Skip to main content
Mediators of Inflammation logoLink to Mediators of Inflammation
. 2014 Jul 22;2014:432685. doi: 10.1155/2014/432685

The Role of Inflammation in the Pathogenesis of Macular Edema Secondary to Retinal Vascular Diseases

Francisco J Ascaso 1,2,*, Valentín Huerva 3,4, Andrzej Grzybowski 5,6
PMCID: PMC4134827  PMID: 25152567

Abstract

Macular edema (ME) is a nonspecific sign of numerous retinal vascular diseases. This paper is an updated overview about the role of inflammatory processes in the genesis of both diabetic macular edema (DME) and ME secondary to retinal vein occlusion (RVO). We focus on the inflammatory mediators implicated, the effect of the different intravitreal therapies, the recruitment of leukocytes mediated by adhesion molecules, and the role of retinal Müller glial (RMG) cells.

1. Macular Edema: A Nonspecific Indication of Numerous Retinal Vascular Disorders

Macular edema (ME) is defined as an accumulation of either extracellular (mainly in the outer plexiform and the inner nuclear layers) or intracellular fluid (swelling of retinal Müller glial (RMG) cells) in the central part of the retina. Indeed, at times, a combination of these types of fluid accumulation occurs [1]. ME is a nonspecific sign of numerous retinal vascular diseases, such as diabetic retinopathy (DR) and retinal vein occlusions (RVO) [2, 3]. In these disorders, inflammatory processes have been considered to be critical [46], and breakdown of the blood retinal barrier (BRB) coupled to the subsequent increase in vascular permeability often causes ME and concomitant visual acuity impairment, secondary to an increased flux in the retinal capillary endothelial cells [7, 8]. Thus, the pathogenesis of diabetic macular edema (DME) includes several interrelated factors such as chronic hyperglycemia, hypoxia, accumulation of free radicals, activation of vascular endothelial growth factor (VEGF), alterations in endothelial intercellular junctions, pericyte loss, retinal vessel leukostasis, disruption of the BRB, and an increase in vascular permeability [9, 10]. Although the pathogenesis of ME when associated with RVO (RVO-ME) is not fully understood, increased rigidity of a crossing artery as a result of an atherosclerotic process has been suggested to cause compression of the underlying vein, provoking turbulent blood flow, endothelial damage, and thrombus formation [11]. Likewise, a common vitreous adhesion at the obstruction site has also been reported, suggesting a possible role of vitreovascular traction in the etiology of some cases of BRVO [12, 13].

Atherosclerosis is a chronic low-grade inflammatory disorder and inflammation within the vascular wall contributes to the development of ME [1416]. Due to BRB breakdown secondary to damage at the tight junctions of endothelial cells, fluid diffusion from the occluded veins into the tissue can lead to ME [17]. In addition, through such mechanisms, inflammatory responses and vascular dysfunction can all interact to cause retinal ischemia, which induces the expression of VEGF [18]. DME and BRVO-ME may differ in terms of pathogenesis because the cytokine concentrations in the aqueous humor are quite different, suggesting that the inflammatory reaction may be more activated in DME than in BRVO-ME, and ischemic insult may play a central role in the development of BRVO-ME [19].

2. The Role of Inflammatory Mediators in the Pathogenesis of Macular Edema

Since Vinores et al. [20] first described the role of VEGF in both ischemic and inflammatory ocular pathologies, it is well known that certain inflammatory mediators are present at the sites of ME, such as the aforementioned VEGF, together with cytokines, chemokines, angiotensin II, prostaglandins, matrix metalloproteinases, interleukins, selectins, vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), and inflammatory cells (macrophages and neutrophils), all of which participate in a complex chain of events that has yet to be fully defined [21, 22]. The vitreous levels of these inflammatory factors appear to be related to the pathological processes [23], although it remains to be seen what blood components are extravasated, how and where they flow into the retinal tissue, and from which vessels they are absorbed [24].

It is important to define which inflammatory mediators are enhanced or dampened in the clinical situation. Indeed, it is known that the concentration of several cytokines in the vitreous cavity increases in eyes with BRVO-ME [2527], including VEGF and interleukin-6 (IL-6), and that such increases are related to the severity and prognosis of ME [28]. Likewise, increased vitreous fluid levels of interleukin-6 (IL-6), monocyte chemotactic protein-1 (MCP-1), pigment epithelium-derived factor (PEDF), and particularly VEGF and ICAM-1 were related to retinal vascular permeability and the severity of DME [29].

However, whereas the aqueous humour is easily accessible and can be examined even in an outpatient setting, it is not possible to evaluate the vitreal levels of these cytokines in a routine examination [30]. When the vitreous levels of VEGF and interleukin-6 (IL-6) have been measured in patients with DME or with ME due to BRVO and CRVO, the vitreal VEGF concentration proved to be very similar in each group [31]. However, the level of IL-6 in the vitreous cavity was significantly higher in DME patients than in those with BRVO or CRVO. Noma et al. investigated whether VEGF or IL-6 contributes to the pathogenesis of ME in eyes with BRVO [26] and CRVO [32]. They found that the vitreous fluid level of VEGF was significantly higher in the patients with BRVO and CRVO than in controls. The vitreous fluid level of IL-6 was also significantly higher in the patients with both types of RVO than in the control subjects. In the BRVO and CRVO patients, there was a significant correlation between the vitreous levels of VEGF and IL-6. Vitreous fluid levels of both VEGF and IL-6 were significantly higher in patients with BRVO/CRVO patients with ischemia than in those without ischemia. In addition, the vitreous levels of both factors were significantly correlated with the severity of macular edema in the BRVO/CRVO patients. Nevertheless, further studies will be needed to fully understand the relationship of certain inflammatory mediators to DME and ME secondary to BRVO or CRVO.

