201
|
McElroy WT, Tan Z, Ho G, Paliwal S, Li G, Seganish WM, Tulshian D, Tata J, Fischmann TO, Sondey C, Bian H, Bober L, Jackson J, Garlisi CG, Devito K, Fossetta J, Lundell D, Niu X. Potent and Selective Amidopyrazole Inhibitors of IRAK4 That Are Efficacious in a Rodent Model of Inflammation. ACS Med Chem Lett 2015; 6:677-82. [PMID: 26101573 DOI: 10.1021/acsmedchemlett.5b00106] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 05/12/2015] [Indexed: 12/26/2022] Open
Abstract
IRAK4 is a critical upstream kinase in the IL-1R/TLR signaling pathway. Inhibition of IRAK4 is hypothesized to be beneficial in the treatment of autoimmune related disorders. A screening campaign identified a pyrazole class of IRAK4 inhibitors that were determined by X-ray crystallography to exhibit an unusual binding mode. SAR efforts focused on the identification of a potent and selective inhibitor with good aqueous solubility and rodent pharmacokinetics. Pyrazole C-3 piperidines were well tolerated, with N-sulfonyl analogues generally having good rodent oral exposure but poor solubility. N-Alkyl piperidines exhibited excellent solubility and reduced exposure. Pyrazoles possessing N-1 pyridine and fluorophenyl substituents were among the most active. Piperazine 32 was a potent enzyme inhibitor with good cellular activity. Compound 32 reduced the in vivo production of proinflammatory cytokines and was orally efficacious in a mouse antibody induced arthritis disease model of inflammation.
Collapse
Affiliation(s)
- William T. McElroy
- Discovery Chemistry, ‡Structural Chemistry, §In Vitro Pharmacology, and ∥Respiratory and Immunology, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Zheng Tan
- Discovery Chemistry, ‡Structural Chemistry, §In Vitro Pharmacology, and ∥Respiratory and Immunology, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Ginny Ho
- Discovery Chemistry, ‡Structural Chemistry, §In Vitro Pharmacology, and ∥Respiratory and Immunology, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Sunil Paliwal
- Discovery Chemistry, ‡Structural Chemistry, §In Vitro Pharmacology, and ∥Respiratory and Immunology, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Guoqing Li
- Discovery Chemistry, ‡Structural Chemistry, §In Vitro Pharmacology, and ∥Respiratory and Immunology, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - W. Michael Seganish
- Discovery Chemistry, ‡Structural Chemistry, §In Vitro Pharmacology, and ∥Respiratory and Immunology, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Deen Tulshian
- Discovery Chemistry, ‡Structural Chemistry, §In Vitro Pharmacology, and ∥Respiratory and Immunology, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - James Tata
- Discovery Chemistry, ‡Structural Chemistry, §In Vitro Pharmacology, and ∥Respiratory and Immunology, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Thierry O. Fischmann
- Discovery Chemistry, ‡Structural Chemistry, §In Vitro Pharmacology, and ∥Respiratory and Immunology, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Christopher Sondey
- Discovery Chemistry, ‡Structural Chemistry, §In Vitro Pharmacology, and ∥Respiratory and Immunology, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Hong Bian
- Discovery Chemistry, ‡Structural Chemistry, §In Vitro Pharmacology, and ∥Respiratory and Immunology, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Loretta Bober
- Discovery Chemistry, ‡Structural Chemistry, §In Vitro Pharmacology, and ∥Respiratory and Immunology, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - James Jackson
- Discovery Chemistry, ‡Structural Chemistry, §In Vitro Pharmacology, and ∥Respiratory and Immunology, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Charles G. Garlisi
- Discovery Chemistry, ‡Structural Chemistry, §In Vitro Pharmacology, and ∥Respiratory and Immunology, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Kristine Devito
- Discovery Chemistry, ‡Structural Chemistry, §In Vitro Pharmacology, and ∥Respiratory and Immunology, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - James Fossetta
- Discovery Chemistry, ‡Structural Chemistry, §In Vitro Pharmacology, and ∥Respiratory and Immunology, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Daniel Lundell
- Discovery Chemistry, ‡Structural Chemistry, §In Vitro Pharmacology, and ∥Respiratory and Immunology, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Xiaoda Niu
- Discovery Chemistry, ‡Structural Chemistry, §In Vitro Pharmacology, and ∥Respiratory and Immunology, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| |
Collapse
|
202
|
Seganish WM, McElroy WT, Herr RJ, Brumfield S, Greenlee WJ, Harding J, Komanduri V, Matasi J, Prakash KC, Tulshian D, Yang J, Yet L, Devito K, Fossetta J, Garlisi CG, Lundell D, Niu X, Sondey C. Initial optimization and series evolution of diaminopyrimidine inhibitors of interleukin-1 receptor associated kinase 4. Bioorg Med Chem Lett 2015; 25:3203-7. [PMID: 26115573 DOI: 10.1016/j.bmcl.2015.05.097] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 05/26/2015] [Accepted: 05/28/2015] [Indexed: 11/24/2022]
Abstract
IRAK4 plays a key role in TLR/IL-1 signaling. Previous efforts identified a series of aminopyrimidine IRAK4 inhibitors that possess good potency, but modest kinase selectivity. Exploration of substituents at the C-2 and C-5 positions generated compounds that maintained IRAK4 potency and improved kinase selectivity. Additionally, it was found that the pyrimidine core could be replaced with a pyridine and still retain potency and kinase selectivity. The optimization efforts led to compound 26 which had an IRAK4 IC50 of 0.7 nM, an IC50 of 55 nM on THP-1 cells stimulated with LPS, a TLR4 agonist, and greater than 100-fold selectivity versus 96% of a panel of 306 kinases.
Collapse
Affiliation(s)
- W Michael Seganish
- Discovery Chemistry, Merck Research Laboratories, 2015 Galloping Hill Rd., Kenilworth, NJ 07033, United States.
| | - William T McElroy
- Discovery Chemistry, Merck Research Laboratories, 2015 Galloping Hill Rd., Kenilworth, NJ 07033, United States
| | - R Jason Herr
- Medicinal Chemistry Department, Albany Molecular Research, Inc. (AMRI), 26 Corporate Circle, Albany, NY 12203, United States
| | - Stephanie Brumfield
- Discovery Chemistry, Merck Research Laboratories, 2015 Galloping Hill Rd., Kenilworth, NJ 07033, United States
| | - William J Greenlee
- Discovery Chemistry, Merck Research Laboratories, 2015 Galloping Hill Rd., Kenilworth, NJ 07033, United States
| | - James Harding
- Medicinal Chemistry Department, Albany Molecular Research, Inc. (AMRI), 26 Corporate Circle, Albany, NY 12203, United States
| | - Venukrishnan Komanduri
- Medicinal Chemistry Department, AMRI Singapore Research Centre, 61 Science Park Road, #05-01, The Galen, Science Park III, Singapore 117525, Singapore
| | - Julius Matasi
- Discovery Chemistry, Merck Research Laboratories, 2015 Galloping Hill Rd., Kenilworth, NJ 07033, United States
| | - Koraboina Chandra Prakash
- Medicinal Chemistry Department, AMRI Singapore Research Centre, 61 Science Park Road, #05-01, The Galen, Science Park III, Singapore 117525, Singapore
| | - Deen Tulshian
- Discovery Chemistry, Merck Research Laboratories, 2015 Galloping Hill Rd., Kenilworth, NJ 07033, United States
| | - Jinhai Yang
- Medicinal Chemistry Department, Albany Molecular Research, Inc. (AMRI), 26 Corporate Circle, Albany, NY 12203, United States
| | - Larry Yet
- Medicinal Chemistry Department, Albany Molecular Research, Inc. (AMRI), 26 Corporate Circle, Albany, NY 12203, United States
| | - Kristine Devito
- In Vitro Pharmacology, Merck Research Laboratories, 2015 Galloping Hill Rd., Kenilworth, NJ 07033, United States
| | - James Fossetta
- Respiratory and Immunology, Merck Research Laboratories, 2015 Galloping Hill Rd., Kenilworth, NJ 07033, United States
| | - Charles G Garlisi
- In Vitro Pharmacology, Merck Research Laboratories, 2015 Galloping Hill Rd., Kenilworth, NJ 07033, United States
| | - Daniel Lundell
- Respiratory and Immunology, Merck Research Laboratories, 2015 Galloping Hill Rd., Kenilworth, NJ 07033, United States
| | - Xiaoda Niu
- In Vitro Pharmacology, Merck Research Laboratories, 2015 Galloping Hill Rd., Kenilworth, NJ 07033, United States
| | - Christopher Sondey
- In Vitro Pharmacology, Merck Research Laboratories, 2015 Galloping Hill Rd., Kenilworth, NJ 07033, United States
| |
Collapse
|
203
|
Liu Y, Yin H, Zhao M, Lu Q. TLR2 and TLR4 in autoimmune diseases: a comprehensive review. Clin Rev Allergy Immunol 2015; 47:136-47. [PMID: 24352680 DOI: 10.1007/s12016-013-8402-y] [Citation(s) in RCA: 282] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Autoimmune diseases are immune disorders characterized by T cell hyperactivity and B cell overstimulation leading to overproduction of autoantibodies. Although the pathogenesis of various autoimmune diseases remains to be elucidated, environmental factors have been thought to contribute to the initiation and maintenance of auto-respond inflammation. Toll-like receptors (TLRs) are pattern recognition receptors belonging to innate immunity that recognize and defend invading microorganisms. Besides these exogenous pathogen-associated molecular patterns, TLRs can also bind with damage-associated molecular patterns produced under strike or by tissue damage or cells apoptosis. It is believed that TLRs build a bridge between innate immunity and autoimmunity. There are five adaptors to TLRs including MyD88, TRIF, TIRAP/MAL, TRAM, and SARM. Upon activation, TLRs recruit specific adaptors to initiate the downstream signaling pathways leading to the production of inflammatory cytokines and chemokines. Under certain circumstances, ligation of TLRs drives to aberrant activation and unrestricted inflammatory responses, thereby contributing to the perpetuation of inflammation in autoimmune diseases. In the past, most studies focused on the intracellular TLRs, such as TLR3, TLR7, and TLR9, but recent studies reveal that cell surface TLRs, especially TLR2 and TLR4, also play an essential role in the development of autoimmune diseases and afford multiple therapeutic targets. In this review, we summarized the biological characteristics, signaling mechanisms of TLR2/4, the negative regulators of TLR2/4 pathway, and the pivotal function of TLR2/4 in the pathogenesis of autoimmune diseases including rheumatoid arthritis, systemic lupus erythematosus, systemic sclerosis, Sjogren's syndrome, psoriasis, multiple sclerosis, and autoimmune diabetes.
