1
|
Neri P, Aljneibi S, Pichi F. Rescue Treatment with Infliximab for a Bilateral, Severe, Sight Threatening Frosted Branch Angiitis Associated with Concomitant Acute Onset of Presumed Dermatomyositis. Ocul Immunol Inflamm 2022; 31:865-869. [PMID: 35394851 DOI: 10.1080/09273948.2022.2057333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE To report a severe bilateral frosted branch angiitis associated with acute onset of systemic dermatomyositis. METHOD History and clinical examination, laboratory evaluation, fundus' and skin's color pictures, fluorescein angiography (FA), optical coherence tomography (OCT). RESULTS A 32-year-old female was referred for a bilateral frosted branch angiitis and vitreitis, with skin rash, muscle pain, weakness, severe headache, compatible with dermatomyositis. After an initial improvement following an aggressive treatment by systemic steroids and mycophenolate mofetil, she was switched to intravenous infliximab (RemicadeTM, Janssen Biotech, Inc. USA) due to lack of efficacy. Nine months later, her visual acuity was improved up to 20/20 in OU, with both retinal vasculitis and vitreitis being solved. CONCLUSION Dermatomyositis is a severe multiorgan disease which might severely involve eye structures. Anti-TNF-Alpha agents, particularly Infliximab, might offer a rapid control and long-term remission.
Collapse
Affiliation(s)
- Piergiorgio Neri
- The Eye Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE.,Cleveland Clinic Lerner College of Medicine, Case Western Reserve University (USA), Cleveland, Ohio (USA).,College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, UAE
| | | | - Francesco Pichi
- The Eye Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE.,Cleveland Clinic Lerner College of Medicine, Case Western Reserve University (USA), Cleveland, Ohio (USA)
| |
Collapse
|
2
|
Luo C, Zhao J, Chen M, Xu H. The expression of C1 inhibitor (C1INH) in macrophages is upregulated by retinal pigment epithelial cells - implication in subretinal immune privilege in the aging eye. Aging (Albany NY) 2018; 10:1380-1389. [PMID: 29905533 PMCID: PMC6046230 DOI: 10.18632/aging.101474] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 06/07/2018] [Indexed: 04/18/2023]
Abstract
Age-related para-inflammation in the retina-choroidal interface is featured by low-levels of complement activation and subretinal macrophage accumulation. This study aimed to understand how complement expression in macrophages is regulated by retinal pigment epithelium (RPE). Bone marrow-derived macrophages (BMDMs) and RPE cells were cultured from 8-10 weeks old C57BL/6J mice. The BMDMs were co-cultured with normal RPE, or oxidized photoreceptor outer segment (oxPOS) or TNF-α pre-treated RPE, or apoptotic RPE, or RPE-choroid eyecups. Macrophages were then isolated and processed for real-time RT-PCR. The expression of complement inhibitor C1INH in BMDMs was significantly upregulated by RPE and RPE-choroid eyecups. The eyecups also upregulated CFH, CD59a, and Crry in BMDMs. oxPOS pre-treated RPE upregulated C1qb but down-regulated C3 expression in BMDMs. TNF-α pre-treated RPE enhanced C1INH and CFB expression. When BMDMs were treated with apoptotic RPE, the expression of C1qb, CFH, and CD59a was reduced, whereas the expression of C3, CFB and C1INH was increased. Our results suggest that RPE can modulate macrophages complement expression at the retina-choroidal interface even under aging or oxidative conditions. However, during inflammation, they may promote the alternative pathway of complement activation through down-regulating CFH and CD59a and upregulating CFB and C3.
Collapse
Affiliation(s)
- Chang Luo
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen’s University Belfast, Belfast, UK
- AIER Eye Institute, Changsha, China
- AIER School of Ophthalmology, Central South University, Changsha, China
| | - Jiawu Zhao
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen’s University Belfast, Belfast, UK
| | - Mei Chen
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen’s University Belfast, Belfast, UK
| | - Heping Xu
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen’s University Belfast, Belfast, UK
- AIER Eye Institute, Changsha, China
- AIER School of Ophthalmology, Central South University, Changsha, China
| |
Collapse
|
3
|
Inhibition of lymphocyte proliferation: An ability shared by murine mesenchymal stem cells, dermal fibroblasts and chondrocytes. Transpl Immunol 2018; 47:55-61. [DOI: 10.1016/j.trim.2018.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 02/08/2018] [Accepted: 02/12/2018] [Indexed: 02/06/2023]
|
4
|
Dry eye disease and uveitis: A closer look at immune mechanisms in animal models of two ocular autoimmune diseases. Autoimmun Rev 2016; 15:1181-1192. [DOI: 10.1016/j.autrev.2016.09.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 07/08/2016] [Indexed: 12/13/2022]
|
5
|
Dick AD. Doyne lecture 2016: intraocular health and the many faces of inflammation. Eye (Lond) 2016; 31:87-96. [PMID: 27636226 DOI: 10.1038/eye.2016.177] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 07/04/2016] [Indexed: 12/14/2022] Open
Abstract
Dogma for reasons of immune privilege including sequestration (sic) of ocular antigen, lack of lymphatic and immune competent cells in the vital tissues of the eye has long evaporated. Maintaining tissue and cellular health to preserve vision requires active immune responses to prevent damage and respond to danger. A priori the eye must contain immune competent cells, undergo immune surveillance to ensure homoeostasis as well as an ability to promote inflammation. By interrogating immune responses in non-infectious uveitis and compare with age-related macular degeneration (AMD), new concepts of intraocular immune health emerge. The role of macrophage polarisation in the two disorders is a tractable start. TNF-alpha regulation of macrophage responses in uveitis has a pivotal role, supported via experimental evidence and validated by recent trial data. Contrast this with the slow, insidious degeneration in atrophic AMD or in neovasular AMD, with the compelling genetic association with innate immunity and complement, highlights an ability to attenuate pathogenic immune responses and despite known inflammasome activation. Yolk sac-derived microglia maintains tissue immune health. The result of immune cell activation is environmentally dependent, for example, on retinal cell bioenergetics status, autophagy and oxidative stress, and alterations that skew interaction between macrophages and retinal pigment epithelium (RPE). For example, dead RPE eliciting macrophage VEGF secretion but exogenous IL-4 liberates an anti-angiogenic macrophage sFLT-1 response. Impaired autophagy or oxidative stress drives inflammasome activation, increases cytotoxicity, and accentuation of neovascular responses, yet exogenous inflammasome-derived cytokines, such as IL-18 and IL-33, attenuate responses.
Collapse
Affiliation(s)
- A D Dick
- UCL Institute of Ophthalmology, London, UK.,Academic unit of Ophthalmology, School of Clinical Sciences, University of Bristol, Bristol, UK.,National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| |
Collapse
|
6
|
Macrophages and Uveitis in Experimental Animal Models. Mediators Inflamm 2015; 2015:671417. [PMID: 26078494 PMCID: PMC4452861 DOI: 10.1155/2015/671417] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 03/14/2015] [Accepted: 03/31/2015] [Indexed: 11/17/2022] Open
Abstract
Resident and infiltrated macrophages play relevant roles in uveitis as effectors of innate immunity and inductors of acquired immunity. They are major effectors of tissue damage in uveitis and are also considered to be potent antigen-presenting cells. In the last few years, experimental animal models of uveitis have enabled us to enhance our understanding of the leading role of macrophages in eye inflammation processes, including macrophage polarization in experimental autoimmune uveoretinitis and the major role of Toll-like receptor 4 in endotoxin-induced uveitis. This improved knowledge should guide advantageous iterative research to establish mechanisms and possible therapeutic targets for human uveitis resolution.
