase in intracellular calcium (iCa2+) concentration, granule trafficking, and exocytosis. Activated mast cells release their intra-granular pre-stored mediator or the newly synthesized mediator in the exocytosis process, in the form of degranulation or secretion. There are at least three types of exocytosis in mast cells, which are suggested to contribute to the release of different mediators, i.e.,, piecemeal, kiss-and-run, and compound exocytosis. The status of mast cells, i.e., activated or resting, is often determined by measuring the concentration of the released mediator such as histamine or β-hexosaminidase. This review summarizes several mast cell components that have been and are generally used as mast cell activation indicator, from the classical histamine and β-hexosaminidase measurement, to eicosanoid and granule trafficking observation. Basic principle of the component determination is also explained with their specified research application and purpose. The information will help to predict the experiment results with a certain study design.
Collapse
Affiliation(s)
- Muhammad Novrizal Abdi Sahid
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada , Yogyakarta, Indonesia.,Curcumin Research Center, Faculty of Pharmacy, Univeristas Gadjah Mada , Yogyakarta, Indonesia
| | - Takeshi Kiyoi
- Division of Analytical Bio-medicine, Advanced Research Support Center, Ehime University , Toon, Ehime, Japan
| |
Collapse
|
6
|
Hu K, Fu M, Wang J, Luo S, Barreto M, Singh R, Chowdhury T, Li M, Zhang M, Guan X, Xiao J, Hu Q. HSV-2 Infection of Human Genital Epithelial Cells Upregulates TLR9 Expression Through the SP1/JNK Signaling Pathway. Front Immunol 2020; 11:356. [PMID: 32194565 PMCID: PMC7065266 DOI: 10.3389/fimmu.2020.00356] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 02/13/2020] [Indexed: 12/24/2022] Open
Abstract
It is known that herpes simplex virus type 2 (HSV-2) triggers the activation of Toll-like receptor (TLR) 9 signaling pathway and the consequent production of antiviral cytokines in dendritic cells. However, the impact of HSV-2 infection on TLR9 expression and signaling in genital epithelial cells, the primary HSV-2 targets, has yet to be determined. In the current study, by using both human genital epithelial cell lines and primary genital epithelial cells as models, we found that HSV-2 infection enhances TLR9 expression at both mRNA and protein levels. Such enhancement is virus replication-dependent and CpG-independent, while the HSV-2-mediated upregulation of TLR9 does not activate TLR9 signaling pathway. Mechanistically, a SP1 binding site on TLR9 promoter appears to be essential for HSV-2-induced TLR9 transactivation. Upon HSV-2 infection, SP1 translocates from the cytoplasm to the nucleus, and consequently binds to TLR9 promoter. By using specific inhibitors, the JNK signaling pathway is shown to be involved in the HSV-2-induced TLR9 transactivation, while HSV-2 infection increases the phosphorylation but not the total level of JNK. In agreement, antagonism of JNK signaling pathway inhibits the HSV-2-induced SP1 nuclear translocation. Taken together, our study demonstrates that HSV-2 infection of human genital epithelial cells promotes TLR9 expression through SP1/JNK signaling pathway. Findings in this study provide insights into HSV-2-host interactions and potential targets for immune intervention.
