Open Access

Functional role of translocator protein and its ligands in ocular diseases (Review)

  • Authors:
    • Mingyi Yu
    • Shaozhen Zhao
  • View Affiliations

  • Published online on: January 8, 2024     https://doi.org/10.3892/mmr.2024.13157
  • Article Number: 33
  • Copyright: © Yu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The 18 kDa translocator protein (TSPO) is an essential outer mitochondrial membrane protein that is responsible for mitochondrial transport, maintenance of mitochondrial homeostasis and normal physiological cell function. The role of TSPO in the pathogenesis of ocular diseases is a growing area of interest. More notably, TSPO exerts positive effects in regulating various pathophysiological processes, such as the inflammatory response, oxidative stress, steroid synthesis and modulation of microglial function, in combination with a variety of specific ligands such as 1‑(2‑chlorophenyl‑N‑methylpropyl)‑3‑isoquinolinecarboxamide, 4'‑chlorodiazepam and XBD173. In the present review, the expression of TSPO in ocular tissues and the functional role of TSPO and its ligands in diverse ocular diseases was discussed.

Introduction

Translocator protein (TSPO; 18 kDa) is a highly structurally conserved hydrophobic protein located on the outer mitochondrial membrane (OMM) (1). Discovered in the 1970s, TSPO was originally termed the ‘peripheral benzodiazepine receptor’ due to its specific binding site for certain benzodiazepines in peripheral tissues (2). In 2006, the name was changed to TSPO to reflect the cholesterol binding and transportation functions of the protein (3). TSPO is also involved in a variety of cellular physiological activities such as mitochondrial quality control, mitochondrial permeability transition pore opening, voltage-dependent anion channel (VDAC) opening, calcium transport, cellular proliferation, programmed apoptosis and reactive oxygen species (ROS) production (48). This extensive range of molecular functions has led to the association of TSPO with the pathogenesis of multiple diseases, including central nervous system diseases and cardiovascular anomalies (912). Increasing research interest in TSPO as a crucial pathogenic factor and therapeutic target has driven the study of TSPO-specific, high-affinity binding molecules, known as TSPO ligands. TSPO ligands have been shown to have a beneficial effect by agonizing or inhibiting TSPO activity in diseases such as Alzheimer's disease (AD), multiple sclerosis and cardiac arrhythmias (1315). It was not until the previous decade, however, that the role of TSPO and its ligands in ocular diseases became an area of active research, and a number of promising developments have been identified (1620). The present article provided a review of the advances in the study of TSPO and its ligands in ocular diseases. First, an overview of TSPO function was presented, and a variety of classical and novel TSPO ligands were described. Next, the expression of TSPO in ocular tissues based on existing studies was summarized. Finally, the role of TSPO and its ligands in different ocular conditions was discussed, including ocular development and aging, age-related macular degeneration (AMD), retinal ischemia, diabetic retinopathy (DR) and glaucoma. The aim of the present review was to highlight the research value of TSPO in ophthalmology and to suggest perspectives on potential therapeutic targets.

Structure and distribution of TSPO

TSPO is a highly hydrophobic protein that is expressed in both prokaryotic and eukaryotic cells, from bacteria to humans (21). TSPO is primarily localized to the OMM, adjacent to the VDAC and adenine nucleotide translocase (ANT) (1,22,23). This 169-amino acid protein in humans is encoded by a nuclear gene containing four exons (24) and the mature protein contains five α-helical transmembrane domains (TMs). A cholesterol-recognition amino acid consensus sequence is located in the C-terminus of the TM structure, through which TSPO binds to the lipid membrane (25,26). The 3D structure of TSPO in complex with the TSPO-targeting ligand, (R)-1-(2-chlorophenyl- N-methylpropyl)-3-isoquinolinecarboxamide (PK11195), comprising five transmembrane helices (TM1-TM5) that tightly pack together in the clockwise order TM1-TM2-TM5-TM4-TM3 viewed from the cytosol, has also been revealed (26). TSPO is highly expressed in a variety of tissues, particularly in the gonads, adrenal glands, placenta and brain, due to its high abundance in steroidogenic cells (27). Therefore, the pathogenic effects of TSPO and the potential therapeutic value of TSPO ligands for anxiety disorders, AD and hypogonadism have been thoroughly investigated in vitro and in vivo (2830).

TSPO is involved in multiple cellular functions

TSPO is involved in a large variety of physiological cellular processes (Fig. 1). First, the subcellular localization of TSPO dictates its critical role in mitochondrial function; it is a regulator of mitochondrial respiration, membrane potential, energy homeostasis and quality control (4,3133). TSPO also reduces mitophagy, which inhibits essential ubiquitination of proteins, leading to a low efficiency of ubiquitin-dependent mitochondrial protein degradation, and thus, an accumulation of dysfunctional mitochondria (33). Previous studies have reported that TSPO modulates mitochondrial permeability transition pores (mPTPs), such as VDAC and ANT (1,5,9). However, there is conflicting evidence regarding these interactions, which suggests that TSPO is not a key mPTP regulator (8). Accordingly, these interactions remain ambiguous, and we hypothesized that these are possibly indirect or cell type-dependent. Another function of TSPO is to control intracellular signaling pathways between mitochondria and the nucleus, thereby affecting nuclear gene expression (6). Finally, TSPO is associated with multiple general cellular activities, such as cell proliferation, cell differentiation (34) and programmed cell death (35), and also acts as a gatekeeper for the biosynthesis of steroid hormones (8). TSPO is involved in the translocation of cholesterol and porphyrins from the outer to the inner mitochondrial membrane (22), which is a rate-limiting step in steroid and neurosteroid synthesis (36). TSPO also regulates calcium ion homeostasis in cells and activates calcium-dependent NADPH oxidase (NOX) (37), thereby increasing ROS levels (38,39). An additional study has demonstrated that TSPO is a key regulator of NOX1-dependent neurotoxic ROS production (40). In fact, the function of TSPO is still regarded as debatable, and thus, more future studies are required to elucidate this. For example, one of the most highly researched areas of TSPO is its key role in steroidogenesis. However, Tu et al (28,29) successfully established a TSPO systemic knockout mouse model and demonstrated that TSPO is not essential for steroidogenesis. In addition, some studies solely focused on TSPO ligand functions without measuring TSPO levels, thus it remains unclear whether TSPO had a direct impact on the biological events (14,25). Therefore, further experiments are required to explain the aforementioned unclear mechanisms in order to ascertain the role of TSPO.