3. Recruitment of Leukocytes Mediated by Adhesion Molecules

Chemokines are multifunctional mediators that can recruit leucocytes to sites of inflammation, promoting further inflammation [33, 34]. The vitreous levels of some chemokines, including MCP-1 and MIP-1a and MIP-1b, have been reported to be affected by different retinal diseases, including DME and RVO [3537]. Mononuclear cell chemoattractants, such as MCP-1, IL-1, IL-6, IL-8, IL-12, and TNF-α, are also known to be expressed in ischemic areas, and these factors may induce the recruitment of leukocytes and their adhesion to the target tissue [38]. Thus, it may not be surprising that MIP-1b is expressed in eyes with DME and ME-RVO given that these disorders lead to retinal ischemia and inflammation [19, 36, 37, 39].

Leukocytes also play a role in increasing vascular permeability, along with VEGF. When they accumulate in the perivascular space, monocytes and lymphocytes initiate this process through leucocyte endothelial interactions [40]. These interactions are mediated by adhesion molecules (selectins, immunoglobulins, integrins, etc.) expressed by the vascular endothelium [41], which contribute to the disruption of tight junctions and the breakdown of the BRB [42, 43]. BRB breakdown may be initiated by different mechanisms, including leucocyte-mediated (recruitment and adhesion) endothelial injury, changes in endothelial cells, activation of protein kinase C, and the induction of fenestrations and vesiculovacuolar organelles [1].

4. The Role of Retinal Müller Glial (RMG) Cells

It is well known that ME develops due to vascular leakage and/or through cytotoxic events (e.g., glial cell swelling) [44, 45]. Although their importance in retinal vascular diseases is not fully known, RMG cells play a crucial role in regulating the volume of the extracellular space and water and ion homeostasis and in preserving the inner BRB [46].

Excess water is absorbed by retinal pigment epithelium (RPE) and RMG cells. RPE cells carry out the subretinal fluid, whereas RMG cells dehydrate the inner retinal tissue [44]. Transcellular water transport is linked to a transport of potassium and chloride ions [47]. Water flow through the RMG and RPE cells membranes is facilitated by water-selective channels: the aquaporins. The major water channel of RPE cells and photoreceptors is aquaporin-1, whereas RMG cells express aquaporin-4 [48, 49]. Water transport is coupled to the spatial-buffering potassium currents flowing through RMG cells [50]. Alteration of the transglial water transport after downregulation of Kir4.1 channels and osmotic swelling of RMG cells under pathologic conditions such as transient retinal ischemia-reperfusion and diabetes mellitus have been implicated in the development of ME [51, 52].

Moreover, they contribute to the survival of ganglion cell neurons and photoreceptors, they are responsible for the stabilization of retinal structure, and they modulate inflammatory and immune responses [53, 54]. Thus, the RMG cells can upregulate the expression of inflammatory mediators, including MCP-1, which recruit microglial cells and phagocytotic monocytes/macrophages to regions of damage [55, 56]. Distinct disorders are associated with BRB breakdown, which results in the extravasation of the blood constituents that inactivate Kir channels and that induce RMG cell depolarization [57, 58].

Vascular leakage is a crucial pathogenic mechanism involved in ME [59]. Retinal capillaries are closely ensheathed by glial processes [53] and RMG cells enhance the barrier function of the vascular endothelium [6062]. Due to inflammation and hypoxia, RMG cells produce factors such as VEGF, TNF-α, IL-1β, and prostaglandins, all of which enhance retinal vascular permeability [6274].

Fluid clearance is usually mediated by osmotic water transport through RMG cells, a process facilitated by Kir channels and water channels, especially AQP4 [7578]. AQP4 acts in combination with K+ channels to maintain osmotic retinal homeostasis. Indeed, Kir4.1 channel dysfunction, such as that observed in retinal vascular disorders, disturbs transcellular water transport [45, 79], resulting in water influx and RMG cells swelling [46]. Although a few studies have investigated the mechanisms of action of corticosteroids in ME, it has been shown that RMG cells express both the glucocorticoid receptor (GR) and the mineralocorticoid receptor (MR) [44]. Moreover, the MR ligand aldosterone increases the expression of AQP4 and Kir4.1, and it induces retinal swelling [80]. Finally, the two main corticosteroids used in intravitreal therapies, TA and dexamethasone, the latter administered through a sustained-release implant, regulate AQP4 and Kir4.1 distinctly, indicating that they are not functionally equivalent [44].