Collapse
Affiliation(s)
- Yu Liu
- Department of Dermatology, Second Xiangya Hospital, Central South University, #139 Renmin Middle Rd, Changsha, Hunan, 410011, People's Republic of China
| | | | | | | |
Collapse
|
204
|
Hameed I, Masoodi SR, Mir SA, Nabi M, Ghazanfar K, Ganai BA. Type 2 diabetes mellitus: From a metabolic disorder to an inflammatory condition. World J Diabetes 2015; 6:598-612. [PMID: 25987957 PMCID: PMC4434080 DOI: 10.4239/wjd.v6.i4.598] [Citation(s) in RCA: 271] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 10/14/2014] [Accepted: 12/31/2014] [Indexed: 02/05/2023] Open
Abstract
Diabetes mellitus is increasing at an alarming rate and has become a global challenge. Insulin resistance in target tissues and a relative deficiency of insulin secretion from pancreatic β-cells are the major features of type 2 diabetes (T2D). Chronic low-grade inflammation in T2D has given an impetus to the field of immuno-metabolism linking inflammation to insulin resistance and β-cell dysfunction. Many factors advocate a causal link between metabolic stress and inflammation. Numerous cellular factors trigger inflammatory signalling cascades, and as a result T2D is at the moment considered an inflammatory disorder triggered by disordered metabolism. Cellular mechanisms like activation of Toll-like receptors, Endoplasmic Reticulum stress, and inflammasome activation are related to the nutrient excess linking pathogenesis and progression of T2D with inflammation. This paper aims to systematically review the metabolic profile and role of various inflammatory pathways in T2D by capturing relevant evidence from various sources. The perspectives include suggestions for the development of therapies involving the shift from metabolic stress to homeostasis that would favour insulin sensitivity and survival of pancreatic β-cells in T2D.
Collapse
|
205
|
Weber DJ, Gracon ASA, Ripsch MS, Fisher AJ, Cheon BM, Pandya PH, Vittal R, Capitano ML, Kim Y, Allette YM, Riley AA, McCarthy BP, Territo PR, Hutchins GD, Broxmeyer HE, Sandusky GE, White FA, Wilkes DS. The HMGB1-RAGE axis mediates traumatic brain injury-induced pulmonary dysfunction in lung transplantation. Sci Transl Med 2015; 6:252ra124. [PMID: 25186179 DOI: 10.1126/scitranslmed.3009443] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Traumatic brain injury (TBI) results in systemic inflammatory responses that affect the lung. This is especially critical in the setting of lung transplantation, where more than half of donor allografts are obtained postmortem from individuals with TBI. The mechanism by which TBI causes pulmonary dysfunction remains unclear but may involve the interaction of high-mobility group box-1 (HMGB1) protein with the receptor for advanced glycation end products (RAGE). To investigate the role of HMGB1 and RAGE in TBI-induced lung dysfunction, RAGE-sufficient (wild-type) or RAGE-deficient (RAGE(-/-)) C57BL/6 mice were subjected to TBI through controlled cortical impact and studied for cardiopulmonary injury. Compared to control animals, TBI induced systemic hypoxia, acute lung injury, pulmonary neutrophilia, and decreased compliance (a measure of the lungs' ability to expand), all of which were attenuated in RAGE(-/-) mice. Neutralizing systemic HMGB1 induced by TBI reversed hypoxia and improved lung compliance. Compared to wild-type donors, lungs from RAGE(-/-) TBI donors did not develop acute lung injury after transplantation. In a study of clinical transplantation, elevated systemic HMGB1 in donors correlated with impaired systemic oxygenation of the donor lung before transplantation and predicted impaired oxygenation after transplantation. These data suggest that the HMGB1-RAGE axis plays a role in the mechanism by which TBI induces lung dysfunction and that targeting this pathway before transplant may improve recipient outcomes after lung transplantation.
Collapse
Affiliation(s)
- Daniel J Weber
- Center for Immunobiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Adam S A Gracon
- Center for Immunobiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Matthew S Ripsch
- Department of Anesthesia, Paul and Carole Stark Neurosciences Research Institute, Indianapolis, IN 46202, USA
| | - Amanda J Fisher
- Center for Immunobiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Department of Anesthesia, Paul and Carole Stark Neurosciences Research Institute, Indianapolis, IN 46202, USA
| | - Bo M Cheon
- Department of Anesthesia, Paul and Carole Stark Neurosciences Research Institute, Indianapolis, IN 46202, USA
| | - Pankita H Pandya
- Center for Immunobiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Ragini Vittal
- Center for Immunobiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Maegan L Capitano
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Youngsong Kim
- Department of Anesthesia, Paul and Carole Stark Neurosciences Research Institute, Indianapolis, IN 46202, USA
| | - Yohance M Allette
- Department of Anesthesia, Paul and Carole Stark Neurosciences Research Institute, Indianapolis, IN 46202, USA
| | - Amanda A Riley
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Brian P McCarthy
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Paul R Territo
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Gary D Hutchins
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - George E Sandusky
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Fletcher A White
- Center for Immunobiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Department of Anesthesia, Paul and Carole Stark Neurosciences Research Institute, Indianapolis, IN 46202, USA
| | - David S Wilkes
- Center for Immunobiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
206
|
Lees S, Golub SB, Last K, Zeng W, Jackson DC, Sutton P, Fosang AJ. Bioactivity in an Aggrecan 32-mer Fragment Is Mediated via Toll-like Receptor 2. Arthritis Rheumatol 2015; 67:1240-9. [DOI: 10.1002/art.39063] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 02/03/2015] [Indexed: 01/27/2023]
Affiliation(s)
- Sophie Lees
- University of Melbourne, Murdoch Childrens Research Institute, and Royal Children's Hospital; Parkville Victoria Australia
| | - Suzanne B. Golub
- University of Melbourne, Murdoch Childrens Research Institute, and Royal Children's Hospital; Parkville Victoria Australia
| | - Karena Last
- University of Melbourne, Murdoch Childrens Research Institute, and Royal Children's Hospital; Parkville Victoria Australia
| | - Weiguang Zeng
- University of Melbourne; Parkville Victoria Australia
| | | | - Philip Sutton
- University of Melbourne, Murdoch Childrens Research Institute, and Royal Children's Hospital; Parkville Victoria Australia
| | - Amanda J. Fosang
- University of Melbourne, Murdoch Childrens Research Institute, and Royal Children's Hospital; Parkville Victoria Australia
| |
Collapse
|
207
|
Brough D, Denes A. Interleukin-1α and brain inflammation. IUBMB Life 2015; 67:323-30. [PMID: 25906979 DOI: 10.1002/iub.1377] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 03/16/2015] [Indexed: 12/12/2022]
Abstract
Acute brain injuries such as caused by stroke are amongst the leading causes of death and are the leading cause of disability. Despite this there are very limited therapeutic options, and new therapeutic strategies and targets are required. Inflammation is known to exacerbate brain injury and is now considered as a potential therapeutic target. The damaging inflammation that occurs after acute brain injury is driven by pro-inflammatory members of the interleukin (IL)-1 cytokine family, namely, IL-1α and IL-1β. Previous research efforts have focussed on the biology and contribution of IL-1β. However, we now recognise that IL-1α is an early and important mediator of inflammation after injury. This review focuses on what is known about IL-1α, its regulation and its contribution to brain injury. Inhibiting mechanisms regulating the processing and release of IL-1α may offer new therapeutic targets for the treatment of devastating acute brain injuries.
Collapse
Affiliation(s)
- David Brough
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - Adam Denes
- Faculty of Life Sciences, University of Manchester, Manchester, UK.,Laboratory of Molecular Neuroendocrinology, Institute of Experimental Medicine, Budapest, Hungary
| |
Collapse
|
208
|
Ilhan F, Kalkanli ST. Atherosclerosis and the role of immune cells. World J Clin Cases 2015; 3:345-352. [PMID: 25879006 PMCID: PMC4391004 DOI: 10.12998/wjcc.v3.i4.345] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 06/30/2014] [Accepted: 01/20/2015] [Indexed: 02/05/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease arising from lipids, specifically low-density lipoproteins, and leukocytes. Following the activation of endothelium with the expression of adhesion molecules and monocytes, inflammatory cytokines from macrophages, and plasmacytoid dendritic cells, high levels of interferon (IFN)-α and β are generated upon the activation of toll-like receptor-9, and T-cells, especially the ones with Th1 profile, produce pro-inflammatory mediators such as IFN-γ and upregulate macrophages to adhere to the endothelium and migrate into the intima. This review presents an exhaustive account for the role of immune cells in the atherosclerosis.
Collapse
|
209
|
Carlsen TG, Kjærsgaard P, Jørgensen TL, Foldbjerg R, Nielsen ML, Poulsen TBG, Zabieglo K, Christiansen G, Birkelund S. Interleukin-1α activation and localization in lipopolysaccharide-stimulated human monocytes and macrophages. J Immunol Methods 2015; 422:59-71. [PMID: 25870118 DOI: 10.1016/j.jim.2015.03.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 03/31/2015] [Accepted: 03/31/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND Interleukin-1α (IL-1α) is a proinflammatory cytokine belonging to the IL-1 family. It is synthesized as a 33kDa precursor peptide that is cleaved by a calpain-like protease to a 16 kDa propiece and a 17 kDa mature IL-1α peptide. In contrast to its close relative, IL-1β, the role of IL-1α in inflammation is only partly understood. RESULTS Human monocyte derived macrophages, stimulated with lipopolysaccharide (LPS) were analysed for production and localization of IL-1α by use of a monoclonal antibody (MAb) generated against recombinant precursor IL-1α. We found that the MAb detected IL-1α within the nuclei of the cells 2h (hours) after LPS stimulation and production continued for up to 20 h. At no time could we demonstrate cleavage of the IL-1α precursor. The MAb was conjugated to fluorescein isothiocyanate (FITC) for use in flow cytometry. Based on the flow cytometric analysis CD68 positive cells were positive for IL-1α in agreement with CD68 being a marker for monocytes. CONCLUSIONS Here, we demonstrate, for the first time, a method to visualize and measure the production of IL-1α in both human monocytes and macrophages.
Collapse
Affiliation(s)
- Thomas Gelsing Carlsen
- Department of Health Science and Technology, Fredrik Bajers Vej 3b, 9220 Aalborg Ø, Denmark.
| | - Pernille Kjærsgaard
- Department of Health Science and Technology, Fredrik Bajers Vej 3b, 9220 Aalborg Ø, Denmark.
| | - Trine Lykke Jørgensen
- Department of Health Science and Technology, Fredrik Bajers Vej 3b, 9220 Aalborg Ø, Denmark.
| | - Rasmus Foldbjerg
- Department of Health Science and Technology, Fredrik Bajers Vej 3b, 9220 Aalborg Ø, Denmark.
| | - Mads Lausen Nielsen
- Department of Health Science and Technology, Fredrik Bajers Vej 3b, 9220 Aalborg Ø, Denmark.
| | | | | | | | - Svend Birkelund
- Department of Health Science and Technology, Fredrik Bajers Vej 3b, 9220 Aalborg Ø, Denmark; Loke Diagnostics, Sindalsvej 17, 8240 Risskov, Denmark.
| |
Collapse
|
210
|
De Nardo D. Toll-like receptors: Activation, signalling and transcriptional modulation. Cytokine 2015; 74:181-9. [PMID: 25846205 DOI: 10.1016/j.cyto.2015.02.025] [Citation(s) in RCA: 309] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 02/26/2015] [Indexed: 02/06/2023]
Abstract
Families of innate immune receptors serve as the bodies primary defence system by recognising and rapidly responding to infection by microorganisms or to endogenous danger signals and initiating inflammatory processes. Whilst Toll-like receptors (TLRs) were the first family to be discovered, important and exciting discoveries continue to emerge into the molecular mechanisms that control their activation and regulation. Herein, I will provide an overview of TLR activation and their downstream signalling cascades, and discuss some of the recent findings concerning the assembly of a TLR oligomeric signalling platform, known as the Myddosome. Further, a brief examination of the importance of crosstalk between multiple TLRs or between TLRs and other innate immune receptors for appropriate and coordinated immune responses will be presented. Finally, I will discuss the importance of mechanisms that regulate TLRs with a focus on the role of activating transcription factor 3 (ATF3) in modulating transcriptional responses downstream of TLRs.
