9
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Müller Cell–Microglia Cross Talk Drives Neuroinflammation in Diabetic Retinopathy

      article-commentary
      Diabetes
      American Diabetes Association

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Diabetic retinopathy (DR) is the most common complication of diabetes and a leading cause of vision loss worldwide (1). Unfortunately, there are no treatments targeting early stages of the disease prior to the onset of sight-threatening vascular defects such as macular edema or neovascularization. A better understanding of the etiology of DR is needed to identify therapeutic targets to halt early disease progression. To this end, numerous studies demonstrated that a low-grade inflammation occurs in retinas of diabetic animal models and suggest that inflammation contributes a role in DR progression. Various mechanisms leading to retinal inflammation in DR have been described, with the majority of studies implicating retinal Müller glial cells and microglia as the initiators of retinal inflammation (for review, see ref. 2). However, seldom does a study make a strong connection between these two cell types. In this issue of Diabetes, Portillo et al. (3) describe a mechanism in which retinal inflammation in diabetic mice is dependent upon expression of the cluster of differentiation gene 40 (CD40) receptor by Müller cells (Fig. 1). The study suggests that CD40 activation induces Müller cells to release ATP, leading to activation of P2X7 purinergic receptors on retinal microglia and their subsequent expression of inflammatory cytokines. Importantly, the requirement of Müller cell–specific CD40 expression to recapitulate the appearance of acellular capillaries in diabetic retinas also suggests that inflammation is necessary for the loss of vascular cells associated with DR pathology. Figure 1 Proposed role of CD40 and P2X7 receptors in diabetic retinal neuroinflammation. The CD40 receptor on Müller cells (yellow) is activated in retinas of diabetic mice, presumably by binding to CD40L. CD40 activation triggers phospholipase Cγ1 (PLC) activation, leading to an increase in intracellular calcium (Ca2+) resulting in release of ATP. CD40 activation also causes Müller cells to express the chemokine CCL2. Extracellular ATP activates P2X7 receptors on a subset of retinal microglia (green), which is necessary for their expression of the cytokines TNF-α and IL-1β. Thus, CD40 activation on Müller cells links macroglial and microglial inflammatory responses in DR. CD40 is mainly known as an immune costimulatory molecule, and interactions between CD40 and its major ligand (CD40L) play key roles in immunological licensing of antigen-presenting cells by CD4+ T cells and for B-cell activation, proliferation, class switching, and immunoglobin production (4). In a previous study Portillo et al. (5) found that germline deletion of the CD40 gene blocked intracellular adhesion molecule 1 expression, leukostasis, and the appearance of acellular capillaries in the retinas of diabetic mice. These authors also found that in the retina Müller glial cells express CD40, as do endothelial cells, microglia, and retinal ganglion cells (RGC) (6). Since CD40 was deleted in all cells of the germline knockout mice, including circulating immune cells, the mechanism and cell type in which CD40 contributed to DR pathology were unclear. In the current study CD40 was “added back” to the knockout mice in such a way that it was expressed exclusively by Müller cells, which was sufficient to restore the diabetes-induced inflammation and vascular pathology. Müller cell–targeted transgenic add-back of CD40 represents an elegant means of testing the hypothesis that CD40 expression by this cell type is sufficient for low-grade retinal inflammation in this diabetic model. However, the choice of targeting these glial cells is not obvious. Müller