5. The Effect of the Different Intravitreal Therapies

It is also important to determine whether there are any differences in the response to different therapies. Intravitreal injection of both triamcinolone acetonide (TA) and bevacizumab has been reported to be effective in reducing macular thickness in DME [39, 81]. Indeed, intravitreal injection of TA is effective in decreasing macular thickness in patients with ME due to BRVO or CRVO, reducing the ocular expression of inflammatory cytokines [31]. Recently, it was shown that intravitreal TA injection significantly diminished MCP-1 (monocyte chemotactic protein-1) and MIP-1b (macrophage inflammatory protein-1b) levels in the aqueous humour of eyes with BRVO-ME [28]. Moreover, the decrease in aqueous humour MIP-1b, a chemokine with proinflammatory activity, was correlated with the basal foveal thickness and its improvement following TA injection. Although the exact mechanism leading to the improvement in BRVO-ME following intravitreal TA injection has not been well established, several possible mechanisms have been considered. For example, TA could downregulate VEGF, which might prevent a decrease in occlusion as well as inhibiting any increase in glial fibrillary acidic protein (GFAP) expression in RMG cells [82]. Likewise, intravitreal TA prevents osmotic swelling of the RMG cells through the opening of K+ (Kir) 4.1 channels and aquaporin-1 and aquaporin-4 (AQP-1 and -4) in the Müller cell membrane [83, 84]. These effects might reduce the BRB breakdown that occurs in BRVO, promoting the resolution of the ME. However, IL-6-independent VEGF secretion might also contribute to the persistence BRVO-ME after intravitreal TA injection [6].

Intravitreal injection of an anti-VEGF antibody has also been reported to be effective in reducing CRVO and DR associated with ME [39, 85]. Antiangiogenic drugs, such as ranibizumab, could be anti-inflammatory as well, and part of their actions could be through an anti-inflammatory process. They would need to be able to prevent the VEGF induced by TNF-α from acting on the RPE outside the cell. Inhibition of VEGF may act through both anti-inflammatory and antiangiogenic processes and human recombinant antiangiogenic isoforms such as VEGF-A165b can be anti-inflammatory on RPE cells stimulated by TNF-α [86].

While intravitreal TA injection may have the same beneficial effects as bevacizumab in decreasing foveal thickness and improving visual acuity in the management of ME due to BRVO, TA seems to be more effective than anti-VEGF therapy in patients with DME [23, 34]. Therefore, regarding the improvement in DME, anti-VEGF therapy would be less beneficial than corticosteroid therapy. This suggests that the pathogenesis of DME can be attributed not only to VEGF alone but also to the other inflammatory molecules that are suppressed by corticosteroids [31, 87]. Although the pathogenesis of DME is not fully understood, steroids can modulate vascular permeability by suppressing the expression of VEGF and its receptor, as well as IL-6 and ICAM-1. In addition, they can also reduce the activity of inflammatory cells that release cytokines, stabilizing cell membranes and tight junctions, acting upstream of pigment epithelium-derived factor (PEDF) expression [88]. Therefore, TA has multiple actions compared with bevacizumab, which only diminishes the intraocular levels of free VEGF.

The use of anti-VEGF and steroid agents in ME secondary to retinal vascular diseases is an evolving field. There is an ongoing debate regarding the safety, efficacy, and economic concerns related to these intravitreal therapies to reduce the treatment burden [89]. The future of treatment for DME and macular edema associated with central and branch retinal vein occlusion will probably be some kind of combination: anti-VEGF inhibitors, steroids, and laser.

In conclusion, inflammatory processes can be considered crucial in the pathogenesis of ME related to retinal vascular disorders, thereby representing important therapeutic targets in these diseases.

Conflict of Interests

Costs of language proofreading and medical writer support were covered by Allergan.