TSPO ligands

TSPO ligands

TSPO ligands are classified as endogenous or synthetic ligands. In previous years, TSPO-ligand interactions have received increasing attention, with multiple studies reporting its positive effects on diseases such as AD, multiple sclerosis, anxiety and vasculopathy (1315,41). Steroidogenesis and anti-inflammation are the main pathways by which TSPO ligands function (42). However, the independent mechanism, ‘mitohormesis’, defined as subtoxic stress on mitochondria, may be responsible for the protective effects of TSPO ligands. It is proposed that the ligands suppress ATP synthesis through mitohormesis, leading to activation of the cellular damage-response network. This, in turn, enhances the tolerance of cells to the degeneration processes, including oxidative damage, phototoxicity or DNA damage (43). A summary of classical and novel ligands and their features and effects based on existing studies is described below and in Table I.

Table I.

Involved pathways or biological effects of TSPO ligands in in vitro and in vivo experimental models.

Table I.

Involved pathways or biological effects of TSPO ligands in in vitro and in vivo experimental models.

First author/s, yearTSPO ligandsCell line/animal modelFindings of involved pathways/biological effects(Refs.)
Yamamoto et al, 2014PPIXTSPO-knockdown glioblastoma cellsTSPO deletion leads to the accumulation of PPIX and ROS(49)
Girard et al, 2012; Ravikumar et al, 2016; Choi et al, 2002PK11195LPS-induced human microgliaDownregulates the expression of TNF-α and COX-2, probably relevant to Ca2+-mediated signaling pathways(5961)
Seneviratne et al, 2012 HeLa cellsRegulates cellular autophagy by targeting F1F0-ATP synthase, which is a TSPO-independent pathway(63)
Ishikawa et al, 2016 Rat ex vivo glaucoma modelInhibits pressure-induced RGC apoptosis by promoting the TSPO-5αRD-mediated AlloP synthesis(57)
Gut et al, 2013Ro5-4864 Isoprenaline-induced zebrafish larvaeReverses the glucose level fluctuations(70)
Bernardi et al, 2022; Baez et al, 2017 Glucose deprivation treated T98G astrocyte cellsMaintains mitochondrial homeostasis by reducing the production of free radicals and inhibits the mitochondrial apoptosis pathway by reducing cytochrome c release and caspase-3 activation(36,74)
Da Pozzo et al, 2015 AD mouse modelModulates the production of steroids to exert neuroprotective effects(71)
Musman et al, 2017 Type-2 diabetes mellitus rat modelsLimits cholesterol transport into mitochondria, inhibits oxysterol accumulation and reduces oxidative stress(139)
Nothdurfter et al, 2012XBD173Anxiety disorder population and mouse modelExerts anxiolytic properties by enhancing neurosteroidogenesis, thereby potentiating GABA-mediated inhibitory postsynaptic currents(75)
Scholz et al, 2015 Retinal degenerationAlleviates neurodegeneration by targeting the mouse model microglia/macrophage marker, CD68, and reducing the levels of pro-inflammatory factors, CCL2 and IL-6(78)
Mages et al, 2019 Retinal ischemia mouse modelReduces microglia and promotes the transformation toward the anti-inflammatory M2 phenotype, thereby protecting inner retinal neurons(18)
Lieth et al, 1998; Wagner et al, 2017; Delyfer et al, 2005 Retinal degeneration/ischemia mouse modelMaintains the metabolism of the ‘neuron-supportive microenvironment’ and protects against neurotoxic injury by regulating glutamine synthetase expression(123,124,126)
Wolf et al, 2020; Nagineni et al, 2012 Laser-induced CNV mouse modelInhibits the generation of CNV by inhibiting pro-angiogenic factor and autocrine, IL-1β, regulating phagocytosis and facilitating the transition of microglia to neuroprotective phenotypes(40,116)
Biswas et al, 2021; Ibrahim et al, 2020EtifoxineHigh-fat diet/obesity mouse modelEnhances cholesterol metabolism by downregulating the total cholesterol, triglyceride and phospholipid mass both in RPE and in serum(84,85)
Biswas et al, 2018; Biswas et al, 2017 Choroidal endothelial cells (RF/6A)Inhibits apoptosis by reducing intracellular lipid accumulation, ox-LDL-induced ROS production and inflammatory cytokine secretion(17,25)
Wang et al, 2014DBI/TTNRetinal inflammation mouse modelDBI-TSPO signaling limits the magnitude of microglial activation and promotes a return to baseline quiescence through macroglia-microglia interactions(16)
Santoro et al, 2016PIGASINFγ/LPS-induced rat C6 glioma cellsDownregulates pro-inflammatory enzymes, iNOS and COX-2, expression by modulating neurosteroid synthesis(91)