Collapse
|
7
|
Crawford GL, Boldison J, Copland DA, Adamson P, Gale D, Brandt M, Nicholson LB, Dick AD. The role of lipoprotein-associated phospholipase A2 in a murine model of experimental autoimmune uveoretinitis. PLoS One 2015; 10:e0122093. [PMID: 25874928 PMCID: PMC4398387 DOI: 10.1371/journal.pone.0122093] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Accepted: 02/21/2015] [Indexed: 12/22/2022] Open
Abstract
Macrophage activation is, in part, regulated via hydrolysis of oxidised low density lipoproteins by Lipoprotein-Associated phospholipase A2 (Lp-PLA2), resulting in increased macrophage migration, pro-inflammatory cytokine release and chemokine expression. In uveitis, tissue damage is mediated as a result of macrophage activation; hence inhibition of Lp-PLA2 may limit macrophage activation and protect the tissue. Utilising Lp-PLA2 gene-deficient (KO) mice and a pharmacological inhibitor of Lp-PLA2 (SB-435495) we aimed to determine the effect of Lp-PLA2 suppression in mediating retinal protection in a model of autoimmune retinal inflammation, experimental autoimmune uveoretinitis (EAU). Following immunisation with RBP-3 (IRBP) 1–20 or 161–180 peptides, clinical disease was monitored and severity assessed, infiltrating leukocytes were enumerated by flow cytometry and tissue destruction quantified by histology. Despite ablation of Lp-PLA2 enzyme activity in Lp-PLA2 KO mice or wild-type mice treated with SB-435495, the number of infiltrating CD45+ cells in the retina was equivalent to control EAU animals, and there was no reduction in disease severity. Thus, despite the reported beneficial effects of therapeutic Lp-PLA2 depletion in a variety of vascular inflammatory conditions, we were unable to attenuate disease, show delayed disease onset or prevent progression of EAU in Lp-PLA2 KO mice. Although EAU exhibits inflammatory vasculopathy there is no overt defect in lipid metabolism and given the lack of effect following Lp-PLA2 suppression, these data support the hypothesis that sub-acute autoimmune inflammatory disease progresses independently of Lp-PLA2 activity.
Collapse
Affiliation(s)
- G. L. Crawford
- Academic unit of Ophthalmology, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - J. Boldison
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - D. A. Copland
- Academic unit of Ophthalmology, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - P. Adamson
- Ophthiris Discovery Performance Unit, GlaxoSmithKline, Stevenage, United Kingdom
| | - D. Gale
- Ophthiris Discovery Performance Unit, GlaxoSmithKline, King of Prussia, Pennsylvania, United States of America
| | - M. Brandt
- Platform Technology Sciences, King of Prussia, Pennsylvania, United States of America
| | - L. B. Nicholson
- Academic unit of Ophthalmology, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - A. D. Dick
- Academic unit of Ophthalmology, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
- * E-mail:
| |
Collapse
|
8
|
Nicholls SM, Copland DA, Vitova A, Kuffova L, Forrester JV, Dick AD. Local targeting of the CD200-CD200R axis does not promote corneal graft survival. Exp Eye Res 2014; 130:1-8. [PMID: 25450061 DOI: 10.1016/j.exer.2014.11.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 10/17/2014] [Accepted: 11/10/2014] [Indexed: 12/14/2022]
Abstract
Corneal graft rejection is primarily a CD4(+) T cell-mediated mechanism in which macrophages may play an important inflammatory role. CD200Fc fusion protein is an artificial agonist of CD200R1, a receptor expressed predominantly on myeloid cells, engagement of which is known to down-regulate macrophage function. We therefore wished to test whether CD200Fc could be used as a therapeutic agent to prolong corneal graft survival. The distribution of CD200R1 and CD200, its natural ligand, was examined by immunohistology in the cornea and conjunctiva of unoperated rats and rats that had received corneal allografts. Mouse CD200Fc was injected subconjunctivally into transplanted rats on six occasions from the day of surgery until day 10 after transplantation. Control groups received injections of mouse IgG or diluent PBS. Allo-transplants were also performed in CD200(-/-) and control mice. The ability of CD200Fc to bind rat macrophages in vitro and to inhibit nitric oxide production was tested. Mean day of rejection in CD200Fc, IgG and PBS-treated rats was 12, 10 and 9 respectively (p=0.24). Mean day of rejection in CD200(-/-) and wild type mice was 17.5 and 16.0 respectively (p=0.07). Mouse CD200Fc bound to rat macrophages in a dose-dependent manner, but was unable to inhibit nitric oxide production. The fact that treatment with CD200Fc did not inhibit graft rejection and the failure of CD200 deficiency to affect graft survival suggests that local targeting of the CD200-CD200R axis to suppress macrophage activation is not a useful therapeutic strategy in corneal graft rejection.
Collapse
Affiliation(s)
- Susan M Nicholls
- School of Clinical Sciences, University of Bristol, Bristol, BS8 1TD, UK.
| | - David A Copland
- School of Clinical Sciences, University of Bristol, Bristol, BS8 1TD, UK
| | - Andrea Vitova
- Section of Immunity, Infection and Inflammation (Ocular Immunology), Division of Applied Medicine, School of Medicine and Dentistry, Institute of Medical Sciences, Foresterhill, University of Aberdeen, AB25 2ZD, Scotland, UK
| | - Lucia Kuffova
- Section of Immunity, Infection and Inflammation (Ocular Immunology), Division of Applied Medicine, School of Medicine and Dentistry, Institute of Medical Sciences, Foresterhill, University of Aberdeen, AB25 2ZD, Scotland, UK
| | - John V Forrester
- Section of Immunity, Infection and Inflammation (Ocular Immunology), Division of Applied Medicine, School of Medicine and Dentistry, Institute of Medical Sciences, Foresterhill, University of Aberdeen, AB25 2ZD, Scotland, UK; Ocular Immunology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia, Western Australia, 6009, Australia; Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, 6009, Australia
| | - Andrew D Dick
- School of Clinical Sciences, University of Bristol, Bristol, BS8 1TD, UK; School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK; National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, EC1V 2PD, UK
| |
Collapse
|
9
|
Lee RW, Nicholson LB, Sen HN, Chan CC, Wei L, Nussenblatt RB, Dick AD. Autoimmune and autoinflammatory mechanisms in uveitis. Semin Immunopathol 2014; 36:581-94. [PMID: 24858699 PMCID: PMC4186974 DOI: 10.1007/s00281-014-0433-9] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 04/13/2014] [Indexed: 12/12/2022]
Abstract
The eye, as currently viewed, is neither immunologically ignorant nor sequestered from the systemic environment. The eye utilises distinct immunoregulatory mechanisms to preserve tissue and cellular function in the face of immune-mediated insult; clinically, inflammation following such an insult is termed uveitis. The intra-ocular inflammation in uveitis may be clinically obvious as a result of infection (e.g. toxoplasma, herpes), but in the main infection, if any, remains covert. We now recognise that healthy tissues including the retina have regulatory mechanisms imparted by control of myeloid cells through receptors (e.g. CD200R) and soluble inhibitory factors (e.g. alpha-MSH), regulation of the blood retinal barrier, and active immune surveillance. Once homoeostasis has been disrupted and inflammation ensues, the mechanisms to regulate inflammation, including T cell apoptosis, generation of Treg cells, and myeloid cell suppression in situ, are less successful. Why inflammation becomes persistent remains unknown, but extrapolating from animal models, possibilities include differential trafficking of T cells from the retina, residency of CD8+ T cells, and alterations of myeloid cell phenotype and function. Translating lessons learned from animal models to humans has been helped by system biology approaches and informatics, which suggest that diseased animals and people share similar changes in T cell phenotypes and monocyte function to date. Together the data infer a possible cryptic infectious drive in uveitis that unlocks and drives persistent autoimmune responses, or promotes further innate immune responses. Thus there may be many mechanisms in common with those observed in autoinflammatory disorders.
Collapse
Affiliation(s)
- Richard W Lee
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, University Hospitals Bristol NHS, Foundation Trust, and University of Bristol, Bristol, UK
| | | | | | | | | | | | | |
Collapse
|
10
|
Schewitz-Bowers LP, Lee RWJ, Dick AD. Immune mechanisms of intraocular inflammation. EXPERT REVIEW OF OPHTHALMOLOGY 2014. [DOI: 10.1586/eop.09.68] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
11
|
Aalaei-andabili SH, Rezaei N. Toll like receptor (TLR)-induced differential expression of microRNAs (MiRs) promotes proper immune response against infections: a systematic review. J Infect 2013; 67:251-64. [PMID: 23850616 DOI: 10.1016/j.jinf.2013.07.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 05/25/2013] [Accepted: 07/06/2013] [Indexed: 12/19/2022]
Abstract
Toll like receptors (TLRs) are one of the major families of pattern recognition receptors (PRRs). MicroRNAs (MiRs) are small noncoding RNAs with regulatory effects on biological process, and it has been recently shown that they can control inflammatory process and the response to an infection by modulating the function of TLRs. In this study, we designed a systematic review to clarify the reciprocal interaction between TLRs and MiRs, in order to identify possible future therapeutic targets and strategies. On the one hand, TLRs stimulation can change expression level of miRs in various ways, which can lead to modulating their effects. On the other hand, MiRs also influence the expression of TLRs and the intensity of the inflammatory reaction. We therefore conclude that the interaction between MiRs and TLRs is a key regulator of innate immune system. Investigations discovering therapeutic approaches by manipulation of miRs expression level may open a new approach for the treatment of inflammatory diseases.