Collapse
Affiliation(s)
- Kai Hu
- Institute for Infection and Immunity, St George's, University of London, London, United Kingdom
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Ming Fu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jun Wang
- Institute for Clinical Research Center, Wuhan Medical and Healthcare Center for Women and Children, Wuhan, China
| | - Sukun Luo
- Institute for Clinical Research Center, Wuhan Medical and Healthcare Center for Women and Children, Wuhan, China
| | - Mariana Barreto
- Institute for Infection and Immunity, St George's, University of London, London, United Kingdom
| | - Rubin Singh
- Institute for Infection and Immunity, St George's, University of London, London, United Kingdom
| | - Tasnim Chowdhury
- Institute for Infection and Immunity, St George's, University of London, London, United Kingdom
| | - Mei Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Mudan Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Xinmeng Guan
- Institute for Infection and Immunity, St George's, University of London, London, United Kingdom
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Juhua Xiao
- Department of Ultrasound, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, China
| | - Qinxue Hu
- Institute for Infection and Immunity, St George's, University of London, London, United Kingdom
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
7
|
The Response of Tissue Mast Cells to TLR3 Ligand Poly(I:C) Treatment. J Immunol Res 2020; 2020:2140694. [PMID: 32185237 PMCID: PMC7060451 DOI: 10.1155/2020/2140694] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/23/2020] [Accepted: 02/01/2020] [Indexed: 12/18/2022] Open
Abstract
Mast cells (MCs) are found mainly at the anatomical sites exposed to the external environment; thus, they are localized close to blood vessels, lymphatic vessels, and a multitude of immune cells. Moreover, those cells can recognize invading pathogens through a range of surface molecules known as pathogen recognition receptors (PRRs), mainly Toll-like receptors (TLRs). MCs are extensively engaged in the control and clearance of bacterial infections, but much less is known about their contribution to antiviral host response as well as pathomechanisms of virus-induced diseases. In the study, we employed in vivo differentiated mature tissue mast cells freshly isolated from rat peritoneal cavity. Here, we demonstrated that rat peritoneal mast cells (rPMCs) express viral dsRNA-specific TLR3 molecule (intracellularly and on the cell surface) as well as other proteins associated with cellular antiviral response: IRF3, type I and II IFN receptors, and MHC I. We found that exposure of rPMCs to viral dsRNA mimic, i.e., poly(I:C), induced transient upregulation of surface TLR3 (while temporarily decreased TLR3 intracellular expression), type II IFN receptor, and MHC I. TLR3 ligand-stimulated rPMCs did not degranulate but generated and/or released type I IFNs (IFN-α and IFNβ) as well as proinflammatory lipid mediators (cysLTs), cytokines (TNF, IL-1β), and chemokines (CCL3, CXCL8). We documented that rPMC priming with poly(I:C) did not affect FcεRI-dependent degranulation. However, their costimulation with TLR3 agonist and anti-IgE led to a significant increase in cysLT and TNF secretion. Our findings confirm that MCs may serve as active participants in the antiviral immune response. Presented data on modulated FcεRI-mediated MC secretion of mediators upon poly(I:C) treatment suggests that dsRNA-type virus infection could influence the severity of allergic reactions.
Collapse
|
8
|
Gaudino SJ, Kumar P. Cross-Talk Between Antigen Presenting Cells and T Cells Impacts Intestinal Homeostasis, Bacterial Infections, and Tumorigenesis. Front Immunol 2019; 10:360. [PMID: 30894857 PMCID: PMC6414782 DOI: 10.3389/fimmu.2019.00360] [Citation(s) in RCA: 246] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 02/12/2019] [Indexed: 11/21/2022] Open
Abstract
Innate immunity is maintained in part by antigen presenting cells (APCs) including dendritic cells, macrophages, and B cells. APCs interact with T cells to link innate and adaptive immune responses. By displaying bacterial and tumorigenic antigens on their surface via major histocompatibility complexes, APCs can directly influence the differentiation of T cells. Likewise, T cell activation, differentiation, and effector functions are modulated by APCs utilizing multiple mechanisms. The objective of this review is to describe how APCs interact with and influence the activation of T cells to maintain innate immunity during exposure to microbial infection and malignant cells. How bacteria and cancer cells take advantage of some of these interactions for their own benefit will also be discussed. While this review will cover a broad range of topics, a general focus will be held around pathogens, cancers, and interactions that typically occur within the gastrointestinal tract.