Collapse
Affiliation(s)
- Dominic De Nardo
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3050, Australia.
| |
Collapse
|
211
|
Discovery and hit-to-lead optimization of 2,6-diaminopyrimidine inhibitors of interleukin-1 receptor-associated kinase 4. Bioorg Med Chem Lett 2015; 25:1836-41. [PMID: 25870132 DOI: 10.1016/j.bmcl.2015.03.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 03/16/2015] [Accepted: 03/18/2015] [Indexed: 01/16/2023]
Abstract
Interleukin receptor-associated kinase 4 (IRAK4) is a critical element of the Toll-like/interleukin-1 receptor inflammation signaling pathway. A screening campaign identified a novel diaminopyrimidine hit that exhibits weak IRAK4 inhibitory activity and a ligand efficiency of 0.25. Hit-to-lead activities were conducted through independent SAR studies of each of the four pyrimidine substituents. Optimal activity was observed upon removal of the pyrimidine C-4 chloro substituent. The intact C-6 carboribose is required for IRAK4 inhibition. Numerous heteroaryls were tolerated at the C-5 position, with azabenzothiazoles conferring the best activities. Aminoheteroaryls were preferred at the C-2 position. These studies led to the discovery of inhibitors 35, 36, and 38 that exhibit nanomolar inhibition of IRAK4, improved ligand efficiencies, and modest kinase selectivities.
Collapse
|
212
|
Role of IL-38 and its related cytokines in inflammation. Mediators Inflamm 2015; 2015:807976. [PMID: 25873772 PMCID: PMC4383490 DOI: 10.1155/2015/807976] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 10/13/2014] [Indexed: 12/13/2022] Open
Abstract
Interleukin- (IL-) 38 is a recently discovered cytokine and is the tenth member of the IL-1 cytokine family. IL-38 shares structural features with IL-1 receptor antagonist (IL-1Ra) and IL-36Ra. IL-36R is the specific receptor of IL-38, a partial receptor antagonist of IL-36. IL-38 inhibits the production of T-cell cytokines IL-17 and IL-22. IL-38 also inhibits the production of IL-8 induced by IL-36γ, thus inhibiting inflammatory responses. IL-38-related cytokines, including IL-1Ra and IL-36Ra, are involved in the regulation of inflammation and immune responses. The study of IL-38 and IL-38-related cytokines might provide new insights for developing anti-inflammatory treatments in the near future.
Collapse
|
213
|
Abstract
Innate immune responses depend on timely recognition of pathogenic or danger signals by multiple cell surface or cytoplasmic receptors and transmission of signals for proper counteractions through adaptor and effector molecules. At the forefront of innate immunity are four major signaling pathways, including those elicited by Toll-like receptors, RIG-I-like receptors, inflammasomes, or cGAS, each with its own cellular localization, ligand specificity, and signal relay mechanism. They collectively engage a number of overlapping signaling outcomes, such as NF-κB activation, interferon response, cytokine maturation, and cell death. Several proteins often assemble into a supramolecular complex to enable signal transduction and amplification. In this article, we review the recent progress in mechanistic delineation of proteins in these pathways, their structural features, modes of ligand recognition, conformational changes, and homo- and hetero-oligomeric interactions within the supramolecular complexes. Regardless of seemingly distinct interactions and mechanisms, the recurring themes appear to consist of autoinhibited resting-state receptors, ligand-induced conformational changes, and higher-order assemblies of activated receptors, adaptors, and signaling enzymes through conserved protein-protein interactions.
Collapse
Affiliation(s)
- Qian Yin
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, and
| | | | | | | |
Collapse
|
214
|
Chi PL, Chuang YC, Chen YW, Lin CC, Hsiao LD, Yang CM. The CO donor CORM-2 inhibits LPS-induced vascular cell adhesion molecule-1 expression and leukocyte adhesion in human rheumatoid synovial fibroblasts. Br J Pharmacol 2015; 171:2993-3009. [PMID: 24628691 DOI: 10.1111/bph.12680] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 12/15/2013] [Accepted: 12/24/2013] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND AND PURPOSE Infection with Gram-negative bacteria has been recognized as an initiator of rheumatoid arthritis, which is characterized by chronic inflammation and infiltration of immune cells. Carbon monoxide (CO) exhibits anti-inflammatory properties. Here we have investigated the detailed mechanisms of vascular cell adhesion molecule-1 (VCAM-1) expression induced by LPS and if CO inhibited LPS-induced leukocyte adhesion to synovial fibroblasts by suppressing VCAM-1 expression. EXPERIMENTAL APPROACH Human rheumatoid arthritis synovial fibroblasts (RASFs) were incubated with LPS and/or the CO-releasing compound CORM-2. Effects of LPS on VCAM-1 levels were determined by analysing mRNA expression, promoter activity, protein expression, and immunohistochemical staining. The molecular mechanisms were investigated by determining the expression, activation, and binding activity of transcriptional factors using target signal antagonists. KEY RESULTS CORM-2 significantly inhibited inflammatory responses in LPS-treated RASFs by down-regulating the expression of adhesion molecule VCAM-1 and leukocyte infiltration. The down-regulation of LPS-induced VCAM-1 expression involved inhibition of the expression of phosphorylated-NF-κB p65 and AP-1 (p-c-Jun, c-Jun and c-Fos mRNA levels). These results were confirmed by chromatin immunoprecipitation assay to detect NF-κB and AP-1 DNA binding activity. CONCLUSIONS AND IMPLICATIONS LPS-mediated formation of the TLR4/MyD88/TRAF6/c-Src complex regulated NF-κB and MAPKs/AP-1 activation leading to VCAM-1 expression and leukocyte adhesion. CORM-2, which liberates CO to elicit direct biological activities, attenuated LPS-induced VCAM-1 expression by interfering with NF-κB and AP-1 activation, and significantly reduced LPS-induced immune cell infiltration of the synovium.
Collapse
Affiliation(s)
- Pei-Ling Chi
- Department of Physiology and Pharmacology and Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | | | | | | | | | | |
Collapse
|
215
|
Vallejo-Vaz AJ. Novel Biomarkers in Heart Failure Beyond Natriuretic Peptides - The Case for Soluble ST2. Eur Cardiol 2015; 10:37-41. [PMID: 30310421 DOI: 10.15420/ecr.2015.10.01.37] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Despite more effective management of heart failure over the past few decades, its burden as a chronic disease has grown and is expected to continue to rise, representing a major health problem for years to come. Having reliable tools for early diagnosis and risk stratification can help managing the condition more efficiently. In this context, the interest for biomarkers has increased considerably in the last years following the useful clinical role of B-type natriuretic peptides. These biomarkers have been extensively studied and have become established diagnostic and prognostic biomarkers in heart failure. Despite their usefulness, limitations still remain a problem in clinical practice and the search for new biomarkers has therefore continued. Amongst the most promising newer biomarkers, soluble ST2 deserves further consideration. The present review will focus on the role of this new biomarker in the context of heart failure.
Collapse
Affiliation(s)
- Antonio J Vallejo-Vaz
- Cardiovascular Sciences, Cardiovascular and Cell Sciences Research Institute, St George's University of London, London, UK
| |
Collapse
|
216
|
Lacy SE, Wu C, Ambrosi DJ, Hsieh CM, Bose S, Miller R, Conlon DM, Tarcsa E, Chari R, Ghayur T, Kamath RV. Generation and characterization of ABT-981, a dual variable domain immunoglobulin (DVD-Ig(TM)) molecule that specifically and potently neutralizes both IL-1α and IL-1β. MAbs 2015; 7:605-19. [PMID: 25764208 PMCID: PMC4622731 DOI: 10.1080/19420862.2015.1026501] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 02/24/2015] [Accepted: 03/01/2015] [Indexed: 12/15/2022] Open
Abstract
Interleukin-1 (IL-1) cytokines such as IL-1α, IL-1β, and IL-1Ra contribute to immune regulation and inflammatory processes by exerting a wide range of cellular responses, including expression of cytokines and chemokines, matrix metalloproteinases, and nitric oxide synthetase. IL-1α and IL-1β bind to IL-1R1 complexed to the IL-1 receptor accessory protein and induce similar physiological effects. Preclinical and clinical studies provide significant evidence for the role of IL-1 in the pathogenesis of osteoarthritis (OA), including cartilage degradation, bone sclerosis, and synovial proliferation. Here, we describe the generation and characterization of ABT-981, a dual variable domain immunoglobulin (DVD-Ig) of the IgG1/k subtype that specifically and potently neutralizes IL-1α and IL-1β. In ABT-981, the IL-1β variable domain resides in the outer domain of the DVD-Ig, whereas the IL-1α variable domain is located in the inner position. ABT-981 specifically binds to IL-1α and IL-1β, and is physically capable of binding 2 human IL-1α and 2 human IL-1β molecules simultaneously. Single-dose intravenous and subcutaneous pharmacokinetics studies indicate that ABT-981 has a half-life of 8.0 to 10.4 d in cynomolgus monkey and 10.0 to 20.3 d in rodents. ABT-981 exhibits suitable drug-like-properties including affinity, potency, specificity, half-life, and stability for evaluation in human clinical trials. ABT-981 offers an exciting new approach for the treatment of OA, potentially addressing both disease modification and symptom relief as a disease-modifying OA drug.
Collapse
Affiliation(s)
- Susan E Lacy
- AbbVie Bioresearch Center; Global Biologics; Worcester, MA USA
| | - Chengbin Wu
- Shanghai CP Guojian Pharmaceutical Co., Ltd.; Shanghai, China
| | | | | | - Sahana Bose
- AbbVie Bioresearch Center; Global Biologics; Worcester, MA USA
| | - Renee Miller
- AbbVie Bioresearch Center; Global Biologics; Worcester, MA USA
| | - Donna M Conlon
- AbbVie Bioresearch Center; Immunology Pharmacology; Worcester, MA USA
| | - Edit Tarcsa
- AbbVie Bioresearch Center; DMPK-BA; Worcester, MA USA
| | - Ravi Chari
- AbbVie Bioresearch Center; Drug Product Development; Worcester, MA USA
| | - Tariq Ghayur
- AbbVie Bioresearch Center; Global Biologics; Worcester, MA USA
| | - Rajesh V Kamath
- AbbVie Bioresearch Center; Foundational Immunology; Worcester, MA USA
| |
Collapse
|
217
|
Neonatal sepsis and inflammatory mediators. Mediators Inflamm 2014; 2014:269681. [PMID: 25614712 PMCID: PMC4295603 DOI: 10.1155/2014/269681] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 12/09/2014] [Indexed: 12/18/2022] Open
Abstract
Neonatal sepsis is a major cause of morbidity and mortality and its signs and symptoms are nonspecific, which makes the diagnosis difficult. The routinely used laboratory tests are not effective methods of analysis, as they are extremely nonspecific and often cause inappropriate use of antibiotics. Sepsis is the result of an infection associated with a systemic inflammatory response with production and release of a wide range of inflammatory mediators. Cytokines are potent inflammatory mediators and their serum levels are increased during infections, so changes from other inflammatory effector molecules may occur. Although proinflammatory and anti-inflammatory cytokines have been identified as probable markers of neonatal infection, in order to characterize the inflammatory response during sepsis, it is necessary to analyze a panel of cytokines and not only the measurement of individual cytokines. Measurements of inflammatory mediators bring new options for diagnosing and following up neonatal sepsis, thus enabling early treatment and, as a result, increased neonatal survival. By taking into account the magnitude of neonatal sepsis, the aim of this review is to address the role of cytokines in the pathogenesis of neonatal sepsis and its value as a diagnostic criterion.