Collapse
|
12
|
Chu CJ, Herrmann P, Carvalho LS, Liyanage SE, Bainbridge JWB, Ali RR, Dick AD, Luhmann UFO. Assessment and in vivo scoring of murine experimental autoimmune uveoretinitis using optical coherence tomography. PLoS One 2013; 8:e63002. [PMID: 23690973 PMCID: PMC3653962 DOI: 10.1371/journal.pone.0063002] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 03/27/2013] [Indexed: 12/02/2022] Open
Abstract
Despite advances in clinical imaging and grading our understanding of retinal immune responses and their morphological correlates in experimental autoimmune uveoretinitis (EAU), has been hindered by the requirement for post-mortem histology. To date, monitoring changes occurring during EAU disease progression and evaluating the effect of therapeutic intervention in real time has not been possible. We wanted to establish whether optical coherence tomography (OCT) could detect intraretinal changes during inflammation and to determine its utility as a tool for accurate scoring of EAU. EAU was induced in C57BL/6J mice and animals evaluated after 15, 26, 36 and 60 days. At each time-point, contemporaneous Spectralis-OCT scanning, topical endoscopic fundal imaging (TEFI), fundus fluorescein angiography (FFA) and CD45-immunolabelled histology were performed. OCT features were further characterised on retinal flat-mounts using immunohistochemistry and 3D reconstruction. Optic disc swelling and vitreous opacities detected by OCT corresponded to CD45+ cell infiltration on histology. Vasculitis identified by FFA and OCT matched perivascular myeloid and T-cell infiltrates and could be differentiated from unaffected vessels. Evolution of these changes could be followed over time in the same eye. Retinal folds were visible and found to encapsulate mixed populations of activated myeloid cells, T-cells and microglia. Using these features, an OCT-based EAU scoring system was developed, with significant correlation to validated histological (Pearson r2 = 0.6392, P<0.0001, n = 31 eyes) and TEFI based scoring systems (r2 = 0.6784, P<0.0001). OCT distinguishes the fundamental features of murine EAU in vivo, permits dynamic assessment of intraretinal changes and can be used to score disease severity. As a result, it allows tissue synchronisation with subsequent cellular and functional assessment and greater efficiency of animal usage. By relating OCT signals with immunohistochemistry in EAU, our findings offer the opportunity to inform the interpretation of OCT changes in human uveitis.
Collapse
Affiliation(s)
- Colin J. Chu
- Department of Genetics, UCL Institute of Ophthalmology, London, United Kingdom
| | - Philipp Herrmann
- Department of Genetics, UCL Institute of Ophthalmology, London, United Kingdom
| | - Livia S. Carvalho
- Department of Genetics, UCL Institute of Ophthalmology, London, United Kingdom
| | - Sidath E. Liyanage
- Department of Genetics, UCL Institute of Ophthalmology, London, United Kingdom
| | - James W. B. Bainbridge
- Department of Genetics, UCL Institute of Ophthalmology, London, United Kingdom
- NIHR Biomedical Research Centre for Ophthalmology, Moorfields Eye Hospital, London, United Kingdom
| | - Robin R. Ali
- Department of Genetics, UCL Institute of Ophthalmology, London, United Kingdom
- NIHR Biomedical Research Centre for Ophthalmology, Moorfields Eye Hospital, London, United Kingdom
| | - Andrew D. Dick
- NIHR Biomedical Research Centre for Ophthalmology, Moorfields Eye Hospital, London, United Kingdom
- Unit of Ophthalmology, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
- * E-mail: (UFL); (ADD)
| | - Ulrich F. O. Luhmann
- Department of Genetics, UCL Institute of Ophthalmology, London, United Kingdom
- * E-mail: (UFL); (ADD)
| |
Collapse
|
13
|
Abstract
Noninfectious intraocular inflammatory disease remains a significant cause of visual loss, even with current systemic immunosuppression. Alternative novel treatments are thus required, particularly for severe forms of posterior uveitis. Encouraging results from several phase I/II clinical trials of gene therapy for monogenic retinal disorders have paved the way for the development of this approach for other ocular conditions. Gene therapy for uveitis offers the possibility of potent, self-regulating, long-term disease control following a single treatment and without systemic side effects. To date, gene therapy approaches using interleukin-10, interleukin-1 receptor antagonist, interferon-alpha, soluble TNF-alpha receptors, and alpha-MSH gene transfer have been used successfully to attenuate experimental animal models of uveitis. This review evaluates these preclinical studies, considers the route to clinical application, and explores future targets and approaches.
Collapse
Affiliation(s)
- C J Chu
- University of Bristol, School of Clinical Sciences, Bristol Eye Hospital, Lower Maudlin Street, Bristol, BS1 2LX, UK
| | | | | | | |
Collapse
|
14
|
Fordham JB, Hua J, Morwood SR, Schewitz-Bowers LP, Copland DA, Dick AD, Nicholson LB. Environmental conditioning in the control of macrophage thrombospondin-1 production. Sci Rep 2012; 2:512. [PMID: 22803005 PMCID: PMC3397322 DOI: 10.1038/srep00512] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 06/25/2012] [Indexed: 12/11/2022] Open
Abstract
Thrombospondin-1 (TSP-1) is a multifunctional protein which is secreted into the extracellular matrix during inflammation, where it modulates numerous components of the immune infiltrate. Macrophages are a source of TSP-1, which they produce in response to TLR4 mediated signals. Their production of TSP-1 is regulated by environmental signals that establish a threshold for the level of protein secretion that can be induced by LPS stimulation. Th1 and Th2 cytokines raise this threshold which leads to less TSP-1 production, while signals that promote the generation of regulatory macrophages lower it. TSP-1 plays no direct role in the regulation of its own secretion. In vivo in uveitis, in the presence of TLR-4 ligands, TSP-1 is initially produced by recruited macrophages but this decreases in the presence of inflammatory cytokines. The adaptive immune system therefore plays a dominant role in regulating TSP-1 production in the target organ during acute inflammation.
Collapse
|
15
|
Khera TK, Copland DA, Boldison J, Lait PJP, Szymkowski DE, Dick AD, Nicholson LB. Tumour necrosis factor-mediated macrophage activation in the target organ is critical for clinical manifestation of uveitis. Clin Exp Immunol 2012; 168:165-77. [PMID: 22471277 DOI: 10.1111/j.1365-2249.2012.04567.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Clinically available anti-tumour necrosis factor (TNF) biologics, which inhibit both soluble (sTNF) and transmembrane forms (tmTNF) of TNF, eliminating all TNF signalling, have successfully treated autoimmune diseases including uveitis. These have potentially serious side effects such as reactivation of latent Mycobacterium tuberculosis and, therefore, more specific inhibition of TNF signalling pathways may maintain clinical efficacy while reducing adverse effects. To determine the effects of specific pharmacological inhibition of sTNF on macrophage activation and migration, we used a mouse model of uveitis (experimental autoimmune uveoretinitis; EAU). We show that selective inhibition of sTNF is sufficient to suppress EAU by limiting inflammatory CD11b(+) macrophages and CD4(+) T cell migration into the eye. However, inhibition of both sTNF and tmTNF is required to inhibit interferon-γ-induced chemokine receptor 2, CD40, major histocompatibility complex class II and nitric oxide (NO) up-regulation, and signalling via tmTNF is sufficient to mediate tissue damage. In confirmation, intravitreal inhibition of sTNF alone did not suppress disease, and inflammatory cells that migrated into the eye were activated, generating NO, thus causing structural damage to the retina. In contrast, intravitreal inhibition of both sTNF and tmTNF suppressed macrophage activation and therefore disease. We conclude that sTNF is required for inflammatory cell infiltration into target tissue, but at the tissue site inhibition of both sTNF and tmTNF is required to inhibit macrophage activation and to protect from tissue damage.