[i] TSPO, translocator protein; PPIX, protoporphyrin IX; ROS, reactive oxygen species; PK11195, 1-(2-chlorophenyl- N-methylpropyl)-3-isoquinolinecarboxamide; LPS, lipopolysaccharide; TNF-α, tumor necrosis factor-α; COX-2, cyclooxygenase-2; RGC, retinal ganglion cell; 5αRD, 5α-reductase; AlloP, allopregnanolone; Ro5-4864, 4′-chlorodiazepam; AD, Alzheimer's disease; CCL2, C-C motif chemokine receptor-2; IL-6, interleukin 6; RPE, retinal pigment epithelium; ox-LDL, oxidized low-density lipoprotein; CNV, choroidal neovascularization; DBI, diazepam-binding inhibitor; TTN, triakontatetraneuropeptide; PIGAS, phenylindolyilglyoxylamydes; INFγ, interferon γ; iNOS, inducible nitric oxide synthase.

Porphyrins

Porphyrins are high-affinity endogenous ligands for TSPO, in the micromolar to nanomolar range (35). Mitochondria are critically involved in porphyrin metabolism and TSPO is closely related to this process (26,44). Porphyrins are involved in heme synthesis, and protoporphyrin IX (PPIX), as a key intermediate in the heme biosynthesis pathway, has been one of the most extensively studied endogenous ligands since its discovery in 1987 (45,46). Previous studies have reported that TSPO-PPIX interactions regulate mitochondrial membrane permeability and thus exert cytoprotective effects (46,47). Porphyrin-induced oxidative stress is thought to be the major mechanism of porphyrin-mediated tissue damage (39,48). Furthermore, Yamamoto et al (49) demonstrated that TSPO knockdown leads to the accumulation of PPIX and ROS in glioblastoma cells. Specifically, TSPO possibly functions by binding or sequestering PPIX rather than via TSPO-mediated transmembrane transport (50). Unbound PPIX inhibits heme oxygenase and prevents free heme degradation, thereby maintaining the balance between free and bound heme and preventing cytotoxic effects (51).

PK11195

PK11195 was the first non-benzodiazepine TSPO synthetic ligand discovered that acts in the nanomolar range (42). PK11195 is considered one of the best-characterized and prototypic ligands of TSPO (50), and is widely used to study the roles and functions of TSPO in various diseases, particularly in in vivo studies. [11C]PK11195 has been additionally applied as a radioligand for positron emission tomography (PET). Despite several limitations, such as poor brain permeability, kinetic instability and signal-to-noise ratio issues due to non-specific binding (5254), [11C]PK11195 remains the standard TSPO-based PET radiotracer for assessing the activation of microglia or macrophages in central nervous system disorders (55,56). PK11195 has also been reported to have anti-neuroinflammation effects in experimental models of AD (10) and pressure-induced retinal ganglion cell (RGC) injury (57). However, further research is needed to elucidate the specific mechanisms underlying these protective effects, including whether they are related to the stimulation effect on steroidogenesis in peripheral tissues and the brain (10,58). PK11195 downregulates the expression of the inflammatory cytokines, tumor necrosis factor-α (TNF-α) and cyclooxygenase-2 (COX-2), in lipopolysaccharide (LPS)-induced human microglia (59,60), and modulation of Ca2+-mediated signaling pathways may be involved in its anti-inflammatory actions (61). In prostate cancer cells, PK11195 can be used as a sensitizer to the chemotherapy agents mda-7/IL-24 (Ad.mda-7, a novel cytokine gene belonging to the IL-10 gene superfamily) (62), both in vitro and in vivo. PK11195 promotes cellular autophagy by inactivating the oncoprotein, B-cell lymphoma-2, and by targeting F1F0-ATP synthase (63), an essential component of mPTPs (64). Notably, Gonzalez-Polo et al (65) reported that PK11195 promotes starvation-induced cell death through an unexpected pathway independent of TSPO, which directly activates the mitochondrial apoptotic pathway, as demonstrated by cytochrome c release and caspase-3 activation. Although PK11195 is generally considered to be a TSPO antagonist, there is evidence that PK11195 displays a partial agonistic effect on TSPO (66), which is possibly dependent on the cell type and drug concentration.

4′-chlorodiazepam (Ro5-4864)

Ro5-4864 is another classical synthetic TSPO ligand that has been used to investigate the characteristics of TSPO in health and disease due to its nanomolar selectivity (67,68). However, the application of Ro5-4864 as a pharmacological agent (67) is limited as its affinity varies with species, from rat (high affinity) to human (low affinity) (21). Another limitation is that the affinity of [3H] Ro5-4864 can be influenced by temperature, as found in brain membrane binding assays (69). Gut et al (70) reported that Ro5-4864 is sufficient to reverse the glucose level fluctuations induced by isoprenaline in zebrafish larvae, suggesting that Ro5-4864 may influence glucose homoeostasis in response to altered mitochondrial energy balance. Ro5-4864 also has certain features that are similar to that of PK11195. For example, Ro5-4864 stimulates steroid formation (10), contributes to the neuroprotective effect (but differentiates from PK11195 activity under different experimental conditions) (71) and inhibits pro-inflammatory factors and ROS production in human microglia incubated with LPS (16,59). Furthermore, Ro5-4864 antagonizes the anxiolytic and antidepressant-like effects of other TSPO ligands. For example, the antidepressant properties of PK11195 induced in mice subjected to the forced swimming test was blocked by Ro5-4864 administration (72,73). Ro5-4864 has also been shown to contribute to the maintenance of mitochondrial homeostasis, reducing cytochrome c release and caspase-3 activation, thereby inhibiting the mitochondrial apoptosis pathway (36). Furthermore, Baez et al (74) demonstrated that Ro5-4864 maintains mitochondrial function in the T89G astrocyte cell line in the presence of glucose deprivation damage by reducing the production of free radicals.