Collapse
Affiliation(s)
- Stephen J Gaudino
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, United States
| | - Pawan Kumar
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
9
|
Majima-Horiuchi H, Komine-Aizawa S, Karasaki-Suzuki M, Izumi Y, Aizawa S, Hayakawa S. Synergistic induction of interferon-γ by interleukin-2, interleukin-12 and poly(I:C) in a human natural killer cell line. Immunol Med 2018; 41:136-141. [DOI: 10.1080/25785826.2018.1531193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Affiliation(s)
- Hiroko Majima-Horiuchi
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
- Majima Clinic, Tokyo, Japan
| | - Shihoko Komine-Aizawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Miki Karasaki-Suzuki
- Division of Physiology, Department of Biomedical Sciences, Nihon University School of Medicine, Tokyo, Japan
| | - Yasuyuki Izumi
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Shin Aizawa
- Division of Anatomical Science, Department of Functional Morphology, Nihon University School of Medicine, Tokyo, Japan
| | - Satoshi Hayakawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
10
|
Cathelicidin LL-37 Affects Surface and Intracellular Toll-Like Receptor Expression in Tissue Mast Cells. J Immunol Res 2018; 2018:7357162. [PMID: 29670923 PMCID: PMC5836302 DOI: 10.1155/2018/7357162] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 11/26/2017] [Indexed: 12/30/2022] Open
Abstract
Undoubtedly, mast cells take part in host defense against microorganisms as they are numerous at the portal of infection, they release many proinflammatory and antimicrobial mediators, and they express pattern recognition receptors, such as TLRs. These receptors play a key role in recognition and binding molecules associated with microorganisms and molecules associated with damage. Cathelicidins exhibit direct antimicrobial activities against a broad spectrum of microbes by perturbing their cell membranes. Accumulating evidence suggests a role for these molecules in supporting cell activation. We examined the impact of human cathelicidin LL-37 on tissue mast cell TLR expression and distribution. Depending on context, we show that LL-37 stimulation resulted in minor to major effects on TLR2, TLR3, TLR4, TLR5, TLR7, and TLR9 expression. Confocal microscopy analysis showed that, upon stimulation, TLRs may translocate from the cell interior to the surface and conversely. FPR2 and EGFR inhibitors reduced the increase in expression of selected receptors. We also established that LL-37 acts as a powerful inducer of CCL3 and ROS generation. These results showed that in response to LL-37, mast cells enhance the capability to detect invading pathogens by modulation of TLR expression in what may be involved FPR2 or EGFR molecules.
Collapse
|
11
|
Expression of surface and intracellular Toll-like receptors by mature mast cells. Cent Eur J Immunol 2017; 41:333-338. [PMID: 28450795 PMCID: PMC5382879 DOI: 10.5114/ceji.2016.65131] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/22/2016] [Indexed: 12/13/2022] Open
Abstract
Nowadays, more and more data indicate that mast cells play an important role in host defense against pathogens. That is why it is essential to understand the expression of Toll-like receptors (TLRs) by mast cells, because these molecules play particularly significant role in initiation host defense against microorganisms as they recognize both wide range of microbial pathogen-associated molecular patterns (PAMPs) and various endogenous damage-associated molecular patterns (DAMPs) released in response to infection. Therefore, we examined the constitutive expression of both surface and endosomal TLRs in rat native fully mature tissue mast cells. By the use of qRT-PCR we found that these cells express mRNAs for TLR2, TLR3, TLR4, TLR5, TLR7, and TLR9. The expression of TLR3, TLR4, TLR5, TLR7, and TLR9 transcripts were low and comparable and only the expression of TLR2 transcript was significant. By the use of flow cytometry technique, we clearly documented that mast cells express TLR2, TLR4, and TLR5 on cell surface, while TLR3, TLR7, and TLR9 proteins are located both on the cell membrane and intracellularly. The highest expression was observed for TLR5 and the lowest for surface TLR7. These observations undoubtedly indicate that mature tissue mast cells have a broad set of TLR molecules, thus can recognize and bind bacterial, viral, and fungal PAMPs as well as various endogenous molecules generated in response to infection.