Collapse
Affiliation(s)
- T K Khera
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK.
| | | | | | | | | | | | | |
Collapse
|
16
|
Fromm PD, Kling J, Mack M, Sedgwick JD, Körner H. Loss of TNF signaling facilitates the development of a novel Ly-6C(low) macrophage population permissive for Leishmania major infection. THE JOURNAL OF IMMUNOLOGY 2012; 188:6258-66. [PMID: 22615203 DOI: 10.4049/jimmunol.1100977] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In the absence of TNF, the normally resistant C57BL/6 (B6.WT) strain develops a fatal, progressive form of leishmaniasis after infection with Leishmania major. It is not yet understood which TNF activity or the lack thereof is responsible for the dramatic progression of leishmaniasis in TNF-negative (B6.TNF(-/-)) mice. To elucidate the underlying mechanisms resulting in the fatal outcome of L. major infection in this gene-deficient mouse strain, we analyzed the monocytic component of the inflammatory infiltrate in the draining popliteal lymph node and the site of the infection using multicolor flow cytometry. The leukocytic infiltrate within the draining lymph node and footpad of B6.TNF(-/-) mice resembled that of B6.WT mice over the first 2 wk of cutaneous L. major infection. Thereafter, the B6.TNF(-/-) mice showed an increase of CD11c(+)Ly-6C(+)CCR2(+) monocytic dendritic cells within the popliteal lymph node in comparison with B6.WT mice. This increase of inflammatory dendritic cells was paired with the accumulation of a novel CD11b(+)Ly-6C(low)CCR2(low) population that was not present in B6.WT mice. This B6.TNF(-/-)- and B6.TNFR1(-/-)-specific cell population was CD115(+)Ly-6G(-)iNOS(-), not apoptotic, and harbored large numbers of parasites.
Collapse
Affiliation(s)
- Phillip D Fromm
- ANZAC Research Institute, Concord Hospital, Sydney, New South Wales 2039, Australia
| | | | | | | | | |
Collapse
|
17
|
Calder CJ, Wang ECY. An essential role for death receptor 3 in experimental autoimmune uveoretinitis. Ocul Immunol Inflamm 2012; 20:212-4. [PMID: 22486262 DOI: 10.3109/09273948.2012.658135] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
18
|
Portillo JAC, Feliciano LM, Okenka G, Heinzel F, Subauste MC, Subauste CS. CD40 and tumour necrosis factor-α co-operate to up-regulate inducuble nitric oxide synthase expression in macrophages. Immunology 2012; 135:140-50. [PMID: 22044243 DOI: 10.1111/j.1365-2567.2011.03519.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Regulation of inducible nitric oxide synthase (NOS2) expression is important given the role of this enzyme in inflammation, control of infections and immune regulation. In contrast to tumour necrosis factor-α (TNF-α) alone or CD40 stimulation alone, simultaneous stimulation of mouse macrophages through CD40 ligation and TNF-α led to up-regulation of NOS2 and nitric oxide production. This response was of functional relevance because CD40/TNF-α-stimulated macrophages acquired nitric oxide-dependent anti-Leishmania major activity. CD40 plus TNF-α up-regulated NOS2 independently of interferon-γ, interferon-α/β and interleukin-1. TNF receptor-associated factor 6 (TRAF6), an adapter protein downstream of CD40, appears to be required for NOS2 up-regulation because a CD40-TRAF6 blocking peptide inhibited up-regulation of NOS2 in CD40/TNF-α-stimulated macrophages. CCAAT/enhancer-binding protein-β (C/EBPβ), a transcription factor activated by TNF-α but not CD40, was required for NOS2 up-regulation because this enzyme was not up-regulated when C/EBPβ(-/-) macrophages received CD40 plus TNF-α stimulation. These results indicate that CD40 and TNF-α co-operate to up-regulate NOS2, probably via the effect of TRAF6 and C/EBPβ.
Collapse
Affiliation(s)
- Jose-Andres C Portillo
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH OH 44106, USA
| | | | | | | | | | | |
Collapse
|
19
|
Willermain F, Rosenbaum JT, Bodaghi B, Rosenzweig HL, Childers S, Behrend T, Wildner G, Dick AD. Interplay between innate and adaptive immunity in the development of non-infectious uveitis. Prog Retin Eye Res 2012; 31:182-94. [PMID: 22120610 PMCID: PMC3288447 DOI: 10.1016/j.preteyeres.2011.11.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 11/10/2011] [Accepted: 11/11/2011] [Indexed: 12/14/2022]
Abstract
In vertebrates, the innate and adaptive immune systems have evolved seamlessly to protect the host by rapidly responding to danger signals, eliminating pathogens and creating immunological memory as well as immunological tolerance to self. The innate immune system harnesses receptors that recognize conserved pathogen patterns and alongside the more specific recognition systems and memory of adaptive immunity, their interplay is evidenced by respective roles during generation and regulation of immune responses. The hallmark of adaptive immunity which requires engagement of innate immunity is an ability to discriminate between self and non-self (and eventually between pathogen and symbiont) as well as peripheral control mechanisms maintaining immunological health and appropriate responses. Loss of control mechanisms and/or regulation of either the adaptive or the innate immune system lead to autoimmunity and autoinflammation respectively. Although autoimmune pathways have been largely studied to date in the context of development of non-infectious intraocular inflammation, the recruitment and activation of innate immunity is required for full expression of the varied phenotypes of non-infectious uveitis. Since autoimmunity and autoinflammation implicate different molecular pathways, even though some convergence occurs, increasing our understanding of their respective roles in the development of uveitis will highlight treatment targets and influence our understanding of immune mechanisms operative in other retinal diseases. Herein, we extrapolate from the basic mechanisms of activation and control of innate and adaptive immunity to how autoinflammatory and autoimmune pathways contribute to disease development in non-infectious uveitis patients.
Collapse
Affiliation(s)
- François Willermain
- Department of Ophthalmology, CHU St-Pierre and Brugmann, Université Libre de Bruxelles, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Makhoul M, Bruyns C, Edimo WE, Relvas LJ, Bazewicz M, Koch P, Caspers L, Willermain F. TNFα suppresses IFNγ-induced MHC class II expression on retinal pigmented epithelial cells cultures. Acta Ophthalmol 2012; 90:e38-42. [PMID: 21957872 DOI: 10.1111/j.1755-3768.2011.02241.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE One major consequence of retinal pigment epithelium (RPE) cell activation during autoimmune uveitis is the induction of MHC II molecules expression at their surface. IFNγ is regarded as the main cytokine involved in this induction. As TNFα plays a central role in autoimmune uveitis, we investigated its effects on IFNγ-mediated MHC II induction on RPE cells. METHODS Retinal pigment epithelium cells (ARPE-19) were stimulated with IFNγ, TNFα and the anti-TNFα antibody infliximab. The expression of MHCII and ICAM-1 was analysed by flow cytometry. The activation and expression of IRF-1 and STAT-1, two proteins involved in IFNγ-signalling pathway, were analysed by WB. Class II transactivator (CIITA) expression was monitored by qRT-PCR and immunoprecipitation. RESULTS TNFα inhibits IFNγ-induced MHC II expression on ARPE cells in a dose-dependent manner. Infliximab completely reverses the inhibitory effect of TNFα. We did not observe an inhibitory effect of TNFα on the expression of ICAM-1 induced by IFNγ. Similarly, IFNγ-induced STAT1 phosphorylation and IRF1 expression were not affected by TNFα. On the contrary, we found that TNFα suppresses IFNγ-induced CIITA mRNA accumulation and protein expression. CONCLUSION TNFα inhibits IFNγ-induced MHC II expression in RPE cells. This inhibitory effect was reversed by infliximab and was not because of a global inhibition of IFNγ -mediated RPE cell activation but rather to a specific down-regulation of CIITA expression. Those findings are consistent with the role of TNFα in the resolution of inflammation and might help to elucidate the complex development of autoimmune uveitis.