XBD173

XBD173 (AC-5216; Emapunil) is a novel high-affinity and selective phenylpurine ligand of TSPO, which has been well investigated and widely used in the research and therapy of anxiety disorders (41,75,76). GABA-mediated inhibitory postsynaptic currents were potentiated by XBD173 in both animal experiments and clinical studies (75), and this neurotransmission appeared to be mediated indirectly through the generation of neurosteroids and could be prevented by the 5α-reductase inhibitor, finasteride (76). According to previous studies, the role of XBD173 in microglia has been well defined and has been shown to alleviate the neurotoxic effect of microglia and inhibit cell proliferation and migration (16,77). The XBD173/TSPO axis appears to target the microglia/macrophage marker, CD68, and inhibit the pro-inflammatory genes, C-C motif chemokine receptor-2 and interleukin (IL-)6, to alleviate apoptosis, thereby alleviating neurodegenerative dysfunction (78). Mages et al (18) further investigated the exact response patterns of the major glial cell types of the retina in a retinal ischemia mouse model. In the study it was reported that XBD173 treatment resulted in lower microglia accumulation, and the microglial activation profile showed a transformation towards the anti-inflammatory M2 phenotype. However, the aforementioned effects of XBD173 on microglia could be reversed to some extent, since the reduced microglial cell number induced by XBD173 could transiently rise again (18). In addition, even with XBD173 intervention, cellular morphological changes (from ramified to amoeboid) representing microglial activation were observed, suggesting that XBD173 only partially inhibits microglial activation (18).

Etifoxine

To the best of our knowledge, Etifoxine (etafenoxine; Stresam®) is currently the only commercially available TSPO ligand (79). Etifoxine was developed in the 1960s for anxiety disorders and is administered for this purpose in >42 different countries (80). However previous studies have since reported its therapeutic potential in peripheral nerve injury and demyelination, and chemotherapy-induced pain (14,80,81). As a non-benzodiazepine anxiolytic, etifoxine appears to exercise its functions by binding to the β2 or β3 subunits of the GABAA receptor complex (82). Etifoxine also regulates GABAA receptors indirectly by stimulating neurosteroid production in both the central and peripheral nervous systems (24,83). Furthermore, Biswas et al (84) suggested that etifoxine modulates cholesterol homeostasis, promoting cholesterol efflux and transporting cholesterol to the liver, which is the most important function of this ligand. The authors also found that etifoxine enhanced the expression of cholesterol metabolism and transport enzymes in high-fat diet mice and significantly decreased the total cholesterol, triglycerides and phospholipid mass in the retinal pigment epithelium (RPE). This finding was consistent with that of a study by Ibrahim et al (85), which suggested that serum lipid levels were downregulated in etifoxine-treated mice. Furthermore, etifoxine leads to a higher conversion of cholesterol into high-density lipoprotein while preventing intracellular lipid accumulation, and therefore protecting cells against apoptosis by inhibiting oxidized low-density lipoprotein-induced ROS production and inflammatory cytokine secretion (17,25). In addition, etifoxine stimulates neurosteroid synthesis; however, Costa et al (86) suggested that this effect depends more on residence time at the TSPO binding site rather than the binding affinities.

Other reported TSPO ligands

Diazepam-binding inhibitor (DBI) is an endogenous agonistic ligand of TSPO (3). DBI-TSPO interactions have been demonstrated to occur in the production of steroids such as pregnenolone, in both in vivo and in vitro experiments (87,88). DBI or its cleavage product, triakontatetraneuropeptide, binds to TSPO during a form of macroglia-microglia interaction, limiting the magnitude of microglial activation and promoting a return of microglia to the resting state, which is a hallmark of the downregulation of inflammation (16). Novel TSPO synthetic ligands include N,N-di-n-hexyl 2-(4-fluorophenyl) indole-3-acetamide (FGIN-1-27), 7-chloro-N,N,5-trimethyl-4-oxo-3-phenyl-3,5-dihydro-4H-pyridazino(4,5-b)indole-1-acetamide (SSR180575) and phenylindolyilglyoxylamydes (PIGAS), all of which possess steroidogenic properties (12) and promote cholesterol metabolism and transporter gene expression (25). SSR180575 is anti-apoptotic and protects mitochondria against dysfunction in ischemia-reperfusion injury models (89,90). PIGAS displays anti-inflammatory capacity, as evidenced by decreased inducible nitric oxide synthase and COX-2 expression, possibly by modulating astrocyte function through neurosteroid synthesis (91).

Functional roles of TSPO and TSPO ligands in ocular diseases

Overview

Next, the expression of TSPO in ocular tissues, the mechanisms by which TSPO is involved in the progression of various ocular diseases (Fig. 2) and the role of TSPO ligands in therapeutic interventions are discussed, based on existing studies.