References

  • 1.Scholl S, Kirchhof J, Augustin AJ. Pathophysiology of macular edema. Ophthalmologica. 2010;224, supplement 1:8–15. doi: 10.1159/000315155. [DOI] [PubMed] [Google Scholar]
  • 2.Marmor MF. Mechanisms of fluid accumulation in retinal edema. Documenta Ophthalmologica. 1999;97(3-4):239–249. doi: 10.1023/a:1002192829817. [DOI] [PubMed] [Google Scholar]
  • 3.Tranos PG, Wickremasinghe SS, Stangos NT, Topouzis F, Tsinopoulos I, Pavesio CE. Macular edema. Survey of Ophthalmology. 2004;49(5):470–490. doi: 10.1016/j.survophthal.2004.06.002. [DOI] [PubMed] [Google Scholar]
  • 4.Kaštelan S, Zjačić-Rotkvić V, Kaštelan Ž. Could diabetic retinopathy be an autoimmune disease? Medical Hypotheses. 2007;68(5):1016–1018. doi: 10.1016/j.mehy.2006.05.073. [DOI] [PubMed] [Google Scholar]
  • 5.Adamis AP, Berman AJ. Immunological mechanisms in the pathogenesis of diabetic retinopathy. Seminars in Immunopathology. 2008;30(2):65–84. doi: 10.1007/s00281-008-0111-x. [DOI] [PubMed] [Google Scholar]
  • 6.Park SP, Ahn JK. Changes of aqueous vascular endothelial growth factor and interleukin-6 after intravitreal triamcinolone for branch retinal vein occlusion. Clinical & Experimental Ophthalmology. 2008;36(9):831–835. doi: 10.1111/j.1442-9071.2009.01909.x. [DOI] [PubMed] [Google Scholar]
  • 7.Vinores SA, Derevjanik NL, Ozaki H, Okamoto N, Campochiaro PA. Cellular mechanisms of blood-retinal barrier dysfunction in macular edema. Documenta Ophthalmologica. 1999;97(3-4):217–228. doi: 10.1023/a:1002136712070. [DOI] [PubMed] [Google Scholar]
  • 8.Antonetti DA, Lieth E, Barber AJ, Gardner TW. Molecular mechanisms of vascular permeability in diabetic retinopathy. Seminars in Ophthalmology. 1999;14(4):240–248. doi: 10.3109/08820539909069543. [DOI] [PubMed] [Google Scholar]
  • 9.Romero-Aroca P. Targeting the pathophysiology of diabetic macular edema. Diabetes Care. 2010;33(11):2484–2485. doi: 10.2337/dc10-1580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Joussen A, Smyth N, Niessen C. Pathophysiology of diabetic macular edema. Developments in Ophthalmology. 2007;39:1–12. doi: 10.1159/000098495. [DOI] [PubMed] [Google Scholar]
  • 11.Fekrat S, Finkelstein D. Venous occlusive disease. In: Regillo CD, Brown GC, Flynn HW Jr., editors. Vitreoretinal Disease: The Essentials. chapter 9. New York, NY, USA: Thieme; 1999. pp. 117–132. [Google Scholar]
  • 12.Ascaso FJ, Huerva V. Vitreoretinal traction in impending branch retinal vein occlusion: a pathogenetic role? Thrombosis and Haemostasis. 2012;108(2):208–209. doi: 10.1160/TH12-03-0190. [DOI] [PubMed] [Google Scholar]
  • 13.Ascaso FJ, Padgett E, Núñez E, Villén L, Grzybowski A, Cristóbal JA. Branch retinal vein occlusion and vitreovascular traction: a preliminary spectral domain OCT case–control study. Graefe's Archive for Clinical and Experimental Ophthalmology. 2014;252(3):375–381. doi: 10.1007/s00417-013-2463-8. [DOI] [PubMed] [Google Scholar]
  • 14.Huber SA, Sakkinen P, Conze D, Hardin N, Tracy R. Interleukin-6 exacerbates early atherosclerosis in mice. Arteriosclerosis, Thrombosis, and Vascular Biology. 1999;19(10):2364–2367. doi: 10.1161/01.atv.19.10.2364. [DOI] [PubMed] [Google Scholar]
  • 15.Pinderski LJ, Fischbein MP, Subbanagounder G, et al. Overexpression of interleukin-10 by activated T lymphocytes inhibits atherosclerosis in LDL receptor-deficient mice by altering lymphocyte and macrophage phenotypes. Circulation Research. 2002;90(10):1064–1071. doi: 10.1161/01.res.0000018941.10726.fa. [DOI] [PubMed] [Google Scholar]
  • 16.Whitman SC, Ravisankar P, Elam H, Daugherty A. Exogenous interferon-γ enhances atherosclerosis in apolipoprotein E-/- mice. The American Journal of Pathology. 2000;157(6):1819–1824. doi: 10.1016/s0002-9440(10)64820-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Silva RM, Faria de Abreu JR, Cunha-Vaz JG. Blood-retina barrier in acute retinal branch vein occlusion. Graefe's Archive for Clinical and Experimental Ophthalmology. 1995;233(11):721–726. doi: 10.1007/BF00164677. [DOI] [PubMed] [Google Scholar]
  • 18.Aiello LP, Northrup JM, Keyt BA, Takagi H, Iwamoto MA. Hypoxic regulation of vascular endothelial growth factor in retinal cells. Archives of Ophthalmology. 1995;113(12):1538–1544. doi: 10.1001/archopht.1995.01100120068012. [DOI] [PubMed] [Google Scholar]
  • 19.Lee WJ, Kang MH, Seong M, Cho HY. Comparison of aqueous concentrations of angiogenic and inflammatory cytokines in diabetic macular oedema and macular oedema due to branch retinal vein occlusion. British Journal of Ophthalmology. 2012;96(11):1426–1430. doi: 10.1136/bjophthalmol-2012-301913. [DOI] [PubMed] [Google Scholar]