Collapse
Affiliation(s)
- Maya Makhoul
- IRIBHM (Institute of Interdisciplinary Research), Université Libre De Bruxelles-Campus Erasme, Brussels, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Therapeutic dosing of fingolimod (FTY720) prevents cell infiltration, rapidly suppresses ocular inflammation, and maintains the blood-ocular barrier. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 180:672-81. [PMID: 22119714 DOI: 10.1016/j.ajpath.2011.10.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 09/14/2011] [Accepted: 10/11/2011] [Indexed: 01/23/2023]
Abstract
Fingolimod (FTY720) is an FDA-approved therapeutic drug with efficacy demonstrated in experimental models of multiple sclerosis and in phase III human multiple sclerosis trials. Fingolimod prevents T-cell migration to inflammatory sites by decreasing expression of the sphingosine-1 phosphate receptor normally required for egress from secondary lymphoid tissue. As a preclinical model of human uveitis, experimental autoimmune uveoretinitis permits assessment of immunotherapeutic efficacy. Murine experimental autoimmune uveoretinitis is induced by activation of retinal antigen-specific CD4(+) T cells that infiltrate the eye. High-dose fingolimod treatment administered before disease onset reduces ocular infiltration within hours of administration and suppresses clinicopathologic expression of experimental autoimmune uveoretinitis. In the present investigation of the efficacy of fingolimod treatment for established disease, single-dose treatment was effective and immunosuppressive ability was maintained through a dose range, demonstrating significant and rapid reduction in CD4(+) cell infiltration at clinically relevant therapeutic doses of fingolimod. A repeated-treatment regimen using a dose similar to that in current multiple sclerosis patient protocols significantly reduced infiltration within 24 hours of administration; importantly, repeated doses did not compromise the vascular integrity of the blood-ocular barrier. On withdrawal of fingolimod, drug-induced remission was lost and recrudescence of clinical disease was observed. These results support a strong therapeutic potential for fingolimod as an acute rescue therapy for the treatment of ocular immune-mediated inflammation.
Collapse
|
22
|
Vinay DS, Kwon BS. The tumour necrosis factor/TNF receptor superfamily: therapeutic targets in autoimmune diseases. Clin Exp Immunol 2011; 164:145-57. [PMID: 21401577 DOI: 10.1111/j.1365-2249.2011.04375.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Autoimmune diseases are characterized by the body's ability to mount immune attacks on self. This results from recognition of self-proteins and leads to organ damage due to increased production of pathogenic inflammatory molecules and autoantibodies. Over the years, several new potential therapeutic targets have been identified in autoimmune diseases, notable among which are members of the tumour necrosis factor (TNF) superfamily. Here, we review the evidence that certain key members of this superfamily can augment/suppress autoimmune diseases.
Collapse
Affiliation(s)
- D S Vinay
- Section of Clinical Immunology, Allergy, and Rheumatology, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA, USA
| | | |
Collapse
|
23
|
Raveney BJE, Copland DA, Calder CJ, Dick AD, Nicholson LB. TNFR1 signalling is a critical checkpoint for developing macrophages that control of T-cell proliferation. Immunology 2011; 131:340-9. [PMID: 20518824 DOI: 10.1111/j.1365-2567.2010.03305.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Macrophages (Mϕ) are professional antigen-presenting cells, but when they accumulate at sites of inflammation, they can inhibit T-cell proliferation. In experimental autoimmune uveoretinitis, this limits the expansion of T cells within the target organ. To define requirements for the elaboration of this outcome, we have generated populations of Mϕ in vitro that could also regulate T-cell responses; stimulating CD4(+) T-cell activation and cytokine production, but simultaneously suppressing T-cell proliferation. When T cells are removed from the influence of such cells, normal T-cell responses are restored. We show that tumour necrosis factor 1 (TNFR1) signalling is a critical checkpoint in the development of such Mϕ, as TNFR1(-/-) Mϕ are unable to suppress T-cell proliferation. This deficit in antigen-presenting cells results in a lack of production of prostaglandin E(2) (PGE(2)) and nitric oxide, which are critical effector mechanisms that inhibit T-cell division. However, TNFR1 signalling is not required for the inhibitory function of Mϕ because we could circumvent the requirement for this receptor, by maturing Mϕ in the presence of exogenous interferon-γ and PGE(2). This produced TNFR1(-/-) Mϕ that inhibited T-cell proliferation and indicates that TNFR1 delivers a signal that is necessary for the development but not the execution of this function.
Collapse
Affiliation(s)
- Ben J E Raveney
- Department of Cellular and Molecular Medicine, School of Medical Sciences, Bristol, UK
| | | | | | | | | |
Collapse
|
24
|
Khera TK, Dick AD, Nicholson LB. Mechanisms of TNFα regulation in uveitis: Focus on RNA-binding proteins. Prog Retin Eye Res 2010; 29:610-21. [DOI: 10.1016/j.preteyeres.2010.08.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
25
|
McLaren JE, Michael DR, Salter RC, Ashlin TG, Calder CJ, Miller AM, Liew FY, Ramji DP. IL-33 reduces macrophage foam cell formation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 185:1222-9. [PMID: 20543107 DOI: 10.4049/jimmunol.1000520] [Citation(s) in RCA: 142] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2025]
Abstract
The development of atherosclerosis, a chronic inflammatory disease characterized by the formation of arterial fibrotic plaques, has been shown to be reduced by IL-33 in vivo. However, whether IL-33 can directly affect macrophage foam cell formation, a key feature of atherosclerotic plaques, has not been determined. In this study, we investigated whether IL-33 reduces macrophage foam cell accumulation in vivo and if IL-33 reduces their formation in vitro using THP-1 and primary human monocyte-derived macrophages. In Apolipoprotein E(-/-) mice fed on a high fat diet, IL-33 treatment significantly reduced the accumulation of macrophage-derived foam cells in atherosclerotic plaques. IL-33 also reduced macrophage foam cell formation in vitro by decreasing acetylated and oxidized low-density lipoprotein uptake, reducing intracellular total and esterified cholesterol content and enhancing cholesterol efflux. These changes were associated with IL-33-mediated reduction in the expression of genes involved in modified low-density lipoprotein uptake, such as CD36, and simultaneous increase in genes involved in cholesterol efflux, including Apolipoprotein E, thereby providing a mechanism for such an action for this cytokine. IL-33 also decreased the expression of key genes implicated in cholesterol esterification and triglyceride storage, including Acyl-CoA:cholesterol acyltransferase 1 and Adipocyte differentiation-related protein. Furthermore, using bone marrow-derived macrophages from ST2(-/-) mice, we demonstrate that the IL-33 receptor, ST2, is integral to the action of IL-33 on macrophage foam cell formation. In conclusion, IL-33 has a protective role in atherosclerosis by reducing macrophage foam cell formation suggesting that IL-33 maybe a potential therapeutic agent against atherosclerosis.
Collapse
Affiliation(s)
- James E McLaren
- Cardiff School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
26
|
McLaren JE, Calder CJ, McSharry BP, Sexton K, Salter RC, Singh NN, Wilkinson GWG, Wang ECY, Ramji DP. The TNF-like protein 1A-death receptor 3 pathway promotes macrophage foam cell formation in vitro. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:5827-34. [PMID: 20410491 PMCID: PMC2874519 DOI: 10.4049/jimmunol.0903782] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
TNF-like protein 1A (TL1A), a TNF superfamily cytokine that binds to death receptor 3 (DR3), is highly expressed in macrophage foam cell-rich regions of atherosclerotic plaques, although its role in foam cell formation has yet to be elucidated. We investigated whether TL1A can directly stimulate macrophage foam cell formation in both THP-1 and primary human monocyte-derived macrophages with the underlying mechanisms involved. We demonstrated that TL1A promotes foam cell formation in human macrophages in vitro by increasing both acetylated and oxidized low-density lipoprotein uptake, by enhancing intracellular total and esterified cholesterol levels and reducing cholesterol efflux. This imbalance in cholesterol homeostasis is orchestrated by TL1A-mediated changes in the mRNA and protein expression of several genes implicated in the uptake and efflux of cholesterol, such as scavenger receptor A and ATP-binding cassette transporter A1. Furthermore, through the use of virally delivered DR3 short-hairpin RNA and bone marrow-derived macrophages from DR3 knockout mice, we demonstrate that DR3 can regulate foam cell formation and contributes significantly to the action of TL1A in this process in vitro. We show, for the first time, a novel proatherogenic role for both TL1A and DR3 that implicates this pathway as a target for the therapeutic intervention of atherosclerosis.
Collapse
Affiliation(s)
- James E. McLaren
- Cardiff School of Biosciences Cardiff University, Cardiff, United Kingdom
| | - Claudia J. Calder
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Brian P. McSharry
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Keith Sexton
- Cardiff School of Biosciences Cardiff University, Cardiff, United Kingdom
| | - Rebecca C. Salter
- Cardiff School of Biosciences Cardiff University, Cardiff, United Kingdom
| | - Nishi N. Singh
- Cardiff School of Biosciences Cardiff University, Cardiff, United Kingdom
| | - Gavin W. G. Wilkinson
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Eddie C. Y. Wang
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Dipak P. Ramji
- Cardiff School of Biosciences Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
27
|
Sharma SM, Nestel AR, Lee RWJ, Dick AD. Clinical review: Anti-TNFalpha therapies in uveitis: perspective on 5 years of clinical experience. Ocul Immunol Inflamm 2010; 17:403-14. [PMID: 20001261 DOI: 10.3109/09273940903072443] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Despite a lack of robust evidence, anti-TNF therapies are in wide use for the treatment of noninfectious ocular inflammatory diseases. There is a clear rationale, based on mechanistic and preclinical efficacy data, for their use in posterior segment intraocular inflammation. However, their increasing use for other indications has been largely extrapolated from the benefit observed in autoinflammatory and autoimmune systemic diseases. Given their cost and the potential for significant adverse events, this review highlights the evidence for their continued use, possibilities for switching anti-TNF agents, and ways of reducing the risk of therapy.