Current status of research on TSPO expression in ocular tissues

In recent years, the role of TSPO in ocular diseases has gained attention. However, it must be acknowledged that the number of studies on TSPO expression in ocular cells and tissues has been limited. Biswas et al (17,25) reported high TSPO expression in the RPE and choroidal endothelium of mice. This observation was consistent with that of a study by Mages et al (18), in which specific cell types were enriched via immunomagnetic separation and then protein expression levels were measured using mass spectrometry analysis. It was noted that TSPO was most highly expressed in RPE cells, vascular cells and Müller cells, while expression was lower in microglia and almost undetectable in retinal neurons (16,18,77). By contrast, Biswas et al (17) reported strong TSPO signals in the ganglial cell layer by immunofluorescence analysis in mouse eye sections. Differences in TSPO expression and localization may be due to differences in sample collection and immunoassay approaches. In addition, TSPO expression has been detected in human retina and RPE (77,92,93), with both containing high expression at the mRNA level. When the central and peripheral sections of the retina and RPE were isolated separately, the peripheral RPE exhibited higher TSPO expression than the central RPE (92). It remains unclear whether this infers any functional implications. To the best of our knowledge, there have been no further reports of TSPO expression in other ocular structures such as the cornea, conjunctiva, lens, meibomian gland and lacrimal gland.

TSPO in ocular development and aging

Wang et al (16) investigated whether the endogenous functions of TSPO expression are relevant to retinal developmental processes. TSPO is expressed in microglia with an amoeboid phenotype that invades the neural retina during early development. In adulthood, as microglia undergo a morphological transformation to a ramified cell shape, TSPO expression is barely detectable (94). There are several theories that might help explain this, including that TSPO is possibly involved in the physiological processes of microglial development (95,96), such as complement-mediated synapse elimination and phagocytosis of dead neurons (66). Temporary expression of TSPO may also be associated with the migration process of microglia during retina development (97), and the expression terminated when microglia reach the final site. On the other hand, certain researchers suggest that TSPO is unnecessary for healthy development and aging following the study of several generated systemic TSPO-knockout mouse lines (28,98,99). For example, Klee et al (92) considered that ocular development is not strongly associated with TSPO expression as no ocular phenotype was observed in TSPO-knockout mice aged 0–48 weeks old. Furthermore, TSPO mRNA expression did not show significant differences in human retina and RPE tissues among different age groups of donors (from 17–90 years old) (92), suggesting that TSPO expression may not be involved in the regulation of normal development and aging. These differential findings led us to consider whether there are compensatory mechanisms for TSPO deletion in the knockout models such that marked phenotypes are not evident during the development process. Furthermore, as a stress-related protein, the regulatory effects of TSPO in multiple pathways may be more critical in disease states but not play a major role in normal physiological development. At present, the potential mechanisms remain to be validated by additional studies.

AMD

AMD is a prevalent cause of severe visual impairment among elderly individuals (100). Clinically, AMD can be categorized into two subtypes: Dry AMD, which accounts for 80–85% of cases and manifests as geographic atrophy, and wet AMD, which accounts for 15–20% of cases and presents as neovascularization (101). The pathogenesis of AMD is multifaceted, with a combination of genetic and environmental factors, while the efficacy of existing therapies is extremely limited since there is no treatment available to prevent the irreversible degeneration of the photoreceptors that leads to loss of central vision (102,103). Dysfunctional cholesterol metabolism is one of the prominent mechanisms underlying the development of AMD (103). The pathological examination of patients with AMD has revealed subretinal deposits of apolipoproteins, cholesterol and cholesteryl ester, indicating a potential association of cholesterol transport disorders in AMD (17,104). Notably, the formation of drusen is associated with poor choroidal endothelial structural and functional integrity, resulting in inadequate nutrient and oxygen delivery to the RPE and photoreceptor cells and impairing their metabolite clearance (105). These processes are particularly associated with the pathogenesis of wet AMD (105,106). Aberrant expression of cholesterol homeostasis genes may lead to cholesterol and lipid dysregulation, thereby worsening AMD progression (107,108). This has been demonstrated in several experimental animal models showing AMD lesion progression (107,109,110). In addition, the release of inflammatory factors, the accumulation of ROS and the phagocytosis of neurons are involved in photoreceptor apoptosis in AMD (111113). TSPO-knockout mice exhibit AMD-like features, including impaired cholesterol efflux, increased cholesterol, triglycerides and phospholipids in the RPE and choroid, increased levels of inflammatory factors (IL-1β and TNF-α) and downregulation of cholesterol-related genes (Nr1h3, Abca1, Abcg1, Cyp27a1 and Cyp46a1) (114). In a laser-induced choroidal neovascularization (CNV) model, TSPO regulated NOX1-mediated paracrine secretion of ROS, which is a crucial pathway for promoting pro-photoreceptor cell apoptosis (40). By contrast, angiogenic factors are generated via a non-NOX-dependent pathway, which can be reversed by TSPO ligand treatment (115). These findings suggest that TSPO deficiency may contribute to the core pathogenesis of AMD. The TSPO ligand, etifoxine, upregulated cholesterol homeostasis gene expression in both RPE and choroidal vascular endothelial cells, promoting cholesterol efflux in a mouse AMD model (25,84). In addition, in human RPE cells (ARPE-19 cells), the FGIN-1-27, XBD173 and etifoxine ligands also significantly promote cholesterol efflux, whereas in TSPO knockdown RPE cells, this therapeutic effect is reversed (17). The aforementioned results imply that targeting the TSPO-mediated cholesterol efflux process may be effective in improving subretinal lipid accumulation in AMD. ROS production in RPE and choroid endothelial cells is also downregulated following etifoxine treatment (84). For neovascular AMD, XBD173 treatment inhibits the generation of CNV, which is thought to be associated with inhibiting pro-angiogenic factor (Vegf, Ang1 and Ang2) expression, regulating phagocytosis and facilitating the transition of microglia to neuroprotective phenotypes (40). In addition, inhibition of autocrine IL-1β levels in RPE, which has previously been shown to induce VEGF production, also provides a similar protective effect (116). In general, the mechanisms by which CNVs are formed are complex, and it is likely that TSPO ligands do not inhibit CNVs via a single pathway; thus, specific mechanisms warrant further exploration.