Collapse
|
218
|
Modulatory effect of three antibiotics on uterus bovine contractility in vitro and likely therapeutic approaches in reproduction. Theriogenology 2014; 82:1287-95. [DOI: 10.1016/j.theriogenology.2014.08.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 08/18/2014] [Accepted: 08/19/2014] [Indexed: 11/17/2022]
|
219
|
Uchikawa S, Yoda M, Tohmonda T, Kanaji A, Matsumoto M, Toyama Y, Horiuchi K. ADAM17 regulates IL-1 signaling by selectively releasing IL-1 receptor type 2 from the cell surface. Cytokine 2014; 71:238-45. [PMID: 25461404 DOI: 10.1016/j.cyto.2014.10.032] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Revised: 09/22/2014] [Accepted: 10/21/2014] [Indexed: 01/22/2023]
Abstract
Interleukin (IL)-1 is one of the most evolutionarily conserved cytokines and plays an essential role in the regulation of innate immunity. IL-1 binds to two different receptors, IL-1R1 and IL-1R2, which share approximately 28% amino acid homology. IL-1R1 contains a cytoplasmic domain and is capable of transducing cellular signals; by contrast, IL-1R2 lacks a functional cytoplasmic domain and serves as a decoy receptor for IL-1. Interestingly, IL-1R2 is proteolytically cleaved and also functions as a soluble receptor that blocks IL-1 activity. In the present study, we examined the shedding properties of IL-1R2 and demonstrate that ADAM17 is de facto the major sheddase for IL-1R2 and that introducing a mutation into the juxta-membrane domain of IL-1R2 significantly desensitizes IL-1R2 to proteolytic cleavage. IL-1R1 was almost insensitive to ADAM17-dependent cleavage; however, the replacement of the juxta-membrane domain of IL-R1 with that of IL-1R2 significantly increased the sensitivity of IL-1R1 to shedding. Furthermore, we demonstrate that ADAM17 indirectly enhances IL-1 signaling in a cell-autonomous manner by selectively cleaving IL-1R2. Taken together, the data collected in the present study indicate that ADAM17 affects sensitivity to IL-1 by changing the balance between IL-1R1 and the decoy receptor IL-1R2.
Collapse
Affiliation(s)
- Shinichi Uchikawa
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | - Masaki Yoda
- Department of Anti-aging Orthopedic Research, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | - Takahide Tohmonda
- Department of Anti-aging Orthopedic Research, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | - Arihiko Kanaji
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | - Morio Matsumoto
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | - Yoshiaki Toyama
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | - Keisuke Horiuchi
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; Department of Anti-aging Orthopedic Research, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| |
Collapse
|
220
|
Jain A, Kaczanowska S, Davila E. IL-1 Receptor-Associated Kinase Signaling and Its Role in Inflammation, Cancer Progression, and Therapy Resistance. Front Immunol 2014; 5:553. [PMID: 25452754 PMCID: PMC4233944 DOI: 10.3389/fimmu.2014.00553] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 10/17/2014] [Indexed: 12/18/2022] Open
Abstract
Chronic inflammation has long been associated with the development of cancer. Among the various signaling pathways within cancer cells that can incite the expression of inflammatory molecules are those that activate IL-1 receptor-associated kinases (IRAK). The IRAK family is comprised of four family members, IRAK-1, IRAK-2, IRAK-3 (also known as IRAK-M), and IRAK-4, which play important roles in both positively and negatively regulating the expression of inflammatory molecules. The wide array of inflammatory molecules that are expressed in response to IRAK signaling within the tumor microenvironment regulate the production of factors which promote tumor growth, metastasis, immune suppression, and chemotherapy resistance. Based on published reports we propose that dysregulated activation of the IRAK signaling pathway in cancer cells contributes to disease progression by creating a highly inflammatory tumor environment. In this article, we present both theoretical arguments and reference experimental data in support of this hypothesis.
Collapse
Affiliation(s)
- Ajay Jain
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, State University of New York Upstate Medical University , Albany, NY , USA
| | - Sabina Kaczanowska
- Department of Microbiology and Immunology, University of Maryland School of Medicine , Baltimore, MD , USA
| | - Eduardo Davila
- Department of Microbiology and Immunology, University of Maryland School of Medicine , Baltimore, MD , USA ; Greenebaum Cancer Center , Baltimore, MD , USA
| |
Collapse
|
221
|
Geng X, Xu T, Niu Z, Zhou X, Zhao L, Xie Z, Xue D, Zhang F, Xu C. Differential proteome analysis of the cell differentiation regulated by BCC, CRH, CXCR4, GnRH, GPCR, IL1 signaling pathways in Chinese fire-bellied newt limb regeneration. Differentiation 2014; 88:85-96. [DOI: 10.1016/j.diff.2014.10.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 10/07/2014] [Accepted: 10/29/2014] [Indexed: 12/11/2022]
|
222
|
Sivick KE, Arpaia N, Reiner GL, Lee BL, Russell BR, Barton GM. Toll-like receptor-deficient mice reveal how innate immune signaling influences Salmonella virulence strategies. Cell Host Microbe 2014; 15:203-13. [PMID: 24528866 DOI: 10.1016/j.chom.2014.01.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 01/08/2014] [Accepted: 01/28/2014] [Indexed: 12/21/2022]
Abstract
Pathogens utilize features of the host response as cues to regulate virulence gene expression. Salmonella enterica serovar Typhimurium (ST) sense Toll-like receptor (TLR)-dependent signals to induce Salmonella Pathogenicity Island 2 (SPI2), a locus required for intracellular replication. To examine pathogenicity in the absence of such cues, we evaluated ST virulence in mice lacking all TLR function (Tlr2(-/-)xTlr4(-/-)xUnc93b1(3d/3d)). When delivered systemically to TLR-deficient mice, ST do not require SPI2 and maintain virulence by replicating extracellularly. In contrast, SPI2 mutant ST are highly attenuated after oral infection of the same mice, revealing a role for SPI2 in the earliest stages of infection, even when intracellular replication is not required. This early requirement for SPI2 is abolished in MyD88(-/-)xTRIF(-/-) mice lacking both TLR- and other MyD88-dependent signaling pathways, a potential consequence of compromised intestinal permeability. These results demonstrate how pathogens use plasticity in virulence strategies to respond to different host immune environments.
Collapse
Affiliation(s)
- Kelsey E Sivick
- Department of Molecular and Cell Biology, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Nicholas Arpaia
- Department of Molecular and Cell Biology, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Gabrielle L Reiner
- Department of Molecular and Cell Biology, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Bettina L Lee
- Department of Molecular and Cell Biology, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Bethany R Russell
- Department of Molecular and Cell Biology, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Gregory M Barton
- Department of Molecular and Cell Biology, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
223
|
Abstract
Cytokine-based immunotherapy is executed by harnessing cytokines to activate the immune system to suppress tumors. Th1-type cytokines including IL-1, IL-2, IL-12 and granulocyte-macrophage colony-stimulating factor are potent stimulators of Th1 differentiation and Th1-based antitumor response. Many preclinical studies demonstrated the antitumor effects of Th1 cytokines but their clinical efficacy is limited. Multiple factors influence the efficacy of immunotherapy for tumors. For instance immunosuppressive cells in the tumor microenvironment can produce inhibitory cytokines which suppress antitumor immune response. Most studies on cytokine immunotherapy focused on how to boost Th1 response; many studies combined cytokine-based therapy with other treatments to reverse immunosuppression in tumor microenvironment. In addition, cytokines have pleiotropic functions and some cytokines show paradoxical activities under different settings. Better understanding the physiological and pathological functions of cytokines helps clinicians to design Th1-based cancer therapy in clinical practice.
Collapse
Affiliation(s)
- Hong-Mei Xu
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE, United Kingdom.
| |
Collapse
|
224
|
Zhu S, Wang H, Shi R, Zhang R, Wang J, Kong L, Sun Y, He J, Kong J, Wang JF, Li XM. Chronic phencyclidine induces inflammatory responses and activates GSK3β in mice. Neurochem Res 2014; 39:2385-93. [PMID: 25270429 DOI: 10.1007/s11064-014-1441-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 09/18/2014] [Accepted: 09/22/2014] [Indexed: 11/26/2022]
Abstract
Use of phencyclidine (PCP) in rodents can mimic some aspects of schizophrenia. However, the underlying mechanism is still unclear. Growing evidence indicates that neuroinflammation plays a significant role in the pathophysiology of schizophrenia. In this study, we focused on inflammatory responses as target of PCP for inducing schizophrenia-like symptoms. 3-month-old C57BL/6J mice received daily injections of PCP (20 mg/kg, i.p.) or saline for one week. PCP-injected mice produced schizophrenia-like behaviours including impaired spatial short-term memory assessed by the Y-maze task and sensorimotor gating deficits in a prepulse inhibition task. Simultaneously, chronic PCP administration induced astrocyte and microglial activation in both the cortex and hippocampus. Additionally, the proinflammatory cytokine interleukin-1β was significantly up-regulated in PCP administrated mice. Furthermore, PCP treatment decreased ratio of the phospho-Ser9 epitope of glycogen synthase kinase-3β (GSK3β) over total GSK3β, which is indicative of increased GSK3β activity. These data demonstrate that chronic PCP in mouse produces inflammatory responses and GSK3β activation.
Collapse
Affiliation(s)
- Shenghua Zhu
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Manitoba, Winnipeg, MB, R3E 0T6, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
225
|
Poole AZ, Weis VM. TIR-domain-containing protein repertoire of nine anthozoan species reveals coral-specific expansions and uncharacterized proteins. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 46:480-488. [PMID: 24933613 DOI: 10.1016/j.dci.2014.06.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 06/04/2014] [Accepted: 06/05/2014] [Indexed: 06/03/2023]
Abstract
The intracellular toll/interleukin-1 receptor (TIR) domain plays an important role in vertebrate immunity, but the evolution and function of invertebrate TIR-domain-containing proteins is not fully understood. This study characterized and compared the TIR-domain-containing protein repertoire of nine cnidarians in class Anthozoa. A diverse set of proteins, including MyD88 (myeloid differentiation primary response protein 88), toll-like receptor (TLR)-like, interleukin-1 receptor (IL-1R)-like, and TIR-only proteins are present in the species surveyed. Increased numbers of TIR-only proteins were observed in corals compared to anemones, especially in the Acroporid and Pocilloporid coral families. This expansion could be linked to diversity of the microbial community on or in hosts and managing both positive and negative associations. Phylogenetic analysis indicates there are two groups of proteins with IL-1R-like domain architecture in anthozoans that potentially evolved independently of the vertebrate family. Bacterial-like TIR_2 domain proteins are also present, including one sequence with novel domain architecture. Overall this work promotes a better understanding of the anthozoan immune repertoire, which is important in the context learning about ancestral immune pathways and host-microbe interactions.