Collapse
|
28
|
Iwata D, Kitamura M, Kitaichi N, Saito Y, Kon S, Namba K, Morimoto J, Ebihara A, Kitamei H, Yoshida K, Ishida S, Ohno S, Uede T, Onoé K, Iwabuchi K. Prevention of experimental autoimmune uveoretinitis by blockade of osteopontin with small interfering RNA. Exp Eye Res 2009; 90:41-8. [PMID: 19766630 DOI: 10.1016/j.exer.2009.09.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2008] [Revised: 09/07/2009] [Accepted: 09/09/2009] [Indexed: 01/13/2023]
Abstract
Osteopontin (OPN) is elevated during the progression of experimental autoimmune uveoretinitis (EAU) in C57BL/6 (B6) mice. Furthermore, EAU symptoms are ameliorated in OPN knockout mice or in B6 mice treated with anti-OPN antibody (M5). Recently, OPN has been shown to promote the Th1 response not only in the extracellular space as a secretory protein but also in cytosol as a signaling component. Thus, we attempted to reduce OPN in both compartments by using a small interfering RNA (siRNA) targeting the OPN coding sequence (OPN-siRNA). EAU was induced in B6 mice by immunization with human interphotoreceptor retinoid-binding protein (hIRBP) peptide sequence 1-20. The OPN- or control-siRNA was administered with hydrodynamic methods 24 h before and simultaneously with immunization (prevention regimen). When plasma OPN levels were quantified following siRNA administration with the prevention regimen, the level in the OPN-siRNA-treated group was significantly lower than that in the control-siRNA-treated group. Accordingly, the clinical and histopathological scores of EAU were significantly reduced in B6 mice when siRNA caused OPN blockade. Furthermore, TNF-alpha, IFN-gamma, IL-2, GM-CSF and IL-17 levels in the culture supernatants were markedly suppressed in the OPN-siRNA-treated group, whereas the proliferative responses of T lymphocytes from regional lymph nodes against immunogenic peptides was not significantly reduced. On the other hand, the protection was not significant if the mice received the OPN-siRNA treatment on day 7 and day 8 after immunization when the clinical symptoms appeared overt (reversal regimen). Our results suggest that OPN blockade with OPN-siRNA can be an alternative choice for the usage of anti-OPN antibody and controlling uveoretinitis in the preventive regimen.
Collapse
Affiliation(s)
- Daiju Iwata
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo 060-0815, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Raveney BJE, Copland DA, Dick AD, Nicholson LB. TNFR1-dependent regulation of myeloid cell function in experimental autoimmune uveoretinitis. THE JOURNAL OF IMMUNOLOGY 2009; 183:2321-9. [PMID: 19635911 DOI: 10.4049/jimmunol.0901340] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Experimental autoimmune uveoretinitis is an autoimmune disease induced in mice, which involves the infiltration of CD11b(+) macrophages and CD4(+) T cells into the normally immune-privileged retina. Damage is produced in the target organ following the activation of Th1 and Th17 T cells and by the release of cytotoxic mediators such as NO by activated macrophages. The majority of immune cells infiltrating into the retina are CD11b(+) myeloid cells, but, despite the presence of these APCs, relatively limited numbers of T cells are observed in the retina during the disease course. These T cells do not proliferate when leukocytes are isolated from the retina and restimulated in vitro, although they do produce both IFN-gamma and IL-17. T cell proliferation was restored by depleting the myeloid cells from the cultures and furthermore those isolated myeloid cells were able to regulate the proliferation of other T cells. The ability of macrophages to regulate proliferation depends on activation by T cell-produced IFN-gamma and autocrine TNF-alpha signaling in the myeloid cells via TNFR1. In the absence of TNFR1 signaling, relative T cell expansion in the retina is increased, indicating that regulatory myeloid cells may also act in vivo. However, TNFR1 signaling is also required for macrophages, but not T cells, to migrate into the target organ. Thus, in TNFR1 knock out mice, the amplification of autoimmunity is limited, leading to resistance to experimental autoimmune uveoretinitis induction.
Collapse
Affiliation(s)
- Ben J E Raveney
- Department of Cellular and Molecular Medicine, School of Medical Sciences, University of Bristol, Bristol, United Kingdom
| | | | | | | |
Collapse
|
30
|
Masli S, Turpie B. Anti-inflammatory effects of tumour necrosis factor (TNF)-alpha are mediated via TNF-R2 (p75) in tolerogenic transforming growth factor-beta-treated antigen-presenting cells. Immunology 2009; 127:62-72. [PMID: 18795974 DOI: 10.1111/j.1365-2567.2008.02933.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Exposure of macrophages to transforming growth factor (TGF)-beta is known to alter their functional phenotype such that antigen presentation by these cells leads to tolerance rather than an inflammatory immune response. Typically, eye-derived antigen-presenting cells (APCs) exposed to TGF-beta in the local environment are known to induce a form of peripheral tolerance and protect the eye from inflammatory immune effector-mediated damage. In response to TGF-beta, APCs increase their expression of tumour necrosis factor (TNF)-alpha and TNF receptor 2 (TNF-R2). Although TNF-alpha has been implicated in tolerance and the associated regulation of the inflammatory immune response, its source and the receptors involved remain unclear. In this report we determined the contribution of TNF-alpha and TNF-R2 expressed by TGF-beta-treated APCs to their anti-inflammatory tolerogenic effect. Our results indicate that APC-derived TNF-alpha is essential for the ability of APCs to regulate the immune response and their IL-12 secretion. Moreover, in the absence of TNF-R2, APCs exposed to TGF-beta failed to induce tolerance or regulatory cells known to participate in this tolerance. Also, blocking of TNF-R1 signalling enhanced the ability of the APCs to secrete increased TGF-beta in response to TGF-beta exposure. Together our results support an anti-inflammatory role of TNF-alpha in regulation of an immune response by TGF-beta-treated APCs and suggest that TNF-R2 contributes significantly to this role.
Collapse
Affiliation(s)
- Sharmila Masli
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
31
|
Giannandrea M, Pierce RH, Crispe IN. Indirect action of tumor necrosis factor-alpha in liver injury during the CD8+ T cell response to an adeno-associated virus vector in mice. Hepatology 2009; 49:2010-20. [PMID: 19291774 PMCID: PMC2871665 DOI: 10.1002/hep.22869] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
UNLABELLED CD8+ T cells can cause hepatocellular injury by two distinct mechanisms. In addition to their direct cytotoxic effect, there is also collateral liver injury, which occurs when cells are killed in an antigen-independent manner. Whereas immune effector cytokines interferon-gamma (IFNgamma) and tumor necrosis factor-alpha (TNFalpha) have both been implicated in various forms of hepatitis, their respective roles in direct and/or collateral liver damage remains unclear. In order to investigate these elements of liver injury, we developed a new experimental model of CD8+ T-cell-mediated hepatitis based on an adeno-associated virus-based gene therapy vector. This vector is used to deliver antigen to hepatocytes, and CD8+ T cells specific for the vector-encoded transgene are adoptively transferred to produce liver immunopathology. In this experimental model, CD8+ T-cell IFNgamma acts on Kupffer cells, inducing TNFalpha secretion and liver injury. Both IFNgamma and TNFalpha are important in this injury process, but TNFalpha acts as an autocrine amplifier of Kupffer cell function, rather than as a direct effector of hepatocellular damage. CONCLUSIONS TNFalpha indirectly promotes liver damage and is not a direct hepatotoxic agent. IFNgamma also indirectly contributes to liver injury through Kupffer cell activation while, in parallel, directly promoting hepatitis through induction of hepatocyte major histocompatability complex class I. In principle, it may be possible to ameliorate this immunopathologic indirect mechanism by developing therapies that target Kupffer cells, without impairing CD8+ T-cell-mediated antiviral immunity. This would have great therapeutic potential in chronic viral hepatitis.