  • 20.Vinores SA, Youssri AI, Luna JD, et al. Upregulation of vascular endothelial growth factor in ischemic and non-ischemic human and experimental retinal disease. Histology and Histopathology. 1997;12(1):99–109. [PubMed] [Google Scholar]
  • 21.Joussen AM, Smyth N, Niessen C. Pathophysiology of diabetic macular edema. Developments in Ophthalmology. 2007;39:1–12. doi: 10.1159/000098495. [DOI] [PubMed] [Google Scholar]
  • 22.Pasqualetti G, Danesi R, del Tacca M, Bocci G. Vascular endothelial growth factor pharmacogenetics: a new perspective for anti-angiogenic therapy. Pharmacogenomics. 2007;8(1):49–66. doi: 10.2217/14622416.8.1.49. [DOI] [PubMed] [Google Scholar]
  • 23.Yoshimura T, Sonoda K, Sugahara M, et al. Comprehensive analysis of inflammatory immune mediators in vitreoretinal diseases. PLoS ONE. 2009;4(12) doi: 10.1371/journal.pone.0008158.e8158 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Ogino K, Murakami T, Tsujikawa A, et al. Characteristics of optical coherence tomographic hyperreflective foci in retinal vein occlusion. Retina. 2012;32(1):77–85. doi: 10.1097/IAE.0b013e318217ffc7. [DOI] [PubMed] [Google Scholar]
  • 25.Noma H, Funatsu H, Yamasaki M, et al. Pathogenesis of macular edema with branch retinal vein occlusion and intraocular levels of vascular endothelial growth factor and interleukin-6. American Journal of Ophthalmology. 2005;140(2):256.e1–256.e7. doi: 10.1016/j.ajo.2005.03.003. [DOI] [PubMed] [Google Scholar]
  • 26.Noma H, Minamoto A, Funatsu H, et al. Intravitreal levels of vascular endothelial growth factor and interleukin-6 are correlated with macular edema in branch retinal vein occlusion. Graefe's Archive for Clinical and Experimental Ophthalmology. 2006;244(3):309–315. doi: 10.1007/s00417-004-1087-4. [DOI] [PubMed] [Google Scholar]
  • 27.Shimura M, Nakazawa T, Yasuda K, et al. Comparative therapy evaluation of intravitreal bevacizumab and triamcinolone acetonide on persistent diffuse diabetic macular edema. American Journal of Ophthalmology. 2008;145(5):854–861. doi: 10.1016/j.ajo.2007.12.031. [DOI] [PubMed] [Google Scholar]
  • 28.Kunikata H, Shimura M, Nakazawa T, et al. Chemokines in aqueous humour before and after intravitreal triamcinolone acetonide in eyes with macular oedema associated with branch retinal vein occlusion. Acta Ophthalmologica. 2012;90(2):162–167. doi: 10.1111/j.1755-3768.2010.01892.x. [DOI] [PubMed] [Google Scholar]
  • 29.Funatsu H, Noma H, Mimura T, Eguchi S, Hori S. Association of vitreous inflammatory factors with diabetic macular edema. Ophthalmology. 2009;116(1):73–79. doi: 10.1016/j.ophtha.2008.09.037. [DOI] [PubMed] [Google Scholar]
  • 30.Noma H, Funatsu H, Yamasaki M, et al. Aqueous humour levels of cytokines are correlated to vitreous levels and severity of macular oedema in branch retinal vein occlusion. Eye. 2008;22(1):42–48. doi: 10.1038/sj.eye.6702498. [DOI] [PubMed] [Google Scholar]
  • 31.Funatsu H, Noma H, Mimura T, Eguchi S. Vitreous inflammatory factors and macular oedema. British Journal of Ophthalmology. 2012;96(2):302–304. doi: 10.1136/bjo.2010.181222. [DOI] [PubMed] [Google Scholar]
  • 32.Noma H, Funatsu H, Mimura T, Harino S, Hori S. Vitreous levels of interleukin-6 and vascular endothelial growth factor in macular edema with central retinal vein occlusion. Ophthalmology. 2009;116(1):87–93. doi: 10.1016/j.ophtha.2008.09.034. [DOI] [PubMed] [Google Scholar]
  • 33.Struyf S, Proost P, van Damme J. Regulation of the immune response by the interaction of chemokines and proteases. Advances in Immunology. 2003;81:1–44. doi: 10.1016/s0065-2776(03)81001-5. [DOI] [PubMed] [Google Scholar]
  • 34.Abu El-Asrar AM, Struyf S, Kangave D, Geboes K, van Damme J. Chemokines in proliferative diabetic retinopathy and proliferative vitreoretinopathy. European Cytokine Network. 2006;17(3):155–165. [PubMed] [Google Scholar]
  • 35.Funk M, Kriechbaum K, Prager F, et al. Intraocular concentrations of growth factors and cytokines in retinal vein occlusion and the effect of therapy with bevacizumab. Investigative Ophthalmology and Visual Science. 2009;50(3):1025–1032. doi: 10.1167/iovs.08-2510. [DOI] [PubMed] [Google Scholar]
  • 36.Ai H, Song HP. Different expression pattern of serum soluble intercellular adhesion molecules-1 and neutrophilic expression of CD18 in patients with diabetic retinopathy. International Journal of Ophthalmology. 2012;5:202–207. doi: 10.3980/j.issn.2222-3959.2012.02.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Huang H, Gandhi JK, Zhong X, et al. TNFα is required for late BRB breakdown in diabetic retinopathy, and its inhibition prevents leukostasis and protects vessels and neurons from apoptosis. Investigative Ophthalmology and Visual Science. 2011;52(3):1336–1344. doi: 10.1167/iovs.10-5768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Frangogiannis NG. Chemokines in the ischemic myocardium: from inflammation to fibrosis. Inflammation Research. 2004;53(11):585–595. doi: 10.1007/s00011-004-1298-5. [DOI] [PubMed] [Google Scholar]