Collapse
|
12
|
Narusawa M, Inoue H, Sakamoto C, Matsumura Y, Takahashi A, Inoue T, Watanabe A, Miyamoto S, Miura Y, Hijikata Y, Tanaka Y, Inoue M, Takayama K, Okazaki T, Hasegawa M, Nakanishi Y, Tani K. TLR7 ligand augments GM-CSF-initiated antitumor immunity through activation of plasmacytoid dendritic cells. Cancer Immunol Res 2014; 2:568-80. [PMID: 24830413 DOI: 10.1158/2326-6066.cir-13-0143] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Vaccination with irradiated granulocyte macrophage colony-stimulating factor (GM-CSF)-transduced autologous tumor cells (GVAX) has been shown to induce therapeutic antitumor immunity. However, its effectiveness is limited. We therefore attempted to improve the antitumor effect by identifying little-known key pathways in GM-CSF-sensitized dendritic cells (GM-DC) in tumor-draining lymph nodes (TDLN). We initially confirmed that syngeneic mice subcutaneously injected with poorly immunogenic Lewis lung carcinoma (LLC) cells transduced with Sendai virus encoding GM-CSF (LLC/SeV/GM) remarkably rejected the tumor growth. Using cDNA microarrays, we found that expression levels of type I interferon (IFN)-related genes, predominantly expressed in plasmacytoid DCs (pDC), were significantly upregulated in TDLN-derived GM-DCs and focused on pDCs. Indeed, mouse experiments demonstrated that the effective induction of GM-CSF-induced antitumor immunity observed in immunocompetent mice treated with LLC/SeV/GM cells was significantly attenuated when pDC-depleted or IFNα receptor knockout (IFNAR(-/-)) mice were used. Importantly, in both LLC and CT26 colon cancer-bearing mice, the combinational use of imiquimod with autologous GVAX therapy overcame the refractoriness to GVAX monotherapy accompanied by tolerability. Mechanistically, mice treated with the combined vaccination displayed increased expression levels of CD86, CD9, and Siglec-H, which correlate with an antitumor phenotype, in pDCs, but decreased the ratio of CD4(+)CD25(+)FoxP3(+) regulatory T cells in TDLNs. Collectively, these findings indicate that the additional use of imiquimod to activate pDCs with type I IFN production, as a positive regulator of T-cell priming, could enhance the immunologic antitumor effects of GVAX therapy, shedding promising light on the understanding and treatment of GM-CSF-based cancer immunotherapy.
Collapse
Affiliation(s)
- Megumi Narusawa
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Hiroyuki Inoue
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, JapanAuthors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, JapanAuthors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Chika Sakamoto
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Yumiko Matsumura
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Atsushi Takahashi
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Tomoko Inoue
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Ayumi Watanabe
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Shohei Miyamoto
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Yoshie Miura
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Yasuki Hijikata
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Yoshihiro Tanaka
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Makoto Inoue
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Koichi Takayama
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Toshihiko Okazaki
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Mamoru Hasegawa
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Yoichi Nakanishi
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Kenzaburo Tani
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, JapanAuthors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| |
Collapse
|
13
|
Zhang A, Chi X, Luo G, Hei Z, Xia H, Luo C, Wang Y, Mao X, Xia Z. Mast cell stabilization alleviates acute lung injury after orthotopic autologous liver transplantation in rats by downregulating inflammation. PLoS One 2013; 8:e75262. [PMID: 24116032 PMCID: PMC3792971 DOI: 10.1371/journal.pone.0075262] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 08/12/2013] [Indexed: 12/16/2022] Open
Abstract
Background Acute lung injury (ALI) is one of the most severe complications after orthotopic liver transplantation. Amplified inflammatory response after transplantation contributes to the process of ALI, but the mechanism underlying inflammation activation is not completely understood. We have demonstrated that mast cell stabilization attenuated inflammation and ALI in a rodent intestine ischemia/reperfusion model. We hypothesized that upregulation of inflammation triggered by mast cell activation may be involve in ALI after liver transplantation. Methods Adult male Sprague–Dawley rats received orthotopic autologous liver transplantation (OALT) and were executed 4, 8, 16, and 24 h after OALT. The rats were pretreated with the mast cell stabilizers