Collapse
Affiliation(s)
- Matthew Giannandrea
- The David H Smith Center for Vaccine Biology and Immunology, Aab Institute for Biomedical Research, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642, USA
| | - Robert H. Pierce
- Schering-Plough Biopharma, 901 California Ave, Palo Alto, CA 94304, USA.
| | - Ian Nicholas Crispe
- The Seattle Biomedical Research Institute, 307 North Westlake Avenue, Seattle, WA 98019
| |
Collapse
|
32
|
Iwakura Y, Nakae S, Saijo S, Ishigame H. The roles of IL-17A in inflammatory immune responses and host defense against pathogens. Immunol Rev 2009; 226:57-79. [PMID: 19161416 DOI: 10.1111/j.1600-065x.2008.00699.x] [Citation(s) in RCA: 378] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
T-helper 17 (Th17) cells are a newly discovered CD4(+) helper T-cell subset that produces interleukin-17A (IL-17A) and IL-17F. IL-17A plays important roles in allergic responses such as delayed-type hypersensitivity, contact hypersensitivity, and allergic airway inflammation. IL-17A promotes inflammation by inducing various proinflammatory cytokines and chemokines, recruiting neutrophils, enhancing antibody production, and activating T cells. IL-17A expression is also augmented in autoimmune diseases such as multiple sclerosis and rheumatoid arthritis. Using mouse models of these diseases, we found that IL-17A plays a central role in their development. IL-6 is required for the development of Th17 cells and tumor necrosis factor functions downstream of IL-17A during the effector phase. IL-1 is important both for developing Th17 cells and eliciting inflammation. Th17 cells, like Th1 and Th2 cells, are involved in host defense against infections, but the contribution of these Th subsets to defense mechanisms differs among pathogens. The roles of IL-17F remain largely unknown. In this review, we introduce how IL-17A/IL-17F are involved in inflammatory immune responses and host defense mechanisms and discuss their relationship with other cytokines in the development of inflammatory and infectious diseases.
Collapse
Affiliation(s)
- Yoichiro Iwakura
- Center for Experimental Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | | | | | | |
Collapse
|
33
|
Kohno K, Miyake M, Sano O, Tanaka-Kataoka M, Yamamoto S, Koya-Miyata S, Arai N, Fujii M, Watanabe H, Ushio S, Iwaki K, Fukuda S. Anti-inflammatory and immunomodulatory properties of 2-amino-3H-phenoxazin-3-one. Biol Pharm Bull 2008; 31:1938-45. [PMID: 18827359 DOI: 10.1248/bpb.31.1938] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Accumulating evidence suggests that nitric oxide (NO) and prostaglandin E(2) (PGE(2)) are involved in the pathogenesis of various chronic inflammatory diseases and cancer. During the course of a screening program to identify natural anti-inflammatory substances, we isolated the compound 2-amino-3H-phenoxazin-3-one (APO) from an extract of the edible brown mushroom Agaricus bisporus IMBACH. APO inhibited NO production by mouse peritoneal macrophages in response to the pro-inflammatory stimuli lipopolysaccharide (LPS) and interferon (IFN)-gamma (LPS/IFN-gamma) at low concentrations (IC(50)=1.5 microM) through reduced inducible NO synthase protein expression. PGE(2) production by LPS/IFN-gamma-stimulated macrophages was inhibited by APO at much lower concentrations (IC(50)=0.27 microM) than those required for the inhibition of NO production. Mechanistic analysis showed that APO inhibited both cyclooxygenase (COX)-1 and COX-2 enzyme activities with almost equal selectivity. Secretion of NO and the pro-inflammatory cytokine IL-6 by IFN-gamma-activated RAW264.7 cells, a murine macrophage-like cell line, was also dose-dependently reduced by APO. Furthermore, APO increased the secretion of the anti-inflammatory cytokine IL-4 by antigen-stimulated T cells and promoted the polarization of CD4(+) Th cells toward the anti-inflammatory Th2 phenotype at equimolar concentrations that inhibited NO production. Our results suggested that APO induced polarization toward the Th2 subset, at least in part through the down-regulation of IL-12 production. Thus, APO appears to have potent anti-inflammatory and immunoregulatory properties that may provide a promising therapeutic strategy for the treatment of T cell-mediated inflammatory autoimmune diseases as well as for bacteria-induced chronic-inflammatory diseases.
Collapse
Affiliation(s)
- Keizo Kohno
- Biomedical Institute, Research Center, Hayashibara Biochemical Laboratories, Inc, 675-1 Fujisaki, Okayama 702-8006, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Bull MJ, Williams AS, Mecklenburgh Z, Calder CJ, Twohig JP, Elford C, Evans BAJ, Rowley TF, Slebioda TJ, Taraban VY, Al-Shamkhani A, Wang ECY. The Death Receptor 3-TNF-like protein 1A pathway drives adverse bone pathology in inflammatory arthritis. J Exp Med 2008; 205:2457-64. [PMID: 18824582 PMCID: PMC2571920 DOI: 10.1084/jem.20072378] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2007] [Accepted: 09/02/2008] [Indexed: 01/21/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease of synovial joints that is associated with cartilage and bone destruction. Death Receptor 3 (DR3), a tumor necrosis factor (TNF) receptor superfamily member, has recently been associated with the pathogenesis of RA. We demonstrate that absence of DR3 confers resistance to the development of adverse bone pathology in experimental antigen-induced arthritis (AIA). DR3(ko) mice exhibited a reduction in all histopathological hallmarks of AIA but, in particular, failed to develop subchondral bone erosions and were completely protected from this characteristic of AIA. In contrast, TNF-like protein 1A (TL1A), the ligand for DR3, exacerbated disease in a dose- and DR3-dependent fashion. Analysis of osteoclast number within AIA joint revealed a reduction in areas susceptible to bone erosion in DR3(ko) mice, whereas in vitro osteoclastogenesis assays showed that TL1A could directly promote osteoclastogenesis in mouse and man. Treatment with antagonistic anti-TL1A mAb protected animals in a systemic model of RA disease collagen-induced arthritis. We therefore conclude that the DR3-TL1A pathway regulates joint destruction in two murine models of arthritis and represents a potential novel target for therapeutic intervention in inflammatory joint disease.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/pathology
- Bone and Bones/pathology
- DNA Primers/genetics
- Humans
- Immunohistochemistry
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Osteoclasts/cytology
- Osteoclasts/immunology
- Receptors, Tumor Necrosis Factor, Member 25/genetics
- Receptors, Tumor Necrosis Factor, Member 25/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction/immunology
- Statistics, Nonparametric
- Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism
Collapse
Affiliation(s)
- Melanie Jane Bull
- Department of Medical Biochemistry and Immunology, School of Medicine, Heath Park, Cardiff CF14 4XN, Wales, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
|
36
|
Calzascia T, Pellegrini M, Hall H, Sabbagh L, Ono N, Elford AR, Mak TW, Ohashi PS. TNF-alpha is critical for antitumor but not antiviral T cell immunity in mice. J Clin Invest 2008; 117:3833-45. [PMID: 17992258 DOI: 10.1172/jci32567] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2007] [Accepted: 09/12/2007] [Indexed: 12/31/2022] Open
Abstract
TNF-alpha antagonists are widely used in the treatment of inflammatory and autoimmune diseases, but their use is associated with reactivation of latent infections. This highlights the importance of TNF-alpha in immunity to certain pathogens and raises concerns that critical aspects of immune function are impaired in its absence. Unfortunately, the role of TNF-alpha in the regulation of T cell responses is clouded by a myriad of contradictory reports. Here, we show a role for TNF-alpha and its receptors, TNFR1 and TNFR2, specifically in antitumor immunity. TNF-alpha-deficient mice exhibited normal antiviral responses associated with strong inflammation. However, TNF-alpha/TNFR1-mediated signals on APCs and TNF-alpha/TNFR2 signals on T cells were critically required for effective priming, proliferation, and recruitment of tumor-specific T cells. Furthermore, in the absence of TNF-alpha signaling, tumor immune surveillance was severely abrogated. Finally, treatment with a CD40 agonist alone or in combination with TLR2 stimuli was able to rescue proliferation of TNF-alpha-deficient T cells. Therefore, TNF-alpha signaling may be required only for immune responses in conditions of limited immunostimulatory capacity, such as tumor surveillance. Importantly, these results suggest that prolonged continuous TNF-alpha blockade in patients may have long-term complications, including potential tumor development or progression.