  • 39.Diabetic Retinopathy Clinical Research Network. A phase II randomized clinical trial of intravitreal bevacizumab for diabetic macular edema. Ophthalmology. 2007;114(10):1860–1867. doi: 10.1016/j.ophtha.2007.05.062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Noma H, Funatsu H, Mimura T, Shimada K. Increase of aqueous inflammatory factors in macular edema with branch retinal vein occlusion: a case control study. Journal of Inflammation. 2010;7, article 44 doi: 10.1186/1476-9255-7-44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Nishijima K, Kiryu J, Tsujikawa A, et al. Inhibitory effects of antithrombin III on interactions between blood cells and endothelial cells during retinal ischemia-reperfusion injury. Investigative Ophthalmology and Visual Science. 2003;44(1):332–341. doi: 10.1167/iovs.02-0493. [DOI] [PubMed] [Google Scholar]
  • 42.Jousssen AM, Poulaki V, Le ML, et al. A central role for inflammation in the pathogenesis of diabetic retinopathy. The FASEB Journal. 2004;18(12):1450–1452. doi: 10.1096/fj.03-1476fje. [DOI] [PubMed] [Google Scholar]
  • 43.Zhang SX, Wang JJ, Gao G, Shao C, Mott R, Ma J. Pigment epithelium-derived factor (PEDF) is an endogenous antiinflammatory factor. The FASEB Journal. 2006;20(2):323–325. doi: 10.1096/fj.05-4313fje. [DOI] [PubMed] [Google Scholar]
  • 44.Bringmann A, Reichenbach A, Wiedemann P. Pathomechanisms of cystoid macular edema. Ophthalmic Research. 2004;36(5):241–249. doi: 10.1159/000081203. [DOI] [PubMed] [Google Scholar]
  • 45.Reichenbach A, Wurm A, Pannicke T, Iandiev I, Wiedemann P, Bringmann A. Müller cells as players in retinal degeneration and edema. Graefe's Archive for Clinical and Experimental Ophthalmology. 2007;245(5):627–636. doi: 10.1007/s00417-006-0516-y. [DOI] [PubMed] [Google Scholar]
  • 46.Bringmann A, Wiedemann P. Müller glial cells in retinal disease. Ophthalmologica. 2012;227(1):1–19. doi: 10.1159/000328979. [DOI] [PubMed] [Google Scholar]
  • 47.Bringmann A, Pannicke T, Grosche J, et al. Müller cells in the healthy and diseased retina. Progress in Retinal and Eye Research. 2006;25(4):397–424. doi: 10.1016/j.preteyeres.2006.05.003. [DOI] [PubMed] [Google Scholar]
  • 48.Stamer WD, Bok D, Hu J, Jaffe GJ, McKay BS. Aquaporin-1 channels in human retinal pigment epithelium: role in transepithelial water movement. Investigative Ophthalmology and Visual Science. 2003;44(6):2803–2808. doi: 10.1167/iovs.03-0001. [DOI] [PubMed] [Google Scholar]
  • 49.Iandiev I, Pannicke T, Reichel MB, Wiedemann P, Reichenbach A, Bringmann A. Expression of aquaporin-1 immunoreactivity by photoreceptor cells in the mouse retina. Neuroscience Letters. 2005;388(2):96–99. doi: 10.1016/j.neulet.2005.06.046. [DOI] [PubMed] [Google Scholar]
  • 50.Nagelhus EA, Horio Y, Inanobe A. Immunogold evidence suggests that coupling of K+ siphoning and water transport in rat retinal Müller cells is mediated by a coenrichment of Kir4.1 and AQP4 in specific membrane domains. Glia. 1999;26(1):47–54. doi: 10.1002/(sici)1098-1136(199903)26:1<47::aid-glia5>3.0.co;2-5. [DOI] [PubMed] [Google Scholar]
  • 51.Pannicke T, Iandiev I, Uckermann O, et al. A potassium channel-linked mechanism of glial cell swelling in the postischemic retina. Molecular and Cellular Neuroscience. 2004;26(4):493–502. doi: 10.1016/j.mcn.2004.04.005. [DOI] [PubMed] [Google Scholar]
  • 52.Pannicke T, Iandiev I, Wurm A, et al. Diabetes alters osmotic swelling characteristics and membrane conductance of glial cells in rat retina. Diabetes. 2006;55(3):633–639. doi: 10.2337/diabetes.55.03.06.db05-1349. [DOI] [PubMed] [Google Scholar]
  • 53.Reichenbach A, Bringmann A. Müller Cells in the Healthy and Diseased Retina. New York, NY, USA: Springer; 2010. [DOI] [PubMed] [Google Scholar]
  • 54.Bringmann A, Pannicke T, Biedermann B, et al. Role of retinal glial cells in neurotransmitter uptake and metabolism. Neurochemistry International. 2009;54(3-4):143–160. doi: 10.1016/j.neuint.2008.10.014. [DOI] [PubMed] [Google Scholar]
  • 55.Nakazawa T, Matsubara A, Noda K, et al. Characterization of cytokine responses to retinal detachment in rats. Molecular Vision. 2006;12:867–878. [PubMed] [Google Scholar]
  • 56.Hollborn M, Francke M, Iandiev I, et al. Early activation of inflammation- and immune response-related genes after experimental detachment of the porcine retina. Investigative Ophthalmology and Visual Science. 2008;49(3):1262–1273. doi: 10.1167/iovs.07-0879. [DOI] [PubMed] [Google Scholar]