Retinal ischemia

Retinal ischemia is a major cause of visual impairment, often leading to irreversible morphological and functional injuries with depleted ATP stores (117). Extensive research has been devoted to clarifying the mechanisms of ischemia-induced neuronal damage, although debates persist. Retinal ischemic injury involves a cascade of self-reinforcing and destructive events, including neuronal depolarization, calcium imbalance and oxidative stress due to increased energy depletion and enhanced glutamate stimulation (118121). TSPO was discovered to be upregulated in microglia during the early stages of retinal ischemia injury, which then decreased 4 days later (16). TSPO expression increases over time in Müller cells, which is the primary immune cell type in the retina, indicating an association between TSPO and the active retinal immune response during ischemic injury (16,18). A number of studies have shown that TSPO is closely associated with microglial activation and microglial energy metabolism (98,122). Thus, the transient increase in TSPO expression may be due to a microglia-mediated immune response or an acute response to an imbalanced energy supply in the postischemic phase. Furthermore, mice treated with the TSPO ligand, XBD173, have a significant reduction in neuron death in the postischemic retina. In addition, they have a decreased microglial number accompanied by phenotypic transformation from the M1 type to the M2 type, indicating anti-inflammatory activation (18). This finding concurred with those of Wang et al (16) and Karlstetter et al (77), who found that XBD173 inhibited microglial neurotoxicity and proliferation. The effect of XBD173 on Müller cells was also notable since it increased the level of DBI in postischemic Müller cells (18). As aforementioned, DBI an endogenous TSPO ligand, can alleviate neuroinflammation and induce microglia to favor the resting state (16). XBD173 regulates glutamine synthetase expression, thereby maintaining the metabolism of a ‘neuron-supportive microenvironment’ and protecting against glutamate-overload induced neurotoxic injury (123126). It should be noted that the aforementioned effect of XBD173 was incomplete, as the transient upregulation of inflammatory markers as well as the downregulation of glutamine synthetase were still detected following XBD173 therapy. Given the high abundance of TSPO in the RPE, its role in retinal ischemia injury should not be overlooked. However, to the best of our knowledge, no relevant studies on the association between TSPO and RPE injury in retinal ischemia have been published to date. In summary, TSPO is involved in the pathogenic reactions of specific cell types to retinal ischemia and XBD173 treatment increased retinal neuron survival, to a certain extent.

DR

DR is the most prevalent complication of diabetes mellitus (DM) (127) and adversely affects the vision of working-aged individuals (128,129). The pathogenesis of DM and DR is complicated and unclear and treatment remains challenging due to recurrent vitreous hemorrhage and progressive loss of function (121). Hyperglycemia is considered to be a leading cause of metabolic disorders in the retinal vasculature via various pathways (130), including the protein kinase C, polyol and hexosamine pathways (131,132). Inflammation and dysfunction of glial cells are also involved in the onset and progression of DR (133,134). Other reported mechanisms of DR include mitochondrial dysfunction and oxidative stress damage, both of which accelerate retinal neuronal apoptosis (135137). Correspondingly, TSPO has been suggested to play a role in the regulation of glucose metabolism, and the TSPO ligands, Ro5-4864 and PK11195, were both found to reduce the overall glucose levels in zebrafish larvae (70). Moreover, activation of TSPO regulates glucose homeostasis in adipocytes and it has been suggested that the TSPO ligands, PK11195 and FGIN-1-27, increase adipogenesis and glucose absorption by regulating mitochondrial activity and cholesterol transport (138). In addition, Ro5-4864 protects rat models of type-2 DM from metabolic disorders by limiting cholesterol transport into mitochondria, lowering oxysterol accumulation and reducing oxidative stress (139). Furthermore, TSPO relieves diabetic neuropathy by reportedly raising neuroactive steroid levels (11). Thus, TSPO has been identified as a potential target for diabetes management. Despite the increasing number of studies regarding the association between TSPO and DM complications, few investigations on the mechanism of TSPO in DR have been conducted. Zhou et al (20) found that TSPO was downregulated in rat models of type-2 DM 12 weeks after commencement of disease modeling. TSPO has previously been proposed to be an appropriate candidate marker of microglial activation (16); it was shown to be transiently upregulated in the early stages of retinal injury along with stress-induced microglial activation, while its expression was lower than that of controls or even absent after microglia returned to a resting state (140,141). Therefore, Zhou et al (20) hypothesized that the downregulation of TSPO in the retina indicated the activation of microglia in the retina during the early stages of diabetes. However, this hypothesis requires further study, as microglial activation was not consistently observed in the diabetic models, nor were changes in TSPO expression levels. Guo et al (19) reported significantly higher expression levels of TSPO in the peripheral blood of patients with diabetes than in the controls, and a higher TSPO level in the active proliferative DR subgroup than in the inactive proliferative DR subgroup. Moreover, Guo et al (19) found that the TSPO/VDAC complex positively associates with several inflammatory cytokines, such as NOD-like receptor pyrin domain-containing 3, suggesting an important role in DR development and progression.