Collapse
Affiliation(s)
- Thomas Calzascia
- The Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Copland DA, Calder CJ, Raveney BJE, Nicholson LB, Phillips J, Cherwinski H, Jenmalm M, Sedgwick JD, Dick AD. Monoclonal antibody-mediated CD200 receptor signaling suppresses macrophage activation and tissue damage in experimental autoimmune uveoretinitis. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 171:580-8. [PMID: 17600119 PMCID: PMC1934542 DOI: 10.2353/ajpath.2007.070272] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Macrophage responses are regulated by multiple secreted factors as well as by cell surface receptors, including the inhibitory signals resulting from ligation of myeloid CD200 receptors (CD200R) by the widely distributed CD200. In the absence of CD200, animals display increased susceptibility to autoimmunity and earlier onset aggressive autoimmune disease. In these current experiments, an agonist monoclonal rat anti-mouse CD200R (DX109) antibody delivered a negative signal to bone marrow-derived macrophages, which suppressed interferon (IFN)gamma-mediated nitric oxide (NO) and interleukin-6 production. Experimental autoimmune uveoretinitis (EAU) was used as a model of organ-specific autoimmunity in the eye, a tissue with extensive neuronal and endothelial CD200 expression. In mice lacking CD200 (CD200(-/-)), increased numbers of retina-infiltrating macrophages displaying heightened NO responses were observed during EAU. In addition, we aimed to suppress disease by maintaining tonic suppression of macrophage activation via CD200R. Systemically administered DX109 monoclonal antibody suppressed EAU despite maintained T-cell proliferation and IFNgamma production. Furthermore, locally administered DX109 monoclonal antibody resulted in an earlier resolution of disease. These experiments demonstrate that promoting CD200R-mediated signaling can successfully prevent full expression of IFNgamma-mediated macrophage activation and protect against tissue damage during autoimmune responses.
Collapse
Affiliation(s)
- David A Copland
- Division of Infection and Immunity, Department of Clinical Science at South Bristol, University of Bristol, Bristol, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kitamura M, Iwabuchi K, Kitaichi N, Kon S, Kitamei H, Namba K, Yoshida K, Denhardt DT, Rittling SR, Ohno S, Uede T, Onoé K. Osteopontin Aggravates Experimental Autoimmune Uveoretinitis in Mice. THE JOURNAL OF IMMUNOLOGY 2007; 178:6567-72. [PMID: 17475887 DOI: 10.4049/jimmunol.178.10.6567] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Human endogenous uveitis is a common sight-threatening intraocular inflammatory disease and has been studied extensively using a murine model of experimental autoimmune uveoretinitis (EAU). It is possibly mediated by Th1 immune responses. In the present study, we investigated the role of osteopontin (OPN), a protein with pleiotropic functions that contributes to the development of Th1 cell-mediated immunity. Accompanying EAU progression, OPN was elevated in wild-type (WT) mice that had been immunized with human interphotoreceptor retinoid-binding protein (hIRBP) peptide 1-20. OPN-deficient (OPN-/-) mice showed milder EAU progression in clinical and histopathological scores compared with those of WT mice. The T cells from hIRBP-immunized OPN-/- mice exhibited reduced Ag-specific proliferation and proinflammatory cytokine (TNF-alpha and IFN-gamma) production compared with those of WT T cells. When hIRBP-immunized WT mice were administered M5 Ab reacting to SLAYGLR sequence, a cryptic binding site to integrins within OPN, EAU development was significantly ameliorated. T cells from hIRBP-immunized WT mice showed significantly reduced proliferative responses and proinflammatory cytokine production upon stimulation with hIRBP peptide in the presence of M5 Ab in the culture. Our present results demonstrate that OPN may represent a novel therapeutic target to control uveoretinitis.
Collapse
Affiliation(s)
- Mizuki Kitamura
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Xu H, Dawson R, Forrester JV, Liversidge J. Identification of novel dendritic cell populations in normal mouse retina. Invest Ophthalmol Vis Sci 2007; 48:1701-10. [PMID: 17389502 PMCID: PMC2446435 DOI: 10.1167/iovs.06-0697] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Whether tissue resident or infiltrating antigen-presenting cells (APCs) are involved in modulating immune responses in the retina and initiating inflammation is controversial. In this histologic study, the authors examine the retinas of mice strains with different susceptibility to experimental autoimmune uveoretinitis (EAU) for tissue resident APC. METHODS Retinal wholemounts from normal and inflamed eyes of B10R III, C57BL/6, BALB/c, and ABH Biozii mice were immunostained for APC markers (33D1, CD11c, CD11b, major histocompatibility complex [MHC] class II, F4/80, CD80, CD86, CD205, mPDCA, B220, and GR1) and analyzed by confocal fluorescence microscopy using emission fingerprinting and three-dimensional reconstruction techniques. Hematoxylin and eosin-stained histologic sections were used to evaluate EAU disease scores and to assess outer blood retina barrier (retinal pigment epithelium [RPE]) structures. RESULTS A population of 33D1(+) cells was identified exclusively in the peripheral margins and juxtapapillary areas of the retina in normal, nonimmunized C57BL/6 adult mice. These cells were also MHC class II(high), and their location corresponded to sites of earliest inflammation in EAU. Numbers in the papillary area were very low (less than 10), but this region marked the predominant anatomic site for initiation of inflammation in this moderately susceptible strain. The distribution and phenotype of these cells within the retinas differed between mouse strains exhibiting different disease susceptibility. In EAU-resistant BALB/c mice, many more 33D1(+) dendritic cells were present in the normal retina but were MHC class II(low/-). Conversely, no 33D1(+) or MHC class II (+) dendriform cells could be found in the normal retinas of highly EAU-susceptible B10.RIII mice. CONCLUSIONS A novel population of 33D1(+) DCs was identified in normal mouse retina. The function of these cells remains to be defined, but increased numbers correlate positively with structural abnormalities in the RPE and increased resistance of the strain to EAU.
Collapse
Affiliation(s)
- Heping Xu
- Department of Ophthalmology, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, UK.
| | | | | | | |
Collapse
|
40
|
O'Connell RM, Taganov KD, Boldin MP, Cheng G, Baltimore D. MicroRNA-155 is induced during the macrophage inflammatory response. Proc Natl Acad Sci U S A 2007; 104:1604-9. [PMID: 17242365 PMCID: PMC1780072 DOI: 10.1073/pnas.0610731104] [Citation(s) in RCA: 1474] [Impact Index Per Article: 81.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The mammalian inflammatory response to infection involves the induction of several hundred genes, a process that must be carefully regulated to achieve pathogen clearance and prevent the consequences of unregulated expression, such as cancer. Recently, microRNAs (miRNAs) have emerged as a class of gene expression regulators that has also been linked to cancer. However, the relationship between inflammation, innate immunity, and miRNA expression is just beginning to be explored. In the present study, we use microarray technology to identify miRNAs induced in primary murine macrophages after exposure to polyriboinosinic:polyribocytidylic acid or the cytokine IFN-beta. miR-155 was the only miRNA of those tested that was substantially up-regulated by both stimuli. It also was induced by several Toll-like receptor ligands through myeloid differentiation factor 88- or TRIF-dependent pathways, whereas up-regulation by IFNs was shown to involve TNF-alpha autocrine signaling. Pharmacological inhibition of the kinase JNK blocked induction of miR-155 in response to either polyriboinosinic:polyribocytidylic acid or TNF-alpha, suggesting that miR-155-inducing signals use the JNK pathway. Together, these findings characterize miR-155 as a common target of a broad range of inflammatory mediators. Importantly, because miR-155 is known to function as an oncogene, these observations identify a potential link between inflammation and cancer.
Collapse
Affiliation(s)
- Ryan M. O'Connell
- *Department of Biology, California Institute of Technology, 330 Braun, 1200 East California Boulevard, Pasadena, CA 91125; and
| | - Konstantin D. Taganov
- *Department of Biology, California Institute of Technology, 330 Braun, 1200 East California Boulevard, Pasadena, CA 91125; and
| | - Mark P. Boldin
- *Department of Biology, California Institute of Technology, 330 Braun, 1200 East California Boulevard, Pasadena, CA 91125; and
| | - Genhong Cheng
- Departments of Microbiology, Immunology, and Molecular Genetics, University of California, 650 Charles East Young Drive South, Los Angeles, CA 90095
| | - David Baltimore
- *Department of Biology, California Institute of Technology, 330 Braun, 1200 East California Boulevard, Pasadena, CA 91125; and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|