cromolyn sodium or ketotifen 15 min before OALT and executed 8 h after OALT. Lung tissues and arterial blood were collected to evaluate lung injury. β-hexosaminidase and mast cell tryptase levels were assessed to determine the activation of mast cells. Tumor necrosis factor α (TNF-α), interleukin (IL)-1β and IL-6 in serum and lung tissue were analyzed by enzyme-linked immunosorbent assay. Nuclear factor-kappa B (NF-κB) p65 translocation was assessed by Western blot. Results The rats that underwent OALT exhibited severe pulmonary damage with a high wet-to-dry ratio, low partial pressure of oxygen, and low precursor surfactant protein C levels, which corresponded to the significant elevation of pro-inflammatory cytokines, β-hexosaminidase, and tryptase levels in serum and lung tissues. The severity of ALI progressed and maximized 8 h after OALT. Mast cell stabilization significantly inhibited the activation of mast cells, downregulated pro-inflammatory cytokine levels and translocation of NF-κB, and attenuated OALT-induced ALI. Conclusions Mast cell activation amplified inflammation and played an important role in the process of post-OALT related ALI.
Collapse
Affiliation(s)
- Ailan Zhang
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou City, People's Republic of China
| | - Xinjin Chi
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou City, People's Republic of China
| | - Gangjian Luo
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou City, People's Republic of China
| | - Ziqing Hei
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou City, People's Republic of China
- * E-mail:
| | - Hua Xia
- Department of Anesthesiology, the Affiliated Hospital of Luzhou Medical College, Luzhou City, People's Republic of China
| | - Chenfang Luo
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou City, People's Republic of China
| | - Yanling Wang
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou City, People's Republic of China
| | - Xiaowen Mao
- Department of Anesthesiology, Research Centre of Heart, Brain, Hormone and Healthy Aging, University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Zhengyuan Xia
- Department of Anesthesiology, Research Centre of Heart, Brain, Hormone and Healthy Aging, University of Hong Kong, Hong Kong SAR, People's Republic of China
| |
Collapse
|
14
|
Andries O, Filette MD, De Smedt SC, Demeester J, Poucke MV, Peelman L, Sanders NN. Innate immune response and programmed cell death following carrier-mediated delivery of unmodified mRNA to respiratory cells. J Control Release 2013; 167:157-66. [DOI: 10.1016/j.jconrel.2013.01.033] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 01/14/2013] [Accepted: 01/30/2013] [Indexed: 12/12/2022]
|
15
|
Hernández JC, Stevenson M, Latz E, Urcuqui-Inchima S. HIV type 1 infection up-regulates TLR2 and TLR4 expression and function in vivo and in vitro. AIDS Res Hum Retroviruses 2012; 28:1313-28. [PMID: 22280204 DOI: 10.1089/aid.2011.0297] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Toll-like receptors (TLRs) play a critical role in innate immunity against pathogens. Their stimulation induces the activation of NF-κB, an important inducer of HIV-1 replication. In recent years, an increasing number of studies using several cells types from HIV-infected patients indicate that TLRs play a key role in regulating the expression of proinflammatory cytokines and viral pathogenesis. In the present study, the effect of HIV-1 stimulation of monocyte-derived macrophage (MDM) and peripheral blood mononuclear cell (PBMC) subpopulations from healthy donors on the expression and functions of TLR2 and TLR4 was examined. In addition, and to complete the in vitro study, the expression pattern of TLR2 and TLR4 in 49 HIV-1-infected patients, classified according to viral load and the use of HAART, was determined and compared with 25 healthy subjects. An increase of TLR expression and production of proinflammatory cytokines were observed in MDMs and PBMCs infected with HIV-1 in vitro and in response to TLR stimulation, compared to the mock. In addition, an association between TLR expression and up-regulation of CD80 in plasmacytoid dendritic cells (pDCs) was observed. The ex vivo analysis indicated increased expression of TLR2 and TLR4 in myeloid dendritic cells (mDCs), but only of TLR2 in monocytes obtained from HIV-1-infected patients, compared to healthy subjects. Remarkably, the expression was higher in cells from patients who do not use HAART. In monocytes, there was a positive correlation between both TLRs and viral load, but not CD4(+) T cell numbers. Together, our in vitro and ex vivo results suggest that TLR expression and function can be up-regulated in response to HIV-1 infection and could affect the inflammatory response. We propose that modulation of TLRs represents a mechanism to promote HIV-1 replication or AIDS progression in HIV-1-infected patients.