  • 57.Puro DG, Stuenke EL. Thrombin-induced inhibition of potassium currents in human retinal glial (Müller) cells. The Journal of Physiology. 1995;485(2):337–348. doi: 10.1113/jphysiol.1995.sp020733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Kusaka S, Kapousta-Bruneau NV, Puro DG. Plasma-induced changes in the physiology of mammalian retinal glial cells: role of glutamate. Glia. 1999;25:205–215. doi: 10.1002/(sici)1098-1136(19990201)25:3<205::aid-glia1>3.0.co;2-s. [DOI] [PubMed] [Google Scholar]
  • 59.Cunha-Vaz JG, Travassos A. Breakdown of the blood-retinal barriers and cystoid macular edema. Survey of Ophthalmology. 1984;28:S485–S492. doi: 10.1016/0039-6257(84)90230-3. [DOI] [PubMed] [Google Scholar]
  • 60.Tout S, Chan-Ling T, Hollander H, Stone J. The role of Müller cells in the formation of the blood-retinal barrier. Neuroscience. 1993;55(1):291–301. doi: 10.1016/0306-4522(93)90473-s. [DOI] [PubMed] [Google Scholar]
  • 61.Diaz CM, Penfold PL, Provis JM. Modulation of the resistance of a human endothelial cell line by human retinal glia. Australian and New Zealand Journal of Ophthalmology. 1998;26(1):S62–S64. doi: 10.1111/j.1442-9071.1998.tb01376.x. [DOI] [PubMed] [Google Scholar]
  • 62.Tretiach M, Madigan MC, Wen L, Gillies MC. Effect of Müller cell co-culture on in vitro permeability of bovine retinal vascular endothelium in normoxic and hypoxic conditions. Neuroscience Letters. 2005;378(3):160–165. doi: 10.1016/j.neulet.2004.12.026. [DOI] [PubMed] [Google Scholar]
  • 63.Eichler W, Yafai Y, Wiedemann P, Reichenbach A. Angiogenesis-related factors derived from retinal glial (Müller) cells in hypoxia. NeuroReport. 2004;15(10):1633–1637. doi: 10.1097/01.wnr.0000133071.00786.a4. [DOI] [PubMed] [Google Scholar]
  • 64.Keck PJ, Hauser SD, Krivi G, et al. Vascular permeability factor, an endothelial cell mitogen related to PDGF. Science. 1989;246(4935):1309–1312. doi: 10.1126/science.2479987. [DOI] [PubMed] [Google Scholar]
  • 65.Murata T, Nakagawa K, Khalil A, Ishibashi T, Inomata H, Sueishi K. The relation between expression of vascular endothelial growth factor and breakdown of the blood-retinal barrier in diabetic rat retinas. Laboratory Investigation. 1996;74(4):819–825. [PubMed] [Google Scholar]
  • 66.Luna JD, Chan CC, Derevjanik NL, et al. Blood-retinal barrier (BRB) breakdown in experimental autoimmune uveoretinitis: comparison with vascular endothelial growth factor, tumor necrosis factor-α, and interleukin-1β-mediated breakdown. Journal of Neuroscience Research. 1997;49(3):268–280. doi: 10.1002/(sici)1097-4547(19970801)49:3<268::aid-jnr2>3.0.co;2-a. [DOI] [PubMed] [Google Scholar]
  • 67.Aiello LP, Bursell SE, Clermont A, et al. Vascular endothelial growth factor-induced retinal permeability is mediated by protein kinase C in vivo and suppressed by an orally effective β-isoform-selective inhibitor. Diabetes. 1997;46(9):1473–1480. doi: 10.2337/diab.46.9.1473. [DOI] [PubMed] [Google Scholar]
  • 68.Claudio L, Martiney JA, Brosnan CF. Ultrastructural studies of the blood-retina barrier after exposure to interleukin-1β or tumor necrosis factor-α . Laboratory Investigation. 1994;70(6):850–861. [PubMed] [Google Scholar]
  • 69.Derevjanik NL, Vinores SA, Xiao W, et al. Quantitative assessment of the integrity of the blood-retinal barrier in mice. Investigative Ophthalmology and Visual Science. 2002;43(7):2462–2467. [PubMed] [Google Scholar]
  • 70.Drescher KM, Whittum-Hudson JA. Modulation of immune-associated surface markers and cytokine production by murine retinal glial cells. Journal of Neuroimmunology. 1996;64(1):71–81. doi: 10.1016/0165-5728(95)00156-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Amin RH, Frank RN, Kennedy A, Eliott D, Puklin JE, Abrams GW. Vascular endothelial growth factor is present in glial cells of the retina and optic nerve of human subjects with nonproliferative diabetic retinopathy. Investigative Ophthalmology and Visual Science. 1997;38(1):36–47. [PubMed] [Google Scholar]
  • 72.Eichler W, Kuhrt H, Hoffmann S, Wiedemann P, Reichenbach A. VEGF release by retinal glia depends on both oxygen and glucose supply. NeuroReport. 2000;11(16):3533–3537. doi: 10.1097/00001756-200011090-00026. [DOI] [PubMed] [Google Scholar]
  • 73.Yafai Y, Iandiev I, Wiedemann P, Reichenbach A, Eichler W. Retinal endothelial angiogenic activity: effects of hypoxia and glial (Müller) cells. Microcirculation. 2004;11(7):577–586. doi: 10.1080/10739680490503375. [DOI] [PubMed] [Google Scholar]
  • 74.Noda K, Ishida S, Shinoda H, et al. Hypoxia induces the expression of membrane-type 1 matrix metalloproteinase in retinal glial cells. Investigative Ophthalmology and Visual Science. 2005;46(10):3817–3824. doi: 10.1167/iovs.04-1528. [DOI] [PubMed] [Google Scholar]
