Archives

  • 2018-07
  • 2018-10
  • 2018-11
  • 2019-04
  • 2019-05
  • 2019-06
  • 2019-07
  • 2019-08
  • 2019-09
  • 2019-10
  • 2019-11
  • 2019-12
  • 2020-01
  • 2020-02
  • 2020-03
  • 2020-04
  • 2020-05
  • 2020-06
  • 2020-07
  • 2020-08
  • 2020-09
  • 2020-10
  • 2020-11
  • 2020-12
  • 2021-01
  • 2021-02
  • 2021-03
  • 2021-04
  • 2021-05
  • 2021-06
  • 2021-07
  • 2021-08
  • 2021-09
  • 2021-10
  • 2021-11
  • 2021-12
  • 2022-01
  • 2022-02
  • 2022-03
  • 2022-04
  • 2022-05
  • 2022-06
  • 2022-07
  • 2022-08
  • 2022-09
  • 2022-10
  • 2022-11
  • 2022-12
  • 2023-01
  • 2023-02
  • 2023-03
  • 2023-04
  • 2023-05
  • 2023-06
  • 2023-07
  • 2023-08
  • 2023-09
  • 2023-10
  • 2023-11
  • 2023-12
  • 2024-01
  • 2024-02
  • 2024-03
  • 2024-04
  • br Experimental section br Introduction Microglia

    2024-03-26


    Experimental section
    Introduction Microglia were discovered as a novel cell population in Ketoprofen australia in 1933 by a Spanish physician and histologist Pío del Río-Hortega [1]. In the eye, scientists used Hortega's staining technique to label the microglia in the retinal outer plexiform layer (OPL) in monkeys and rabbits [2], or in rats [3]. The origin of microglia has been debated for a long time. In the early 19 century, scientists hypothesized that microglia were derived from mesenchyme and used the term “mesoglia” [1], which subsequently supported by other researchers in the late 19th [4] and early 20th centuries [5]. Other observations pointed to alternative origins for microglia, including the nervous system [6], [7], monocytes/macrophages [8], [9], or even yolk sac [10], [11], [12]. Retinal microglia (RMG) migrate from the vasculature [13] and reside in the inner or outer plexiform layers, respectively. However, RMG become activated in response to proinflammatory signals or retinal injury. RMG activation has been studied in a variety of disease models, including studies of choroidal neovascularization (CNV) in wet AMD [14], [15], [16], experimental autoimmune uveoretinitis (EAU) [17], [18], and mechanisms leading to diabetic retinopathy [19]. Given the devastating vision loss associated with robust inflammation in the retina, prevention of RMG activation is an important therapeutic goal for management of many ocular diseases. Aldose reductase (AR), best studied as an enzyme of the polyol pathway, is involved in ocular complications of diabetes including diabetic cataract [20], [21], [22] and diabetic retinopathy [23], [24]. Recent studies also implicate AR in the pathogenesis of uveitis [25], [26], [27] and fibrotic changes associated with posterior capsular opacification [28], [29]. Genetic or pharmacological blockade of AR successfully alleviates inflammatory responses induced by endotoxin [25], [26], [30], [31], [32] or hyperglycemia [33], [34], [35]. Our previous study showed that genetic or pharmacological downregulation of AR prevents endotoxin-induced inflammatory responses in RMG primary cell cultures [26]. However, understanding of AR-regulated RMG behavior in vivo is still unclear. In this study, we took a dual approach to investigate the AR effect on RMG in vivo by using AR inhibitors or AR knockout mice (ARKO). Experiments were conducted in CX3CR1GFP mice, where microglia express green fluorescent protein (GFP) [36], allowing us to easily track RMG localization and migration in the eye. By crossing the CX3CR1GFP allele with our AR Transgenic (AR-Tg) mice, we additionally tested the effect of AR expression level on RMG activation in response to endotoxin exposure. Taken together, these studies implicate AR as an effective mediator of RMG activation in the mouse eye.
    Materials and methods
    Results
    Discussion Many studies have indicated that RMG are involved in ocular diseases such as uveitis [42] and diabetic retinopathy (DR) [43], [44]. Therefore, understanding the mechanism of RMG activation is a necessity for preventing these ocular diseases. Ramana and colleagues conducted many studies showing that AR plays a key role in modulating inflammatory responses in macrophages [30], [31], [32]. In agreement with Ramana's group, we previously demonstrated that either genetic ablation or pharmacological inhibition of AR attenuates endotoxin-induced inflammatory responses in macrophages and primary RMG [25], [26]. In this study, we further demonstrated the in vivo effect of AR using AR mutant mice or studies involving AR blockade with Sorbinil, an effective AR inhibitor (Fig. 1, Fig. 2). LPS-induced RMG migration (Fig. 1, Fig. 2) was attenuated by either Sorbinil (Fig. 1F) or AR knockout (Fig. 2D). Expression levels of AR appear to correlate with the activation phenotype observed in RMG cell populations and abundance of ocular TNF-α and CX3CL-1. Levels of RMG activation and migration ability are reduced by AR inhibition (Fig. 1) and gene inactivation (Fig. 2). In addition, ocular TNF-α and CX3CL-1 are reduced by AR inhibition or genetic inactivation (Fig. 3). The functional linkage between AR and RMG behavior is also apparent when AR levels are increased. Elevated levels of AR expression in transgenic mice are associated with increased RMG activation and migration to INL and ONL layers in the retina (Fig. 4). Our observations point to AR as a drugable target for anti-inflammatory therapy in conditions such as uveitis. In addition, these studies suggest that blinding complications of diabetes, such as diabetic retinopathy and diabetic cataracts, which are associated with elevated inflammatory markers, may result from an AR-mediated mechanism [45].