6 research outputs found

    Cellular Responses of the Retina to West Nile Virus Infection

    Get PDF
    Age-related macular degeneration (AMD) is the leading cause of blindness in the developing world in people aged over 60 years, manifested as a loss of central vision in one or both eyes, with significant morbidity including loss of mobility and depression. This condition involves the degeneration of the macula, and although the exact aetiology of this disease is unknown, various epidemiological studies have shown it to be multifactorial. Current research points towards the involvement of a dysregulated immune system in the pathogenesis and progression of the disease: as the body ages, the immune system increasingly adopts a more inflammatory basal state. However, not all of the aged population develops AMD and it is highly likely that an additional stimulus or stimuli is/are needed to exploit this dysregulated immune environment to initiate this disease. Given the range of pathogens that can infect the retina, we hypothesize that this breaking point could manifest as a chronic inflammation as a result of a low-level infection. West Nile Virus (WNV) is a flavivirus that has come into international prominence ever since its spread into previously WNV-free regions following the 1999 New York outbreak. As several case reports have shown that WNV is capable of infecting the retina, and given its immunopathogenic properties, we believe the virus is a useful tool to model key immune pathways and responses that may be involved in the development and progression of AMD. Of significant interest are the processes involved in the breakdown of the outer blood-retinal barrier (BRB), which is an important step in the progression of AMD from an early stage to a more severe one. Additionally, deciphering and understanding the profile and populations of leukocytes that are recruited during an immunopathic infection in an organ regarded as being immunoprivileged is of great appeal. With this in mind, we set out to investigate the effects of WNV infection on the retinal pigment epithelium (RPE), which comprises the outer BRB. Previously, our laboratory established the WNV BRB model by quantitating various parameters, such as level of infectivity, viral output by WNV-infected RPE and effects of WNV infection on RPE proliferation/migration. The effect of WNV on the extracellular matrix (ECM) production by RPE was also investigated and increases in collagen I, IV and fibronectin were noted. Global ECM production induced a lowered rate of proliferation of RPE seeded on WNV-infected RPE ECM as opposed to mock-infected ECM. A full genome microarray was also undertaken on WNV-infected RPE to analyse differentially regulated gene mRNA production, and increases in several immune genes, as well as genes involved in the stress-response pathway and the TGFβ pathway were found. This current investigation expanded upon these results, and found that WNV infection produces a predominantly CCL5 chemokine response rather than a CCL2 response. Additionally, a lack of TNF production was noted, despite a high initial upregulation of the TNF gene in WNV-infected cells. WNV attenuation was found to be predominantly IFNβ-1-driven, while induction of indoleamine 2,3 dioxygenase activity was induced in part by IFNλ-1 and -2. The effects of WNV infection on RPE barrier integrity was investigated, and an initial increase in infected cells of barrier integrity was observed. Several investigations resulted in a conclusion of a soluble-mediator as the likely mechanism behind this initial increase, and while none of the chemokines tested appeared to contribute to this change, the results suggest that it may be TLR3/RIG-I independent. Finally, establishment of a murine WNV intravitreal model was also undertaken, and several key parameters were determined, including confirmation of WNV-infection of the murine retina, effect of WNV titre on mortality, and histological analysis of the effects of WNV infection on the murine retina. Quantification of the leukocyte profile recruited into the WNV-infected murine retina and choroid revealed significant increases in inflammatory Ly6Chi monocytes, as well as significant differences between immune mice and naïve mice intravitreally infected with WNV, and differences between 2 month old and 5 month old mice. Collectively, these results highlight the importance of the interferon response in both direct and indirect anti-WNV activities and immunomodulation, the changes in outer BRB integrity and possible contributors to its degradation, and the establishment of the murine intravitreal WNV model along with identification of several key leukocytes that are recruited at the peak of infection. These results will help guide further research and highlight possible immune pathways that may contribute to dysregulated inflammatory processes that may occur during the pathogenesis of AMD

    Microarray analysis of gene expression in West Nile virus-infected human retinal pigment epithelium

    No full text
    Purpose: To identify key genes differentially expressed in the human retinal pigment epithelium (hRPE) following lowlevel West Nile virus (WNV) infection. Methods: Primary hRPE and retinal pigment epithelium cell line (ARPE-19) cells were infected with WNV (multiplicity of infection 1). RNA extracted from mock-infected and WNV-infected cells was assessed for differential expression of genes using Affymetrix microarray. Quantitative real-time PCR analysis of 23 genes was used to validate the microarray results. Results: Functional annotation clustering of the microarray data showed that gene clusters involved in immune and antiviral responses ranked highly, involving genes such as chemokine (C-C motif) ligand 2 (CCL2), chemokine (C-C motif) ligand 5 (CCL5), chemokine (C-X-C motif) ligand 10 (CXCL10), and toll like receptor 3 (TLR3). In conjunction with the quantitative real-time PCR analysis, other novel genes regulated by WNV infection included indoleamine 2,3-dioxygenase (IDO1), genes involved in the transforming growth factor-β pathway (bone morphogenetic protein and activin membrane-bound inhibitor homolog [BAMBI] and activating transcription factor 3 [ATF3]), and genes involved in apoptosis (tumor necrosis factor receptor superfamily, member 10d [TNFRSF10D]). WNV-infected RPE did not produce any interferon-γ, suggesting that IDO1 is induced by other soluble factors, by the virus alone, or both. Conclusions: Low-level WNV infection of hRPE cells induced expression of genes that are typically associated with the host cell response to virus infection. We also identified other genes, including IDO1 and BAMBI, that may influence the RPE and therefore outer blood-retinal barrier integrity during ocular infection and inflammation, or are associated with degeneration, as seen for example in aging

    Homeostatic IL-13 in healthy skin directs dendritic cell differentiation to promote TH2 and inhibit TH17 cell polarization

    No full text
    The signals driving the adaptation of type 2 dendritic cells (DC2s) to diverse peripheral environments remain mostly undefined. We show that differentiation of CD11blo migratory DC2s-a DC2 population unique to the dermis-required IL-13 signaling dependent on the transcription factors STAT6 and KLF4, whereas DC2s in lung and small intestine were STAT6-independent. Similarly, human DC2s in skin expressed an IL-4 and IL-13 gene signature that was not found in blood, spleen and lung DCs. In mice, IL-13 was secreted homeostatically by dermal innate lymphoid cells and was independent of microbiota, TSLP or IL-33. In the absence of IL-13 signaling, dermal DC2s were stable in number but remained CD11bhi and showed defective activation in response to allergens, with diminished ability to support the development of IL-4+GATA3+ helper T cells (TH), whereas antifungal IL-17+RORγt+ TH cells were increased. Therefore, homeostatic IL-13 fosters a noninflammatory skin environment that supports allergic sensitization.</p
    corecore