Collapse
Affiliation(s)
- Angela Z Poole
- Oregon State University, Department of Integrative Biology, 3029 Cordley Hall, Corvallis, OR 97331, United States.
| | - Virginia M Weis
- Oregon State University, Department of Integrative Biology, 3029 Cordley Hall, Corvallis, OR 97331, United States.
| |
Collapse
|
226
|
Sipos F, Fűri I, Constantinovits M, Tulassay Z, Műzes G. Contribution of TLR signaling to the pathogenesis of colitis-associated cancer in inflammatory bowel disease. World J Gastroenterol 2014; 20:12713-12721. [PMID: 25278673 PMCID: PMC4177458 DOI: 10.3748/wjg.v20.i36.12713] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 04/28/2014] [Accepted: 05/26/2014] [Indexed: 02/06/2023] Open
Abstract
In the intestine a balance between proinflammatory and repair signals of the immune system is essential for the maintenance of intestinal homeostasis. The innate immunity ensures a primary host response to microbial invasion, which induces an inflammatory process to localize the infection and prevent systemic dissemination of pathogens. The key elements of this process are the germline encoded pattern recognition receptors including Toll-like receptors (TLRs). If pathogens cannot be eliminated, they may elicit chronic inflammation, which may be partly mediated via TLRs. Additionally, chronic inflammation has long been suggested to trigger tissue tumorous transformation. Inflammation, the seventh hallmark of cancer, may affect all phases of tumor development, and evade the immune system. Inflammation acts as a cellular stressor and may trigger DNA damage or genetic instability. Furthermore, chronic inflammation can provoke genetic mutations and epigenetic mechanisms that promote malignant cell transformation. Colorectal cancers in inflammatory bowel disease patients are considered typical examples of inflammation-related cancers. Although data regarding the role of TLRs in the pathomechanism of cancer-associated colitis are rather conflicting, functionally these molecules can be classified as ”largely antitumorigenic” and ”largely pro-tumorigenic” with the caveat that the underlying signaling pathways are mainly context (i.e., organ-, tissue-, cell-) and ligand-dependent.
Collapse
|
227
|
Cell membrane penetrating function of the nuclear localization sequence in human cytokine IL-1α. Mol Biol Rep 2014; 41:8117-26. [PMID: 25205122 DOI: 10.1007/s11033-014-3711-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 08/28/2014] [Indexed: 10/24/2022]
Abstract
Cytokines are released from the cell, bind to their receptors, and affect cellular responses. The precursor form of interleukin 1 alpha (pIL-1α) has a nuclear localization sequence (NLS) that causes it to be localized to the nucleus and regulate specific gene expression. The amino acids of the NLS are basic amino acid-rich sequences, as is the cell penetrating peptide (CPP), which has been widely studied as a way to deliver macromolecules into cells. Here, we hypothesized that the NLS in pIL-1α (pIL-1αNLS) can penetrate the cell membrane and it could deliver macromolecules such as protein in vivo. We characterized cell membrane penetration ability of pIL-1αNLS or its tandem repeated form (2pIL-1αNLS) to enhance its intracellular delivery efficiency. 2pIL-1αNLS showed comparable protein delivery efficiency to TAT-CPP and it mediates endocytosis following heparan sulfate interaction. 2pIL-1αNLS conjugated enhanced green fluorescence protein was localized to the nucleus and the cytoplasm. Intra-peritoneal administration of 2pIL-1αNLS conjugated dTomato protein showed remarkable in vivo intracellular delivery efficiency in various tissues including spleen, liver, and intestine in mice. Moreover, cytotoxicity of 2pIL-1αNLS was not observed even at 100 μM. Our results demonstrate cell membrane-penetrating function of NLS in pIL-1α, which can be used as a safe therapeutic macromolecular delivery peptide.
Collapse
|
228
|
Bastien D, Lacroix S. Cytokine pathways regulating glial and leukocyte function after spinal cord and peripheral nerve injury. Exp Neurol 2014; 258:62-77. [PMID: 25017888 DOI: 10.1016/j.expneurol.2014.04.006] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Revised: 02/20/2014] [Accepted: 04/08/2014] [Indexed: 01/13/2023]
Abstract
Injury to the nervous system causes the almost immediate release of cytokines by glial cells and neurons. These cytokines orchestrate a complex array of responses leading to microgliosis, immune cell recruitment, astrogliosis, scarring, and the clearance of cellular debris, all steps that affect neuronal survival and repair. This review will focus on cytokines released after spinal cord and peripheral nerve injury and the primary signalling pathways triggered by these inflammatory mediators. Notably, the following cytokine families will be covered: IL-1, TNF, IL-6-like, TGF-β, and IL-10. Whether interfering with cytokine signalling could lead to novel therapies will also be discussed. Finally, the review will address whether manipulating the above-mentioned cytokine families and signalling pathways could exert distinct effects in the injured spinal cord versus peripheral nerve.
Collapse
Affiliation(s)
- Dominic Bastien
- Centre de recherche du Centre hospitalier universitaire de Québec-CHUL, Département de médecine moléculaire, Université Laval, Québec, QC, Canada
| | - Steve Lacroix
- Centre de recherche du Centre hospitalier universitaire de Québec-CHUL, Département de médecine moléculaire, Université Laval, Québec, QC, Canada..
| |
Collapse
|
229
|
Legido A, Katsetos CD. Experimental studies in epilepsy: immunologic and inflammatory mechanisms. Semin Pediatr Neurol 2014; 21:197-206. [PMID: 25510941 DOI: 10.1016/j.spen.2014.10.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In this article, we review the literature based on experimental studies lending credence to a relationship between epilepsy and immune-mediated mechanisms linked to central nervous system innate immunity. The brain innate immunity responses to neuronal injury or excessive neuronal activity are mediated by resident microglia and astroglia, but also neurons play an immunomodulatory role. Antigens or antibodies applied to the brain trigger an epileptogenic and inflammatory response. Furthermore, seizure activity and status epilepticus elicit the production and release of proinflammatory cytokines and chemokines. The immune pathogenesis of epilepsy involves complex cell-to-cell interactions including a cross talk between astrocytes and neurons, between astrocytes and brain microvascular endothelial cells, as well as reciprocal leukocyte-endothelial interactions in the context of disruption of the blood-brain barrier. There is a large body of literature from experimental studies showing that seizures can initiate a cascade of innate and adaptive immune responses from various cellular sources and perpetuate neuroinflammation through mechanisms involving transcription of inflammatory genes or posttranslational changes in cytokine release machinery. These inflammatory processes could also possibly contribute to the pathogenesis of comorbidities often associated with epilepsy. This opens exciting possibilities for the development of disease-modifying drugs aimed at mitigating neuroinflammation as a means of ameliorating epileptogenesis and lessening or preventing postictal brain injury.
Collapse
Affiliation(s)
- Agustín Legido
- Section of Neurology, St. Christopher's Hospital for Children, Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA.
| | - Christos D Katsetos
- Section of Neurology, St. Christopher's Hospital for Children, Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA
| |
Collapse
|
230
|
Abstract
The human pathogenic fungus Candida albicans is the predominant cause of both superficial and invasive forms of candidiasis. C. albicans primarily infects immunocompromised individuals as a result of either immunodeficiency or intervention therapy, which highlights the importance of host immune defences in preventing fungal infections. The host defence system utilises a vast communication network of cells, proteins, and chemical signals distributed in blood and tissues, which constitute innate and adaptive immunity. Over the last decade the identity of many key molecules mediating host defence against C. albicans has been identified. This review will discuss how the host recognises this fungus, the events induced by fungal cells, and the host innate and adaptive immune defences that ultimately resolve C. albicans infections during health.
Collapse
|
231
|
Appropriate development of the liver Treg compartment is modulated by the microbiota and requires TGF-β and MyD88. J Immunol Res 2014; 2014:279736. [PMID: 25177709 PMCID: PMC4142300 DOI: 10.1155/2014/279736] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Accepted: 06/30/2014] [Indexed: 01/22/2023] Open
Abstract
Neither the early postnatal development of the liver Treg compartment nor the factors that regulate its development has been characterized. We compared the early developmental patterns of Treg cell accumulation in murine liver, thymus, and spleen. A FoxP3EGFP reporter mouse was employed to identify Treg cells. Mononuclear cells were isolated from organs postnatally, stained for CD4, and examined by flow cytometry to enumerate FoxP3+CD4hi cells. To assess roles for TGF-β1, MyD88, and TLR2, gene-specific knockout pups were generated from heterozygous breeders. To test the role of commensal bacteria, pregnant dams were administered antibiotics during gestation and after parturition. The pattern of appearance of Treg cells differed in liver, spleen, and thymus. Notably, at 1-2 weeks, the frequency of CD4hi FoxP3+ T cells in liver exceeded that in spleen by 1.5- to 2-fold. The relative increase in liver Treg frequency was transient and was dependent upon TGF-β1 and MyD88, but not TLR2, and was abrogated by antibiotic treatment. A relative increase in liver Treg frequency occurs approximately 1-2 weeks after parturition that appears to be driven by colonization of the intestine with commensal bacteria and is mediated by a pathway that requires TGF-β1 and MyD88, but not TLR2.
Collapse
|
232
|
Vogelpoel LTC, Hansen IS, Visser MW, Nagelkerke SQ, Kuijpers TW, Kapsenberg ML, de Jong EC, den Dunnen J. FcγRIIa cross-talk with TLRs, IL-1R, and IFNγR selectively modulates cytokine production in human myeloid cells. Immunobiology 2014; 220:193-9. [PMID: 25108563 DOI: 10.1016/j.imbio.2014.07.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 07/01/2014] [Accepted: 07/16/2014] [Indexed: 01/05/2023]
Abstract
Myeloid antigen-presenting cells (APCs) tailor immune responses to the pathogen involved through the production of specific pro- and anti-inflammatory cytokines. It is becoming increasingly clear that the ultimate cytokine profile produced by myeloid APCs crucially depends on interaction between multiple pathogen recognizing receptors. In this respect, we recently identified an important role for cross-talk between Fc gamma receptor IIa (FcγRIIa) and Toll-like receptors (TLRs) in human dendritic cells (DCs), which induces anti-bacterial immunity through the selective induction of TNFα and Th17-promoting cytokines. Here, we show that FcγRIIa-TLR cross-talk is not restricted to DCs, but is a common feature of various human myeloid APC subsets including monocytes and macrophages. Interestingly, FcγRIIa-TLR cross-talk in monocytes resulted in the induction of a cytokine profile distinct from that in DCs and macrophages, indicating that FcγRIIa stimulation induces cell-type and tissue specific responses. Surprisingly, we show that the FCGR2A H131R single nucleotide polymorphism (SNP), which is known to greatly affect FcγRIIa-mediated uptake of IgG2-opsonized bacteria, did not affect FcγRIIa-dependent cytokine production, indicating that these processes are differently regulated. In addition, we demonstrate that FcγRIIa selectively synergized with TLRs, IL-1R, and IFNγR, but did not affect cytokine production induced by other receptors such as C-type lectin receptor Dectin-1. Taken together, these data demonstrate that FcγRIIa-dependent modulation of cytokine production is more widespread than previously considered, and indicate that cross-talk of FcγRIIa with various receptors and in multiple cell types contributes to the induction of pathogen and tissue-specific immunity.