Collapse
Affiliation(s)
- Juan C. Hernández
- Grupo Inmunovirología, Sede de Investigación Universitaria, Universidad de Antioquia, Medellín, Colombia
- INFETARE, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellin, Colombia
| | - Mario Stevenson
- Developmental Center for AIDS Research, University of Miami, Miami, Florida
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital, University of Bonn, Bonn, Germany
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Sede de Investigación Universitaria, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
16
|
Gou Z, Liu R, Zhao G, Zheng M, Li P, Wang H, Zhu Y, Chen J, Wen J. Epigenetic modification of TLRs in leukocytes is associated with increased susceptibility to Salmonella enteritidis in chickens. PLoS One 2012; 7:e33627. [PMID: 22438967 PMCID: PMC3306431 DOI: 10.1371/journal.pone.0033627] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2011] [Accepted: 02/14/2012] [Indexed: 12/21/2022] Open
Abstract
Toll-like receptors (TLRs) signaling pathways are the first lines in defense against Salmonella enteritidis (S. enteritidis) infection but the molecular mechanism underlying susceptibility to S. enteritidis infection in chicken remains unclear. SPF chickens injected with S. enteritidis were partitioned into two groups, one consisted of those from Salmonella-susceptible chickens (died within 5 d after injection, n = 6), the other consisted of six Salmonella-resistant chickens that survived for 15 d after injection. The present study shows that the bacterial load in susceptible chickens was significantly higher than that in resistant chickens and TLR4, TLR2-1 and TLR21 expression was strongly diminished in the leukocytes of susceptible chickens compared with those of resistant chickens. The induction of expression of pro-inflammatory cytokine genes, IL-6 and IFN-β, was greatly enhanced in the resistant but not in susceptible chickens. Contrasting with the reduced expression of TLR genes, those of the zinc finger protein 493 (ZNF493) gene and Toll-interacting protein (TOLLIP) gene were enhanced in the susceptible chickens. Finally, the expression of TLR4 in peripheral blood mononuclear cells (PBMCs) infected in vitro with S. enteritidis increased significantly as a result of treatment with 5-Aza-2-deoxycytidine (5-Aza-dc) while either 5-Aza-dc or trichostatin A was effective in up-regulating the expression of TLR21 and TLR2-1. DNA methylation, in the predicted promoter region of TLR4 and TLR21 genes, and an exonic CpG island of the TLR2-1 gene was significantly higher in the susceptible chickens than in resistant chickens. Taken together, the results demonstrate that ZNF493-related epigenetic modification in leukocytes probably accounts for increased susceptibility to S. enteritidis in chickens by diminishing the expression and response of TLR4, TLR21 and TLR2-1.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jie Wen
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Haidian, Beijing, China
- * E-mail:
| |
Collapse
|
17
|
Bezemer GFG, Sagar S, van Bergenhenegouwen J, Georgiou NA, Garssen J, Kraneveld AD, Folkerts G. Dual role of Toll-like receptors in asthma and chronic obstructive pulmonary disease. Pharmacol Rev 2012; 64:337-58. [PMID: 22407613 DOI: 10.1124/pr.111.004622] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