Glaucoma

Glaucoma is a leading cause of irreversible blindness worldwide and is characterized by progressive optic nerve injury and visual field loss (142,143). Glaucoma is a multifactorial disorder and elevated intraocular pressure is thought to be a major risk factor, leading to morphological deformation of the lamina cribrosa and disturbance of axoplasmic transport in RGC axons (144). Acute or chronic deficiency of these RGC neurotrophic signals ultimately leads to apoptosis (144). Other related pathogeneses of glaucoma include ocular hemodynamic abnormalities, low intracranial pressure, autoimmune dysfunction and mitochondrial dysfunction, while further studies are needed to investigate the detailed mechanisms of RGC injury (145). The neurosteroid, allopregnanolone (AlloP), has been shown to synthesize and exert neuroprotective effects on the retina through the GABAA receptor pathway in a rat model of acute glaucoma in vitro (146,147). As a major mitochondrial cholesterol carrier, TSPO mediates the transport of cholesterol into the mitochondria, which is a key process in the synthesis of pregnenolone (57). Pregnenolone then functions as the starting material for AlloP synthesis (83,148). Under elevated pressure (75 mmHg), TSPO expression is found to be upregulated and TSPO colocalizes with AlloP and 5α-reductase (5αRD; a rate-limiting enzyme in the AlloP synthesis pathway) in RGCs, suggesting that RGCs are the principal site of AlloP synthesis (57). Furthermore, the TSPO antagonist, atriol, significantly inhibits the production of AlloP (149), suggesting that TSPO is important in AlloP synthesis. In addition, atriol induces changes in retinal excitotoxicity, including edema-like changes in the inner plexiform layer and bull's eye formation in the inner nuclear layer (150,151). Moreover, the TSPO ligand, PK11195, acts as a selective agonist that promotes TSPO expression and inhibits pressure-induced RGC apoptosis (57). Atriol and 5αRD inhibitors (24) reverse this effect, suggesting that PK11195 may exert neuroprotective effects by promoting the TSPO-5αRD-mediated AlloP synthesis pathway, as aforementioned. Hence, TSPO agonists may have therapeutic potential for ocular hypertension injury.

Conclusion

Overall, the present review concluded that TSPO is broadly involved in multiple cellular processes as well as mitochondrial functions. TSPO participates in the occurrence and development of various ocular diseases, including AMD, DR, retinal ischemia and glaucoma, with evidence of both protective and aggravating effects in the observed pathology of the diseases. There is increasing evidence that endogenous or synthetic TSPO ligands, which modulate TSPO functions, play beneficial roles in the aforementioned diseases primarily by promoting steroidogenesis, maintaining cholesterol homeostasis, exerting anti-inflammatory effects, decreasing ROS production and regulating microglial activation. These findings suggest that TSPO may be a key potential therapeutic target. Despite the progress made in understanding the role of TSPO in ocular diseases, the expression of TSPO in ocular surface tissues, the lens, the lacrimal gland and the optic nerve and whether TSPO plays a key role in these diseases remain to be investigated. Moreover, whether TSPO is an essential protein for ocular tissue in healthy individuals is another question that requires more extensive study. At present, the use of TSPO ligands in the ocular environment is limited and there is a lack of comprehensive evaluation of their potential adverse effects. Only by addressing these issues can TSPO and its ligands be applied on a larger scale.

Acknowledgements

Not applicable.

Funding

The present research was funded by Tianjin Key Medical Discipline (Specialty) Construction Project (grant no. TJYXZDXK-037A).

Availability of data and materials

Not applicable.

Authors' contributions

MY and SZ designed the study; MY performed the figure preparation and manuscript draft; and SZ reviewed and edited the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

TSPO

translocator protein

OMM

outer mitochondrial membrane

VDAC

voltage-dependent anion channel

ANT

adenine nucleotide translocase

mPTPs

mitochondrial permeability transition pores

ROS

reactive oxygen species

NOX

NADPH oxidase

AD

Alzheimer's disease

PPIX

protoporphyrin IX

PK11195

1-(2-chlorophenyl-N-methylpropyl)-3-isoquinolinecarboxamide

Ro5-4864

4′-chlorodiazepam

DBI

diazepam-binding inhibitor

PIGAS

phenylindolyilglyoxylamydes

FGIN-1-27

N,N-di-n-hexyl 2-(4-fluorophenyl) indole-3-acetamide

PET

positron emission tomography

TNF-α

tumor necrosis factor-α

COX-2

cyclooxygenase-2

LPS

lipopolysaccharide

RGC

retinal ganglion cell

RPE

retinal pigment epithelium

AMD

age-related macular degeneration

CNV

choroidal neovascularization

DR

diabetic retinopathy

DM

diabetes mellitus

References

1 

Shoshan-Barmatz V, Pittala S and Mizrachi D: VDAC1 and the TSPO: Expression, Interactions, and associated functions in health and disease states. Int J Mol Sci. 20:33482019. View Article : Google Scholar : PubMed/NCBI

2 

Braestrup C and Squires RF: Specific benzodiazepine receptors in rat brain characterized by high-affinity (3H)diazepam binding. Proc Natl Acad Sci USA. 74:3805–3809. 1977. View Article : Google Scholar : PubMed/NCBI