Collapse
Affiliation(s)
- Lisa T C Vogelpoel
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Ivo S Hansen
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Marijke W Visser
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Sietse Q Nagelkerke
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Taco W Kuijpers
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Pediatric Hematology, Immunology and Infectious Disease, Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Martien L Kapsenberg
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Esther C de Jong
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jeroen den Dunnen
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
233
|
The TIR Domain Containing Locus of Enterococcus faecalis Is Predominant among Urinary Tract Infection Isolates and Downregulates Host Inflammatory Response. Int J Microbiol 2014; 2014:918143. [PMID: 25147569 PMCID: PMC4131471 DOI: 10.1155/2014/918143] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 06/11/2014] [Accepted: 06/30/2014] [Indexed: 12/18/2022] Open
Abstract
Based on Toll/interleukin-1 receptor (TIR) domain structure homology, we detected a previously uncharacterized gene encoding for a TIR domain containing protein (Tcp) in the genome of Enterococcus faecalis. We assigned this gene the name tcpF (as in Tcp of E. faecalis). Screening of E. faecalis samples revealed that tcpF is more common in isolates from urinary tract infections (UTIs) than in human faecal flora. tcpF alleles showed moderate single nucleotide polymorphism (SNP) among UTI isolates. Infection of mouse RAW264.7 macrophages with a tcpF knock-out mutant led to elevated cytokine response compared to the isogenic wild type E. faecalis strain. In silico analysis predicted significant tertiary structure homology to the TIR domain of human TLR1 (TLR1-TIR). When transiently expressed in cultured eukaryotic cells, TcpF caused suppression of TLR2-dependent NF-κB activation suggesting for TcpF a role as a factor in E. faecalis that benefits colonization by modulating the host's immune responses.
Collapse
|
234
|
Wang H, Flannery SM, Dickhöfer S, Huhn S, George J, Kubarenko AV, Lascorz J, Bevier M, Willemsen J, Pichulik T, Schafmayer C, Binder M, Manoury B, Paludan SR, Alarcon-Riquelme M, Bowie AG, Försti A, Weber ANR. A coding IRAK2 protein variant compromises Toll-like receptor (TLR) signaling and is associated with colorectal cancer survival. J Biol Chem 2014; 289:23123-23131. [PMID: 24973222 DOI: 10.1074/jbc.m113.492934] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Within innate immune signaling pathways, interleukin-1 receptor-associated kinases (IRAKs) fulfill key roles downstream of multiple Toll-like receptors and the interleukin-1 receptor. Although human IRAK4 deficiency was shown to lead to severe immunodeficiency in response to pyogenic bacterial infection during childhood, little is known about the role of human IRAK2. We here identified a non-synonymous IRAK2 variant, rs35060588 (coding R214G), as hypofunctional in terms of NF-κB signaling and Toll-like receptor-mediated cytokine induction. This was due to reduced ubiquitination of TRAF6, a key step in signal transduction. IRAK2 rs35060588 occurs in 3-9% of individuals in different ethnic groups, and our studies suggested a genetic association of rs35060588 with colorectal cancer survival. This for the first time implicates human IRAK2 in a human disease and highlights the R214G IRAK2 variant as a potential novel and broadly applicable biomarker for disease or as a therapeutic intervention point.
Collapse
Affiliation(s)
- Hui Wang
- Junior Research Group Toll-like Receptors and Cancer and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany; Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Sinead M Flannery
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Sabine Dickhöfer
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Stefanie Huhn
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Julie George
- Junior Research Group Toll-like Receptors and Cancer and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Andriy V Kubarenko
- Junior Research Group Toll-like Receptors and Cancer and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Jesus Lascorz
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Melanie Bevier
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Joschka Willemsen
- Department of Infectious Diseases/Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, 69120 Heidelberg, Germany
| | - Tica Pichulik
- Junior Research Group Toll-like Receptors and Cancer and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Clemens Schafmayer
- Department of General and Thoracic Surgery, University Hospital Schleswig-Holstein, 24105 Kiel, Germany,; POPGEN Biobank Project, Christian-Albrechts University, 24105 Kiel, Germany
| | - Marco Binder
- Department of Infectious Diseases/Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, 69120 Heidelberg, Germany
| | - Bénédicte Manoury
- INSERM, Unité 1013 and Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, 75015 Paris, France
| | - Søren R Paludan
- Department of Biomedicine, Aarhus University, Bartholin Building, 8000 Aarhus, Denmark
| | - Marta Alarcon-Riquelme
- Pfizer-Universidad de Granada-Junta de Andalucía Centre for Genomics and Oncological Research, Avenida de la Ilustración 114, 18016 Granada, Spain,; Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104,; BIOLUPUS Network, European Science Foundation, F-67080 Strasbourg Cedex, France, and
| | - Andrew G Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Asta Försti
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany,; Center for Primary Health Care Research, Clinical Research Center, Lund University, 20502 Malmö, Sweden
| | - Alexander N R Weber
- Junior Research Group Toll-like Receptors and Cancer and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany; Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany,.
| |
Collapse
|
235
|
Günther S, Sundberg EJ. Molecular Determinants of Agonist and Antagonist Signaling through the IL-36 Receptor. THE JOURNAL OF IMMUNOLOGY 2014; 193:921-30. [DOI: 10.4049/jimmunol.1400538] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
236
|
Turner MD, Nedjai B, Hurst T, Pennington DJ. Cytokines and chemokines: At the crossroads of cell signalling and inflammatory disease. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2563-2582. [PMID: 24892271 DOI: 10.1016/j.bbamcr.2014.05.014] [Citation(s) in RCA: 1408] [Impact Index Per Article: 128.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 05/22/2014] [Accepted: 05/23/2014] [Indexed: 12/14/2022]
Abstract
Inflammation occurs as a result of exposure of tissues and organs to harmful stimuli such as microbial pathogens, irritants, or toxic cellular components. The primary physical manifestations of inflammation are redness, swelling, heat, pain, and loss of function to the affected area. These processes involve the major cells of the immune system, including monocytes, macrophages, neutrophils, basophils, dendritic cells, mast cells, T-cells, and B-cells. However, examination of a range of inflammatory lesions demonstrates the presence of specific leukocytes in any given lesion. That is, the inflammatory process is regulated in such a way as to ensure that the appropriate leukocytes are recruited. These events are in turn controlled by a host of extracellular molecular regulators, including members of the cytokine and chemokine families that mediate both immune cell recruitment and complex intracellular signalling control mechanisms that characterise inflammation. This review will focus on the role of the main cytokines, chemokines, and their receptors in the pathophysiology of auto-inflammatory disorders, pro-inflammatory disorders, and neurological disorders involving inflammation.
Collapse
Affiliation(s)
- Mark D Turner
- Interdisciplinary Biomedical Research Centre, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham NG11 8NS, United Kingdom.
| | - Belinda Nedjai
- Leukocyte Biology Section, National Heart and Lung Institute, Imperial College, South Kensington, London SW7 2AZ, United Kingdom
| | - Tara Hurst
- Interdisciplinary Biomedical Research Centre, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham NG11 8NS, United Kingdom
| | - Daniel J Pennington
- Blizard Institute, Barts and The London School of Medicine, Queen Mary University of London, Whitechapel, London E1 2AT, United Kingdom
| |
Collapse
|
237
|
RETRACTED ARTICLE: Variant Interleukin-1 Receptor-Associated Kinase-2 Mediates Increased NF-κB and p38 Activity Induced by Lipopolysaccharide. Inflammation 2014; 37:993. [DOI: 10.1007/s10753-014-9823-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
238
|
Ueno-Shuto K, Kato K, Tasaki Y, Sato M, Sato K, Uchida Y, Sakai H, Ono T, Suico MA, Mitsutake K, Tokutomi N, Kai H, Shuto T. Lipopolysaccharide decreases single immunoglobulin interleukin-1 receptor-related molecule (SIGIRR) expression by suppressing specificity protein 1 (Sp1) via the Toll-like receptor 4 (TLR4)-p38 pathway in monocytes and neutrophils. J Biol Chem 2014; 289:18097-109. [PMID: 24821721 DOI: 10.1074/jbc.m113.532093] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Single immunoglobulin interleukin-1 receptor-related molecule (SIGIRR) is one of the immunoglobulin-like membrane proteins that is crucial for negative regulation of toll-like receptor 4 (TLR4) and interleukin-1 receptor. Despite the importance of understanding its expression and function, knowledge is limited on the regulatory mechanism in the epithelial tissues, such as the liver, lung, and gut, where its predominant expression is originally described. Here, we found expression of SIGIRR in non-epithelial innate immune cells, including primary peripheral blood monocytes, polymorphonuclear neutrophils, monocytic RAW264 cells, and neutrophilic-differentiated HL-60 cells. Consistent with previous findings in epithelial tissues, SIGIRR gene and protein expression were also down-regulated by LPS treatment in a time-dependent manner in primary blood monocytes and polymorphonuclear neutrophils. A reduction was also observed in RAW264 and differentiated HL-60 cells. Notably, exogenous introduction of the dominant negative form of TLR4 and siRNA of p38 resulted in inhibition of LPS-induced SIGIRR down-regulation, whereas treatment with p38 activator anisomycin showed a dose-dependent decrease in SIGIRR expression, suggesting TLR4-p38 signal as a critical pathway for LPS-induced SIGIRR down-regulation. Finally, reporter gene and chromatin immunoprecipitation assays demonstrated that Sp1 is a key factor that directly binds to the proximal promoter of SIGIRR gene and consequently regulates basal SIGIRR expression, which is negatively regulated by the LPS-dependent TLR4-p38 pathway. In summary, the data precisely demonstrate how LPS down-regulates SIGIRR expression and provide a role of LPS signal that counteracts Sp1-dependent basal promoter activation of SIGIRR gene via TLR4-p38 pathway in non-epithelial innate immune cells.
Collapse
Affiliation(s)
- Keiko Ueno-Shuto
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan, the Laboratory of Pharmacology, Division of Life Science, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082, Japan
| | - Kosuke Kato
- the Department of Physiology and Lung Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, and
| | - Yukihiro Tasaki
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Miki Sato
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Keizo Sato
- the School of Pharmacy, Kyushu University of Health and Welfare, Nobeoka, Miyazaki 882-8508, Japan
| | - Yuji Uchida
- the Laboratory of Pharmacology, Division of Life Science, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082, Japan
| | - Hiromichi Sakai
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Tomomi Ono
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Mary Ann Suico
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Kazunori Mitsutake
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Naofumi Tokutomi
- the Laboratory of Pharmacology, Division of Life Science, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082, Japan
| | - Hirofumi Kai
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Tsuyoshi Shuto
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan,
| |
Collapse
|
239
|
Verstak B, Stack J, Ve T, Mangan M, Hjerrild K, Jeon J, Stahl R, Latz E, Gay N, Kobe B, Bowie AG, Mansell A. The TLR signaling adaptor TRAM interacts with TRAF6 to mediate activation of the inflammatory response by TLR4. J Leukoc Biol 2014; 96:427-36. [PMID: 24812060 DOI: 10.1189/jlb.2a0913-487r] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
TLRs act as sentinels in professional immune cells to detect and initiate the innate immune response to pathogen challenge. TLR4 is a widely expressed TLR, responsible for initiating potent immune responses to LPS. TRAM acts to bridge TLR4 with TRIF, orchestrating the inflammatory response to pathogen challenge. We have identified a putative TRAF6-binding motif in TRAM that could mediate a novel signaling function for TRAM in TLR4 signaling. TRAM and TRAF6 association was confirmed by immunoprecipitation of endogenous, ectopically expressed and recombinant proteins, which was ablated upon mutation of a key Glu residue in TRAM (TRAM E183A). TRAF6 and TRAM were observed colocalizing using confocal microscopy following ectopic expression in cells and the ability of TRAM and TRAM E183A to activate luciferase-linked reporter assays was determined in HEK293 and TRAF6-deficient cells. Importantly, TRAM-deficient macrophages reconstituted with TRAM E183A display significantly reduced inflammatory TNF-α, IL-6, and RANTES protein production compared with WT TRAM. These results demonstrate a novel role for TRAM in TLR4-mediated signaling in regulating inflammatory responses via its interaction with TRAF6, distinct from its role as a bridging adaptor between TLR4 and TRIF.