During the last decade, significant research has been focused on Toll-like receptors (TLRs) in the pathogenesis of airway diseases. TLRs are pattern recognition receptors that play pivotal roles in the detection of and response to pathogens. Because of the involvement of TLRs in innate and adaptive immunity, these receptors are currently being exploited as possible targets for drug development. Asthma and chronic obstructive pulmonary disease (COPD) are chronic inflammatory airway diseases in which innate and adaptive immunity play an important role. To date, asthma is the most common chronic disease in children aged 5 years and older. COPD is prevalent amongst the elderly and is currently the fifth-leading cause of death worldwide with still-growing prevalence. Both of these inflammatory diseases result in shortness of breath, which is treated, often ineffectively, with bronchodilators and glucocorticosteroids. Symptomatic treatment approaches are similar for both diseases; however, the underlying immunological mechanisms differ greatly. There is a clear need for improved treatment specific for asthma and for COPD. This review provides an update on the role of TLRs in asthma and in COPD and discusses the merits and difficulties of targeting these proteins as novel treatment strategies for airway diseases. TLR agonist, TLR adjuvant, and TLR antagonist therapies could all be argued to be effective in airway disease management. Because of a possible dual role of TLRs in airway diseases with shared symptoms and risk factors but different immunological mechanisms, caution should be taken while designing pulmonary TLR-based therapies.
Collapse
Affiliation(s)
- Gillina F G Bezemer
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Functional gastrointestinal disorders (FGIDs) are common and currently defined by a symptom-based classification with no discernable pathology. In functional dyspepsia (FD), the duodenum is now implicated as a key area where symptoms originate.This is attributed to immune activation with increasing evidence indicating a role for duodenal eosinophilia. In irritable bowel syndrome (IBS), mastocytosis has been documented throughout the small and large intestine. Eosinophils and mast cells are an important link between innate and adaptive immunity, and are important in allergic type TH2 inflammation. Eosinophils may give rise to symptoms due to release of preformed cytokine proteins, which trigger neural excitation, muscle spasm, and pain. The close relationship of mast cells to nerves in IBS may similarly give rise to symptoms. Genetic studies also support of the role of innate immunity in FGIDs. The data supporting a prime role for eosinophils and mast cells in subsets of FD and IBS has become credible, and these data should be used to implement advances in diagnosis and therapeutic trials.
Collapse
|
19
|
Surface TLR2 and TLR4 expression on mature rat mast cells can be affected by some bacterial components and proinflammatory cytokines. Mediators Inflamm 2011; 2011:427473. [PMID: 21765618 PMCID: PMC3134222 DOI: 10.1155/2011/427473] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2011] [Accepted: 02/22/2011] [Indexed: 01/24/2023] Open
Abstract
The aim of our study was to determine whether some bacterial components as well as some proinflammatory cytokines can affect surface mast cell levels. By the use of flow cytometry technique, we documented that freshly isolated mature rat peritoneal mast cells do express surface TLR2 and TLR4 protein, but not CD14 molecules, and respond to stimulation with TLR2 and TLR4 ligands by cysteinyl leukotriene generation. The level of TLR2 protein is modulated by PGN and CCL5 treatment, but not by LPS, LAM, TNF, or IL-6. Surface mast cell TLR4 expression is affected by LPS, LAM, IL-6, and CCL5. Considering that TLR-mediated activation conditions not only engaged these cells in antibacterial defense and development of inflammation but also might influence allergic processes, our observations that surface TLR2 and TLR4 expression can be regulated both bacterial components and proinflammatory cytokines seem to be very intriguing and importance.