  • 75.Iandiev I, Wurm A, Hollborn M, et al. Müller cell response to blue light injury of the rat retina. Investigative Ophthalmology and Visual Science. 2008;49(8):3559–3567. doi: 10.1167/iovs.08-1723. [DOI] [PubMed] [Google Scholar]
  • 76.Nagelhus EA, Horio Y, Inanobe A, et al. Immunogold evidence suggests that coupling of K+ siphoning and water transport in rat retinal Müller cells is mediated by a coenrichment of Kir4.1 and AQP4 in specific membrane domains. Glia. 1999;26:47–54. doi: 10.1002/(sici)1098-1136(199903)26:1<47::aid-glia5>3.0.co;2-5. [DOI] [PubMed] [Google Scholar]
  • 77.Nagelhus EA, Veruki ML, Torp R, et al. Aquaporin-4 water channel protein in the rat retina and optic nerve: polarized expression in Muller cells and fibrous astrocytes. Journal of Neuroscience. 1998;18(7):2506–2519. doi: 10.1523/JNEUROSCI.18-07-02506.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Iandiev I, Pannicke T, Biedermann B, Wiedemann P, Reichenbach A, Bringmann A. Ischemia-reperfusion alters the immunolocalization of glial aquaporins in rat retina. Neuroscience Letters. 2006;408(2):108–112. doi: 10.1016/j.neulet.2006.08.084. [DOI] [PubMed] [Google Scholar]
  • 79.Iandiev I, Pannicke T, Reichenbach A, Wiedemann P, Bringmann A. Diabetes alters the localization of glial aquaporins in rat retina. Neuroscience Letters. 2007;421(2):132–136. doi: 10.1016/j.neulet.2007.04.076. [DOI] [PubMed] [Google Scholar]
  • 80.Zhao M, Bousquet E, Valamanesh F, et al. Differential regulations of AQP4 and Kir4.1 by triamcinolone acetonide and dexamethasone in the healthy and inflamed retina. Investigative Ophthalmology and Visual Science. 2011;52(9):6340–6347. doi: 10.1167/iovs.11-7675. [DOI] [PubMed] [Google Scholar]
  • 81.Ip MS, Scott IU, Van Veldhuisen PC, et al. A randomized trial comparing the efficacy and safety of intravitreal triamcinolone with observation to treat vision loss associated with macular edema secondary to central retinal vein occlusion: the standard care vs corticosteroid for retinal vein occlusion (SCORE) study report 5. Archives of Ophthalmology. 2009;127(9):1101–1114. doi: 10.1001/archophthalmol.2009.234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.McAllister IL, Vijayasekaran S, Chen SD, Yu D. Effect of triamcinolone acetonide on vascular endothelial growth factor and occludin levels in branch retinal vein occlusion. American Journal of Ophthalmology. 2009;147(5):838–846. doi: 10.1016/j.ajo.2008.12.006. [DOI] [PubMed] [Google Scholar]
  • 83.Uckermann O, Kutzera F, Wolf A, et al. The glucocorticoid triamcinolone acetonide inhibits osmotic swelling of retinal glial cells via stimulation of endogenous adenosine signaling. Journal of Pharmacology and Experimental Therapeutics. 2005;315(3):1036–1045. doi: 10.1124/jpet.105.092353. [DOI] [PubMed] [Google Scholar]
  • 84.Rehak M, Hollborn M, Iandiev I, et al. Retinal gene expression and Müller cell responses after branch retinal vein occlusion in the rat. Investigative Ophthalmology & Visual Science. 2009;50(5):2359–2367. doi: 10.1167/iovs.08-2332. [DOI] [PubMed] [Google Scholar]
  • 85.Spaide RF, Chang LS, Klancnik JM, et al. Prospective study of intravitreal ranibizumab as a treatment for decreased visual acuity secondary to central retinal vein occlusion. American Journal of Ophthalmology. 2009;147(2):298–306. doi: 10.1016/j.ajo.2008.08.016. [DOI] [PubMed] [Google Scholar]
  • 86.Thichanpiang P, Harper SJ, Wongprasert K, Bates DO. TNF-α-induced ICAM-1 expression and monocyte adhesion in human RPE cells is mediated in part through autocrine VEGF stimulation. Molecular Vision. 2014;20:781–789. [PMC free article] [PubMed] [Google Scholar]
  • 87.Paccola L, Costa RA, Folgosa MS, Barbosa JC, Scott IU, Jorge R. Intravitreal triamcinolone versus bevacizumab for treatment of refractory diabetic macular oedema (IBEME study) British Journal of Ophthalmology. 2008;92(1):76–80. doi: 10.1136/bjo.2007.129122. [DOI] [PubMed] [Google Scholar]
  • 88.Edelman JL, Lutz D, Castro MR. Corticosteroids inhibit VEGF-induced vascular leakage in a rabbit model of blood-retinal and blood-aqueous barrier breakdown. Experimental Eye Research. 2005;80(2):249–258. doi: 10.1016/j.exer.2004.09.013. [DOI] [PubMed] [Google Scholar]
  • 89.Koss MJ, Naser H, Sener A, et al. Combination therapy in diabetic macular oedema and retinal vein occlusion—past and present. Acta Ophthalmologica. 2012;90(6):580–589. doi: 10.1111/j.1755-3768.2010.01962.x. [DOI] [PubMed] [Google Scholar]

Articles from Mediators of Inflammation are provided here courtesy of Wiley

RESOURCES