Collapse
Affiliation(s)
- Brett Verstak
- Centre for Innate Immunity and Infectious Diseases, MIMR-PHI Institute of Medical Research, Monash University, Melbourne, Victoria, Australia; Department of Biochemistry, University of Cambridge, United Kingdom
| | - Julianne Stack
- Immunology Research Centre, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Thomas Ve
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, and Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Matthew Mangan
- Centre for Innate Immunity and Infectious Diseases, MIMR-PHI Institute of Medical Research, Monash University, Melbourne, Victoria, Australia
| | - Kathryn Hjerrild
- Centre for Innate Immunity and Infectious Diseases, MIMR-PHI Institute of Medical Research, Monash University, Melbourne, Victoria, Australia
| | - Jannah Jeon
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, and Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Rainer Stahl
- Institute of Innate Immunity, University Hospitals, University of Bonn, Germany; and
| | - Eicke Latz
- Institute of Innate Immunity, University Hospitals, University of Bonn, Germany; and Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Nick Gay
- Department of Biochemistry, University of Cambridge, United Kingdom
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, and Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Andrew G Bowie
- Immunology Research Centre, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Ashley Mansell
- Centre for Innate Immunity and Infectious Diseases, MIMR-PHI Institute of Medical Research, Monash University, Melbourne, Victoria, Australia;
| |
Collapse
|
240
|
Hahn WS, Kuzmicic J, Burrill JS, Donoghue MA, Foncea R, Jensen MD, Lavandero S, Arriaga EA, Bernlohr DA. Proinflammatory cytokines differentially regulate adipocyte mitochondrial metabolism, oxidative stress, and dynamics. Am J Physiol Endocrinol Metab 2014; 306:E1033-45. [PMID: 24595304 PMCID: PMC4010657 DOI: 10.1152/ajpendo.00422.2013] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Proinflammatory cytokines differentially regulate adipocyte mitochondrial metabolism, oxidative stress, and dynamics. Macrophage infiltration of adipose tissue and the chronic low-grade production of inflammatory cytokines have been mechanistically linked to the development of insulin resistance, the forerunner of type 2 diabetes mellitus. In this study, we evaluated the chronic effects of TNFα, IL-6, and IL-1β on adipocyte mitochondrial metabolism and morphology using the 3T3-L1 model cell system. TNFα treatment of cultured adipocytes led to significant changes in mitochondrial bioenergetics, including increased proton leak, decreased ΔΨm, increased basal respiration, and decreased ATP turnover. In contrast, although IL-6 and IL-1β decreased maximal respiratory capacity, they had no effect on ΔΨm and varied effects on ATP turnover, proton leak, or basal respiration. Only TNFα treatment of 3T3-L1 cells led to an increase in oxidative stress (as measured by superoxide anion production and protein carbonylation) and C16 ceramide synthesis. Treatment of 3T3-L1 adipocytes with cytokines led to decreased mRNA expression of key transcription factors and control proteins implicated in mitochondrial biogenesis, including PGC-1α and eNOS as well as deceased expression of COX IV and Cyt C. Whereas each cytokine led to effects on expression of mitochondrial markers, TNFα exclusively led to mitochondrial fragmentation and decreased the total level of OPA1 while increasing OPA1 cleavage, without expression of levels of mitofusin 2, DRP-1, or mitofilin being affected. In summary, these results indicate that inflammatory cytokines have unique and specialized effects on adipocyte metabolism, but each leads to decreased mitochondrial function and a reprogramming of fat cell biology.
Collapse
Affiliation(s)
- Wendy S Hahn
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | | | | | | | | | | | | | | | | |
Collapse
|
241
|
England H, Summersgill HR, Edye ME, Rothwell NJ, Brough D. Release of interleukin-1α or interleukin-1β depends on mechanism of cell death. J Biol Chem 2014; 289:15942-50. [PMID: 24790078 PMCID: PMC4047367 DOI: 10.1074/jbc.m114.557561] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The cytokine interleukin-1 (IL-1) has two main pro-inflammatory forms, IL-1α and IL-1β, which are central to host responses to infection and to damaging sterile inflammation. Processing of IL-1 precursor proteins to active cytokines commonly occurs through activation of proteases, notably caspases and calpains. These proteases are instrumental in cell death, and inflammation and cell death are closely associated, hence we sought to determine the impact of cell death pathways on IL-1 processing and release. We discovered that apoptotic regulation of caspase-8 specifically induced the processing and release of IL-1β. Conversely, necroptosis caused the processing and release of IL-1α, and this was independent of IL-1β processing and release. These data suggest that the mechanism through which an IL-1-expressing cell dies dictates the nature of the inflammatory mechanism that follows. These insights may allow modification of inflammation through the selective targeting of cell death mechanisms during disease.
Collapse
Affiliation(s)
- Hazel England
- From the Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Holly R Summersgill
- From the Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Michelle E Edye
- From the Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Nancy J Rothwell
- From the Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester M13 9PT, United Kingdom
| | - David Brough
- From the Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester M13 9PT, United Kingdom
| |
Collapse
|
242
|
Yadav VR, Vilekar P, Awasthi S, Awasthi V. Hemorrhage-induced interleukin-1 receptor pathway in lung is suppressed by 3,5-bis(2-fluorobenzylidene)-4-piperidone in a rat model of hypovolemic shock. Artif Organs 2014; 38:675-83. [PMID: 24749913 DOI: 10.1111/aor.12305] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Severe blood loss in victims of trauma creates an exaggerated inflammatory background that contributes to the development of intravascular coagulopathy and multiple organ dysfunction syndrome. We hypothesized that treatment with diphenyldifluoroketone EF24, an inhibitor of nuclear factor kappa-B, would have salutary effects in hemorrhagic shock. The objective of this study was to investigate the effect of EF24 on the expression of the interleukin-1 receptor (IL-1R) superfamily in a rat model of hypovolemic shock. Hypovolemia was induced by gradually withdrawing approximately 50% of circulating blood, and EF24 was administered intraperitoneally (0.2 mg/kg) in 50 μL of saline. After 6 h of shock, lung tissue was probed immunohistochemically and by immunoblotting to study the expression of Toll-like receptor 4 (TLR4), IL-1R, suppression of tumorigenicity 2 (ST2), and single immunoglobulin IL-1R-related (SIGIRR). The tissue-associated pro-inflammatory cytokines, tumor necrosis factor alpha (TNF-α) and IL-6, were measured by enzyme-linked immunosorbent assay. We observed a reduction in immunoreactive TLR4 and IL-1R1 in lung tissue of rats treated with EF24. Simultaneously, the pulmonary expression of ST2 and SIGIRR (the putative down-regulators of the pro-inflammatory IL-1R pathway) was increased in EF24-treated hemorrhaged rats. The concentration of hemorrhage-induced TNF-α and IL-6 in lung tissue homogenates was also reduced by EF24 treatment. These results confirm our previous in vitro observations in lipopolysaccharide-stimulated dendritic cells that EF24 beneficially modulates the IL-1R pathway and suggest that it could be investigated as an adjunct therapeutic in managing inflammation associated with hemorrhagic shock.
Collapse
Affiliation(s)
- Vivek R Yadav
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | | | | |
Collapse
|
243
|
Liu J, Ren Y, Kang L, Zhang L. Overexpression of CCN3 inhibits inflammation and progression of atherosclerosis in apolipoprotein E-deficient mice. PLoS One 2014; 9:e94912. [PMID: 24722330 PMCID: PMC3983261 DOI: 10.1371/journal.pone.0094912] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 03/21/2014] [Indexed: 11/18/2022] Open
Abstract
Background Cysteine-rich 61/connective tissue growth factor/nephroblastoma overexpressed (CCN) 3 has been recently reported to play a role in regulating inflammation of vascular endothelial cells. However, the role of CCN3 in atherosclerosis, which is characterized by vascular inflammation, remains unclear. Hypothesis and Objectives Overexpression of CCN3 may relieve the inflammation response in and inhibit the progress of atherosclerosis. We aimed to explore the potential roles of CCN3 in inflammation in atherosclerosis. Strategy and Main Results In in vitro studies using cultured human aortic endothelial cells and human umbilical vein endothelial cells, CCN3 mRNA and protein expression significantly decreased in response to tumor necrosis factor-α and interleukin-1β treatments (p<0.05), when analyzed by quantitative real-time polymerase chain reaction and Western blot. Using a mouse model of atherosclerosis, the mRNA and protein levels of CCN3 decreased by 72.2% (p = 0.041) and 86.4% (p = 0.036), respectively, compared with levels in wild-type control mice, respectively. Overexpression of CCN3 by adenovirus-mediated gene overexpression decreased low-density lipoprotein cholesterol by 48.9% (p = 0.017), total cholesterol by 58.9% (p = 0.031), and triglycerides by 56.8% (p = 0.022), and it increased high-density lipoprotein cholesterol level by 2.16-fold (p = 0.039), compared with control groups. Additionally, a reduced plaque area and increased fibrous cap were observed (p<0.05). Furthermore, CCN3 overexpression decreased cell adhesion molecule-1 mRNA expression by 84.7% (p = 0.007) and intercellular adhesion molecule-1 mRNA expression by 61.2% (p = 0.044). Inflammatory factors, including matrix metalloproteinases, cyclooxygenase 2, and tissue factor also significantly (p<0.05) decreased with CCN3 overexpression in the atherosclerotic mouse model. Additionally, CCN1 and CCN2, which have been reported to be highly expressed in aortic atherosclerotic plaques, were significantly downregulated (p<0.05) by CCN3 overexpression. Conclusion CCN3 overexpression is associated with control of inflammatory processes and reversion of dyslipidemia in the process of atherosclerosis, which implies that CCN3 may be a promising target in the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Jun Liu
- Department of Geriatrics, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yingang Ren
- Department of Geriatrics, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Li Kang
- Department of Geriatrics, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Lihua Zhang
- Department of Geriatrics, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
- * E-mail:
| |
Collapse
|
244
|
Critical role of TLR2 and MyD88 for functional response of macrophages to a group IIA-secreted phospholipase A2 from snake venom. PLoS One 2014; 9:e93741. [PMID: 24718259 PMCID: PMC3981733 DOI: 10.1371/journal.pone.0093741] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 03/06/2014] [Indexed: 01/10/2023] Open
Abstract
The snake venom MT-III is a group IIA secreted phospholipase A2 (sPLA2) enzyme with functional and structural similarities with mammalian pro-inflammatory sPLA2s of the same group. Previously, we demonstrated that MT-III directly activates the innate inflammatory response of macrophages, including release of inflammatory mediators and formation of lipid droplets (LDs). However, the mechanisms coordinating these processes remain unclear. In the present study, by using TLR2−/− or MyD88−/− or C57BL/6 (WT) male mice, we report that TLR2 and MyD88 signaling have a critical role in MT-III-induced inflammatory response in macrophages. MT-III caused a marked release of PGE2, PGD2, PGJ2, IL-1β and IL-10 and increased the number of LDs in WT macrophages. In MT-III-stimulated TLR2−/− macrophages, formation of LDs and release of eicosanoids and cytokines were abrogated. In MyD88−/− macrophages, MT-III-induced release of PGE2, IL-1β and IL-10 was abrogated, but release of PGD2 and PGJ2 was maintained. In addition, COX-2 protein expression seen in MT-III-stimulated WT macrophages was abolished in both TLR2−/− and MyD88−/− cells, while perilipin 2 expression was abolished only in MyD88−/− cells. We further demonstrated a reduction of saturated, monounsaturated and polyunsaturated fatty acids and a release of the TLR2 agonists palmitic and oleic acid from MT-III-stimulated WT macrophages compared with WT control cells, thus suggesting these fatty acids as major messengers for MT-III-induced engagement of TLR2/MyD88 signaling. Collectively, our findings identify for the first time a TLR2 and MyD88-dependent mechanism that underlies group IIA sPLA2-induced inflammatory response in macrophages.