Collapse
|
20
|
MiR-146a inhibits oxidized low-density lipoprotein-induced lipid accumulation and inflammatory response via targeting toll-like receptor 4. FEBS Lett 2011; 585:854-60. [DOI: 10.1016/j.febslet.2011.02.009] [Citation(s) in RCA: 189] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 02/04/2011] [Accepted: 02/04/2011] [Indexed: 02/06/2023]
|
21
|
Chang ZL. Important aspects of Toll-like receptors, ligands and their signaling pathways. Inflamm Res 2010; 59:791-808. [PMID: 20593217 DOI: 10.1007/s00011-010-0208-2] [Citation(s) in RCA: 156] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 04/06/2010] [Accepted: 04/22/2010] [Indexed: 12/17/2022] Open
Abstract
Due to the rapid increase of new information on the multiple roles of Toll-like receptors (TLRs), this paper reviews several main properties of TLRs and their ligands and signaling pathways. The investigation of pathogen infections in knockout mice suggests that specific TLRs play a key role in the activation of immune responses. Although the investigation of TLR biology is just beginning, a number of important findings are emerging. This review focuses on the following seven aspects of this emerging field: (a) a history of TLR and ligand studies; (b) the molecular basis of recognition by TLRs: TLR structures, pathogen-associated molecular pattern binding sites, TLR locations and functional responses; (c) cell types in TLR expression; (d) an overview of TLRs and their ligands: expression and ligands of cell-surface TLRs and of intracellular TLRs; (e) TLR-signaling pathways; (f) discussion: TLRs control of innate and adaptive systems; the trafficking of intracellular TLRs to endolysosomes; investigation of TLRs in regulating microRNA; investigation of crystal structure of TLRs with ligand binding; incidence of infectious diseases associated with single nucleotide polymorphisms (SNPs) in TLR genes; risk of cancer related to SNPs in TLR genes; TLR-ligand mediated anti-cancer effects; and TLR-ligand induced chronic inflammation and tumorigenesis; and (g) conclusions.
Collapse
Affiliation(s)
- Z L Chang
- Institute of Biochemistry and Cell Biology, Shanghai Institute for Biological Science, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
22
|
Zhang H, Lin L, Yang H, Zhang Z, Yang X, Zhang L, He S. Induction of IL-13 production and upregulation of gene expression of protease activated receptors in P815 cells by IL-6. Cytokine 2010; 50:138-45. [PMID: 20189822 DOI: 10.1016/j.cyto.2010.02.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2009] [Revised: 12/22/2009] [Accepted: 02/02/2010] [Indexed: 01/08/2023]
Abstract
Interleukin (IL)-6 is a multifunctional cytokine which has been showed to induce up-regulated expression of Fc epsilon RI receptor and histamine production in mast cells. However, little is known of its effects on Th2 cytokine secretion and protease activated receptor (PAR) expression in mast cells. In the present study, we examined potential influence of IL-6 on IL-13, IL-4 and IL-10 release from P815 cells and PAR expression on P815 cells by using flow cytometry analysis, quantitative real-time PCR, ELISA and cellular activation of signaling ELISA (CASE) techniques. The results showed that IL-6 induced up to 1.8-fold increase in IL-13, but not IL-4 or IL-10 release from P815 cells, and FSLLRY-NH(2) did not affect IL-6 induced IL-13 release. Tryptase elicited 2.0-fold increase in IL-13 release from P815 cells, which can be inhibited by IL-6. IL-6 elicited the up-regulated expression of PAR-1, PAR-2, PAR-3 and PAR-4 mRNAs, but had little effects on expression of PAR proteins. U0126, PD98059 and LY204002 abolished IL-6 induced IL-13 release when they were preincubated with P815 cells, indicating ERK and Akt cell signaling pathways may be involved in the event. In conclusion, IL-6 can stimulate IL-13 release from mast cells through an ERK and Akt cell signaling pathway dependent, but PAR independent mechanism.
Collapse
Affiliation(s)
- Huiyun Zhang
- Department of Pathophysiology, Hainan Medical College, Haikou, Hainan, China.
| | | | | | | | | | | | | |
Collapse
|