cells span radially across retina layers providing structural, metabolic, and neurotrophic support necessary for homeostasis (7). CD40 is an immune costimulatory molecule, and a role for CD40–CD40L interaction in Müller cell pathophysiology is unprecedented. Furthermore, one might have expected leukostasis and capillary dropout in the diabetic retina to be dependent on CD40 expression on endothelial cells, since the interaction of luminal CD40 with CD40L on activated platelets causes endothelial activation and adhesion molecule upregulation leading to leukocyte adherence (8,9). Importantly, Portillo et al. (3) provide compelling evidence that Müller cells initiate retinal inflammation in the diabetic retina and signal to microglia to elicit their participation. The study concludes that diabetes triggers retinal neuroinflammation directly through CD40 stimulation on Müller cells and indirectly through ATP release by Müller cells, leading to stimulation of P2X7 purinergic receptors on microglia/macrophages. One might have expected inflammatory responses to be dependent on direct CD40 stimulation on microglia. Microglia represent the major resident innate immune cells of the retina and other neuronal tissues and are uniquely equipped to mount inflammatory responses to infection and tissue damage. In the retina, the conventional view is that microglia are the first responders, initiating an inflammatory response that leads to Müller cell reactive gliosis (10). Furthermore, in the brain, CD40 is mainly found on microglia, where CD40 deficiency or neutralization of CD40L inhibits microglial activation, alleviates brain pathology, and improves cognitive performance in mouse models of Alzheimer disease (11). The ATP-mediated mechanism described by Portillo et al. (3) may have implications for other aspects of DR pathology as well. Several purinergic receptors, including P2X7, play key roles in retinal physiology and pathophysiology, including modulation of retinal neurotransmission, control of vascular tone, and Müller cell swelling and gliosis, as well as RGC apoptosis (12). Diabetes was found to increase the susceptibility of retinal microvessels to transmembrane pore formation in response to P2X7 activation, suggesting that extracellular ATP may cause mural cell loss in DR (13). In addition, P2X7 and P2X4 receptor antagonists inhibited the induction of endothelial cell inflammation and permeability by high glucose (14). Release of ATP by Müller cells could also contribute to the death of RGC observed in DR, as evidence suggests that the release of ATP by gliotic Müller cells induces RGC apoptosis through P2X7 activation (15,16). Finally, the current study suggests the possibility that CD40–CD40L and ATP–P2X7 interactions represent promising new therapeutic targets for prevention of DR progression. Although the study did not establish the source of CD40L in diabetic retinas, plasma levels of soluble CD40L were significantly increased in the diabetic mice, suggesting a systemic influence (platelet activation, perhaps) on DR pathology in this model. Plasma levels of soluble CD40L are also elevated in patients with type 1 and type 2 diabetes (17,18). Several systemic treatments targeting the CD40–CD40L system have been developed to treat cancer, inflammation, and autoimmune disease (4). Although clinical trials of monoclonal antibodies blocking CD40L failed due to occurrence of thromboembolisms (19), such a complication is unlikely with intravitreal applications of these biologics. Also, development of blood-brain-barrier–penetrant P2X7 antagonists for treatment of neuroinflammation has been under way for some time (20). Although the role for this receptor in normal retinal function suggests caution, it is possible that inhibition of P2X7 on retinal microglia could modulate their function to prevent neuroinflammation from progressing toward sight-threatening DR.