Collapse
|
245
|
Schenten D, Nish SA, Yu S, Yan X, Lee HK, Brodsky I, Pasman L, Yordy B, Wunderlich FT, Brüning JC, Zhao H, Medzhitov R. Signaling through the adaptor molecule MyD88 in CD4+ T cells is required to overcome suppression by regulatory T cells. Immunity 2014; 40:78-90. [PMID: 24439266 DOI: 10.1016/j.immuni.2013.10.023] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 10/30/2013] [Indexed: 10/25/2022]
Abstract
Innate immune recognition controls adaptive immune responses through multiple mechanisms. The MyD88 signaling adaptor operates in many cell types downstream of Toll-like receptors (TLRs) and interleukin-1 (IL-1) receptor family members. Cell-type-specific functions of MyD88 signaling remain poorly characterized. Here, we have shown that the T cell-specific ablation of MyD88 in mice impairs not only T helper 17 (Th17) cell responses, but also Th1 cell responses. MyD88 relayed signals of TLR-induced IL-1, which became dispensable for Th1 cell responses in the absence of T regulatory (Treg) cells. Treg cell-specific ablation of MyD88 had no effect, suggesting that IL-1 acts on naive CD4(+) T cells instead of Treg cells themselves. Together, these findings demonstrate that IL-1 renders naive CD4(+) T cells refractory to Treg cell-mediated suppression in order to allow their differentiation into Th1 cells. In addition, IL-1 was also important for the generation of functional CD4(+) memory T cells.
Collapse
Affiliation(s)
- Dominik Schenten
- Department of Immunobiology, Yale University, New Haven, CT 06519, USA; Howard Hughes Medical Institute, School of Medicine, Yale University, New Haven, CT 06519, USA.
| | - Simone A Nish
- Department of Immunobiology, Yale University, New Haven, CT 06519, USA; Howard Hughes Medical Institute, School of Medicine, Yale University, New Haven, CT 06519, USA
| | - Shuang Yu
- Department of Immunobiology, Yale University, New Haven, CT 06519, USA; Howard Hughes Medical Institute, School of Medicine, Yale University, New Haven, CT 06519, USA
| | - Xiting Yan
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06520, USA
| | - Heung Kyu Lee
- Department of Immunobiology, Yale University, New Haven, CT 06519, USA; Howard Hughes Medical Institute, School of Medicine, Yale University, New Haven, CT 06519, USA
| | - Igor Brodsky
- Department of Immunobiology, Yale University, New Haven, CT 06519, USA; Howard Hughes Medical Institute, School of Medicine, Yale University, New Haven, CT 06519, USA
| | - Lesley Pasman
- Department of Immunobiology, Yale University, New Haven, CT 06519, USA; Howard Hughes Medical Institute, School of Medicine, Yale University, New Haven, CT 06519, USA
| | - Brian Yordy
- Department of Immunobiology, Yale University, New Haven, CT 06519, USA
| | - F Thomas Wunderlich
- Max Planck Institute for Neurological Research, Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), Institute for Genetics, University of Cologne, 50931 Cologne, Germany
| | - Jens C Brüning
- Max Planck Institute for Neurological Research, Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), Institute for Genetics, University of Cologne, 50931 Cologne, Germany
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06520, USA
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale University, New Haven, CT 06519, USA; Howard Hughes Medical Institute, School of Medicine, Yale University, New Haven, CT 06519, USA.
| |
Collapse
|
246
|
Martinho FC, Leite FR, Chiesa WM, Nascimento GG, Feres M, Gomes BP. Signaling Pathways Activation by Primary Endodontic Infectious Contents and Production of Inflammatory Mediators. J Endod 2014; 40:484-9. [DOI: 10.1016/j.joen.2013.10.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 10/11/2013] [Accepted: 10/18/2013] [Indexed: 11/29/2022]
|
247
|
Voronov E, Carmi Y, Apte RN. The role IL-1 in tumor-mediated angiogenesis. Front Physiol 2014; 5:114. [PMID: 24734023 PMCID: PMC3975103 DOI: 10.3389/fphys.2014.00114] [Citation(s) in RCA: 170] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 03/07/2014] [Indexed: 12/12/2022] Open
Abstract
Tumor angiogenesis is one of the hallmarks of tumor progression and is essential for invasiveness and metastasis. Myeloid inflammatory cells, such as immature myeloid precursor cells, also termed myeloid-derived suppressor cells (MDSCs), neutrophils, and monocytes/macrophages, are recruited to the tumor microenvironment by factors released by the malignant cells that are subsequently “educated” in situ to acquire a pro-invasive, pro-angiogenic, and immunosuppressive phenotype. The proximity of myeloid cells to endothelial cells (ECs) lining blood vessels suggests that they play an important role in the angiogenic response, possibly by secreting a network of cytokines/chemokines and inflammatory mediators, as well as via activation of ECs for proliferation and secretion of pro-angiogenic factors. Interleukin-1 (IL-1) is an “alarm,” upstream, pro-inflammatory cytokine that is generated primarily by myeloid cells. IL-1 initiates and propagates inflammation, mainly by inducing a local cytokine network and enhancing inflammatory cell infiltration to affected sites and by augmenting adhesion molecule expression on ECs and leukocytes. Pro-inflammatory mediators were recently shown to play an important role in tumor-mediated angiogenesis and blocking their function may suppress tumor progression. In this review, we summarize the interactions between IL-1 and other pro-angiogenic factors during normal and pathological conditions. In addition, the feasibility of IL-1 neutralization approaches for anti-cancer therapy is discussed.
Collapse
Affiliation(s)
- Elena Voronov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences and The Cancer Research Center, Ben-Gurion University of the Negev Beer-Sheva, Israel
| | - Yaron Carmi
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences and The Cancer Research Center, Ben-Gurion University of the Negev Beer-Sheva, Israel
| | - Ron N Apte
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences and The Cancer Research Center, Ben-Gurion University of the Negev Beer-Sheva, Israel
| |
Collapse
|
248
|
Kurabi A, Lee J, Wong C, Pak K, Hoffman HM, Ryan AF, Wasserman SI. The inflammasome adaptor ASC contributes to multiple innate immune processes in the resolution of otitis media. Innate Immun 2014; 21:203-14. [PMID: 24652041 DOI: 10.1177/1753425914526074] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
This study was designed to understand the contribution of the inflammasome and IL-1β activation in otitis media (OM). We examined the middle ear (ME) response to non-typeable Haemophilus influenzae (NTHi) in wild type (WT) mice using gene microarrays and a murine model of acute OM. Expression of members of the NOD domain-like receptor family of inflammasome genes was significantly up-regulated early in NTHi infection of the ME, potentially activating specific downstream regulatory cascades that contribute to the proliferative inflammatory response observed during OM. Expression of the pro-forms of the inflammasome targets IL-1β and IL-18 were also up-regulated. To evaluate the role of inflammasome-mediated cytokine maturation, NTHi-induced OM was examined in Asc(-/-)-deficient mice and compared with that seen in WT mice. Mice lacking the Asc gene showed near absence of IL-1β maturation in the ME and a reduction in leukocyte recruitment and infiltration to the cavity, and their macrophages exhibited reduced phagocytosis of NTHi. These inflammatory defects were linked to an increase in the degree and duration of mucosal epithelial hyperplasia in the ME of Asc(-/-) mice, as well as a delay in bacterial clearance from their MEs. These data demonstrate an important role for the inflammasome and cytokine processing in the course and resolution of OM.
Collapse
Affiliation(s)
- Arwa Kurabi
- School of Medicine, Department of Surgery, Division of Otolaryngology, University of California San Diego, La Jolla, CA, USA
| | - Jasmine Lee
- Department of Biology, University of California La Jolla, La Jolla, CA, USA
| | - Chelsea Wong
- Department of Biology, University of California La Jolla, La Jolla, CA, USA
| | - Kwang Pak
- School of Medicine, Department of Surgery, Division of Otolaryngology, University of California San Diego, La Jolla, CA, USA San Diego Veterans Administration Healthcare System, San Diego, CA, USA
| | - Hal M Hoffman
- Department of Medicine, Division of Rheumatology, Allergy and Immunology, University of California San Diego, La Jolla, CA, USA Ludwig Institute of Cancer Research, San Diego Branch, La Jolla, CA, USA
| | - Allen F Ryan
- School of Medicine, Department of Surgery, Division of Otolaryngology, University of California San Diego, La Jolla, CA, USA San Diego Veterans Administration Healthcare System, San Diego, CA, USA
| | - Stephen I Wasserman
- Department of Medicine, Division of Rheumatology, Allergy and Immunology, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
249
|
Steinberg GR, Schertzer JD. AMPK promotes macrophage fatty acid oxidative metabolism to mitigate inflammation: implications for diabetes and cardiovascular disease. Immunol Cell Biol 2014; 92:340-5. [PMID: 24638063 DOI: 10.1038/icb.2014.11] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 02/04/2014] [Accepted: 02/04/2014] [Indexed: 02/07/2023]
Abstract
Metabolic inflammation, a low-grade chronic pro-inflammatory environment in metabolic or vascular tissues during nutrient excess, has emerged as an important factor underpinning the development of type 2 diabetes (T2D) and cardiovascular disease (CVD). Macrophages are a primary source of inflammatory effectors that contribute to insulin resistance and atherosclerosis, the precursors of T2D and CVD, respectively. Oxidative metabolism dictates the inflammatory status of macrophages, effects that may be upstream of endoplasmic reticulum (ER) stress and the NLRP3 inflammasome. The AMP-activated protein kinase (AMPK) lies at the crossroads of metabolically driven macrophage inflammation and exerts control over mitochondrial metabolism, and therefore is vital for dictating the inflammatory status of macrophages. Understanding how AMPK regulates oxidative metabolism and substrate selection to control both ER stress and NLRP3 inflammasome-mediated inflammation holds promise for identifying new therapies and the tailoring of current therapies for the treatment of T2D and CVD.
Collapse
Affiliation(s)
- Gregory R Steinberg
- 1] Faculty of Health Sciences, Department of Medicine, McMaster University, Hamilton, Ontario, Canada [2] Faculty of Health Sciences, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan D Schertzer
- 1] Faculty of Health Sciences, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada [2] Faculty of Health Sciences, Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
250
|
Krumm B, Xiang Y, Deng J. Structural biology of the IL-1 superfamily: key cytokines in the regulation of immune and inflammatory responses. Protein Sci 2014; 23:526-38. [PMID: 24677376 DOI: 10.1002/pro.2441] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 02/07/2014] [Accepted: 02/10/2014] [Indexed: 12/15/2022]
Abstract
Interleukin-1 superfamily of cytokines (IL-1, IL-18, IL-33) play key roles in inflammation and regulating immunity. The mechanisms of agonism and antagonism in the IL-1 superfamily have been pursued by structural biologists for nearly 20 years. New insights into these mechanisms were recently provided by the crystal structures of the ternary complexes of IL-1β and its receptors. We will review here the structural biology related to receptor recognition by IL-1 superfamily cytokines and the regulation of its cytokine activities by antagonists.
Collapse
Affiliation(s)
- Brian Krumm
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, Oklahoma, 74078
| | | | | |
Collapse
|