          Related collections

          Most cited references17

          • Record: found
          • Abstract: found
          • Article: not found

          CD40 ligand on activated platelets triggers an inflammatory reaction of endothelial cells.

          CD40 ligand (CD40L, CD154), a transmembrane protein structurally related to the cytokine TNF-alpha, was originally identified on stimulated CD4+ T cells, and later on stimulated mast cells and basophils. Interaction of CD40L on T cells with CD40 on B cells is of paramount importance for the development and function of the humoral immune system. CD40 is not only constitutively present on B cells, but it is also found on monocytes, macrophages and endothelial cells, suggesting that CD40L has a broader function in vivo. We now report that platelets express CD40L within seconds of activation in vitro and in the process of thrombus formation in vivo. Like TNF-alpha and interleukin-1, CD40L on platelets induces endothelial cells to secrete chemokines and to express adhesion molecules, thereby generating signals for the recruitment and extravasation of leukocytes at the site of injury. Our results indicate that platelets are not only involved in haemostasis but that they also directly initiate an inflammatory response of the vessel wall.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            New functions of Müller cells.

            Müller cells, the major type of glial cells in the retina, are responsible for the homeostatic and metabolic support of retinal neurons. By mediating transcellular ion, water, and bicarbonate transport, Müller cells control the composition of the extracellular space fluid. Müller cells provide trophic and anti-oxidative support of photoreceptors and neurons and regulate the tightness of the blood-retinal barrier. By the uptake of glutamate, Müller cells are more directly involved in the regulation of the synaptic activity in the inner retina. This review gives a survey of recently discoved new functions of Müller cells. Müller cells are living optical fibers that guide light through the inner retinal tissue. Thereby they enhance the signal/noise ratio by minimizing intraretinal light scattering and conserve the spatial distribution of light patterns in the propagating image. Müller cells act as soft, compliant embedding for neurons, protecting them in case of mechanical trauma, and also as soft substrate required for neurite growth and neuronal plasticity. Müller cells release neuroactive signaling molecules which modulate neuronal activity, are implicated in the mediation of neurovascular coupling, and mediate the homeostasis of the extracellular space volume under hypoosmotic conditions which are a characteristic of intense neuronal activity. Under pathological conditions, a subset of Müller cells may differentiate to neural progenitor/stem cells which regenerate lost photoreceptors and neurons. Increasing knowledge of Müller cell function and responses in the normal and diseased retina will have great impact for the development of new therapeutic approaches for retinal diseases. Copyright © 2013 Wiley Periodicals, Inc.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              CD40 in Retinal Müller Cells Induces P2X 7 -Dependent Cytokine Expression in Macrophages/Microglia in Diabetic Mice and Development of Early Experimental Diabetic Retinopathy

              Müller cells and macrophages/microglia are likely important for the development of diabetic retinopathy; however, the interplay between these cells in this disease is not well understood. An inflammatory process is linked to the onset of experimental diabetic retinopathy. CD40 deficiency impairs this process and prevents diabetic retinopathy. Using mice with CD40 expression restricted to Müller cells, we identified a mechanism by which Müller cells trigger proinflammatory cytokine expression in myeloid cells. During diabetes, mice with CD40 expressed in Müller cells upregulated retinal tumor necrosis factor-α (TNF-α), interleukin 1β (IL-1β), intracellular adhesion molecule 1 (ICAM-1), and nitric oxide synthase (NOS2), developed leukostasis and capillary degeneration. However, CD40 did not cause TNF-α or IL-1β secretion in Müller cells. TNF-α was not detected in Müller cells from diabetic mice with CD40+ Müller cells. Rather, TNF-α was upregulated in macrophages/microglia. CD40 ligation in Müller cells triggered phospholipase C–dependent ATP release that caused P2X7-dependent production of TNF-α and IL-1β by macrophages. P2X7 −/− mice and mice treated with a P2X7 inhibitor were protected from diabetes-induced TNF-α, IL-1β, ICAM-1, and NOS2 upregulation. Our studies indicate that CD40 in Müller cells is sufficient to upregulate retinal inflammatory markers and appears to promote experimental diabetic retinopathy and that Müller cells orchestrate inflammatory responses in myeloid cells through a CD40-ATP-P2X7 pathway.
                Bookmark

                Author and article information

                Journal
                Diabetes
                Diabetes
                diabetes
                diabetes
                Diabetes
                Diabetes
                American Diabetes Association
                0012-1797
                1939-327X
                February 2017
                13 January 2017
                : 66
                : 2
                : 261-263
                Affiliations
                [1]University of Michigan Kellogg Eye Center, Ann Arbor, MI
                Author notes
                Corresponding author: Steve F. Abcouwer, sabcouwe@ 123456umich.edu .
                Article
                0047
                10.2337/dbi16-0047
                5248992
                28108606
                038c1638-4d00-42b8-bc22-5c55c20d54aa
                © 2017 by the American Diabetes Association.

                Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at http://www.diabetesjournals.org/content/license.

                History
                Page count
                Figures: 1, Tables: 0, Equations: 0, References: 20, Pages: 3
                Funding
                Funded by: National Eye Institute, DOI http://dx.doi.org/10.13039/100000053;
                Award ID: R01EY007739
                Award ID: R01EY020582
                Funded by: National Institute of Diabetes and Digestive and Kidney Diseases, DOI http://dx.doi.org/10.13039/100000062;
                Award ID: R24DK082841
                Categories
                Commentaries

                Endocrinology & Diabetes
                Endocrinology & Diabetes

                Comments

                Comment on this article