3 

Papadopoulos V, Baraldi M, Guilarte TR, Knudsen TB, Lacapère JJ, Lindemann P, Norenberg MD, Nutt D, Weizman A, Zhang MR and Gavish M: Translocator protein (18kDa): New nomenclature for the peripheral-type benzodiazepine receptor based on its structure and molecular function. Trends Pharmacol Sci. 27:402–409. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Tu LN, Zhao AH, Hussein M, Stocco DM and Selvaraj V: Translocator Protein (TSPO) affects mitochondrial fatty acid oxidation in steroidogenic cells. Endocrinology. 157:1110–1121. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Bonsack F and Sukumari-Ramesh S: TSPO: An evolutionarily conserved protein with elusive functions. Int J Mol Sci. 19:16942018. View Article : Google Scholar : PubMed/NCBI

6 

Fan J and Papadopoulos V: Mitochondrial TSPO deficiency triggers retrograde signaling in MA-10 mouse tumor leydig cells. Int J Mol Sci. 22:2522020. View Article : Google Scholar : PubMed/NCBI

7 

Vainshtein A, Veenman L, Shterenberg A, Singh S, Masarwa A, Dutta B, Island B, Tsoglin E, Levin E, Leschiner S, et al: Quinazoline-based tricyclic compounds that regulate programmed cell death, induce neuronal differentiation, and are curative in animal models for excitotoxicity and hereditary brain disease. Cell Death Discov. 1:150272015. View Article : Google Scholar : PubMed/NCBI

8 

Sileikyte J, Blachly-Dyson E, Sewell R, Carpi A, Menabò R, Di Lisa F, Ricchelli F, Bernardi P and Forte M: Regulation of the mitochondrial permeability transition pore by the outer membrane does not involve the peripheral benzodiazepine receptor (Translocator Protein of 18 kDa (TSPO)). J Biol Chem. 289:13769–13781. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Ilkan Z and Akar FG: The mitochondrial translocator protein and the emerging link between oxidative stress and arrhythmias in the diabetic heart. Front Physiol. 9:15182018. View Article : Google Scholar : PubMed/NCBI

10 

Arbo BD, Ribeiro MF and Garcia-Segura LM: Development of new treatments for Alzheimer's disease based on the modulation of translocator protein (TSPO). Ageing Res Rev. 54:1009432019. View Article : Google Scholar : PubMed/NCBI

11 

Mitro N, Cermenati G, Giatti S, Abbiati F, Pesaresi M, Calabrese D, Garcia-Segura LM, Caruso D and Melcangi RC: LXR and TSPO as new therapeutic targets to increase the levels of neuroactive steroids in the central nervous system of diabetic animals. Neurochem Int. 60:616–621. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Veenman L and Gavish M: The peripheral-type benzodiazepine receptor and the cardiovascular system. Implications for drug development. Pharmacol Ther. 110:503–524. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Barron AM, Garcia-Segura LM, Caruso D, Jayaraman A, Lee JW, Melcangi RC and Pike CJ: Ligand for translocator protein reverses pathology in a mouse model of Alzheimer's disease. J Neurosci. 33:8891–8897. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Daugherty DJ, Selvaraj V, Chechneva OV, Liu XB, Pleasure DE and Deng W: A TSPO ligand is protective in a mouse model of multiple sclerosis. EMBO Mol Med. 5:891–903. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Wu LP, Gong ZF, Wang H, Zhou ZS, Zhang MM, Liu C, Ren HM, Yang J, Han Y and Zeng CY: TSPO ligands prevent the proliferation of vascular smooth muscle cells and attenuate neointima formation through AMPK activation. Acta Pharmacol Sin. 41:34–46. 2020. View Article : Google Scholar : PubMed/NCBI

16 

Wang M, Wang X, Zhao L, Ma W, Rodriguez IR, Fariss RN and Wong WT: Macroglia-microglia interactions via TSPO signaling regulates microglial activation in the mouse retina. J Neurosci. 34:3793–3806. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Biswas L, Zhou X, Dhillon B, Graham A and Shu X: Retinal pigment epithelium cholesterol efflux mediated by the 18 kDa translocator protein, TSPO, a potential target for treating age-related macular degeneration. Hum Mol Genet. 26:4327–4339. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Mages K, Grassmann F, Jagle H, Rupprecht R, Weber BHF, Hauck SM and Grosche A: The agonistic TSPO ligand XBD173 attenuates the glial response thereby protecting inner retinal neurons in a murine model of retinal ischemia. J Neuroinflammation. 16:432019. View Article : Google Scholar : PubMed/NCBI

19 

Guo Y, Sun Z, Wang L, Jiang R, Shu Q and Xu G: Increased expression of TSPO-VDAC complex is correlated with NLRP3 inflammasome activation in diabetic retinopathy. Mol Med Rep. 26:3532022. View Article : Google Scholar : PubMed/NCBI

20 

Zhou Y, Ou Y, Ju Z, Zhang X, Zheng L, Li J, Sun Y and Liu X: Visualization of translocator protein (18 kDa) (TSPO) in the retina of diabetic retinopathy rats using fluorine-18-DPA-714. Ann Nucl Med. 34:675–681. 2020. View Article : Google Scholar : PubMed/NCBI

21 

Lacapere JJ, Duma L, Finet S, Kassiou M and Papadopoulos V: Insight into the Structural Features of TSPO: Implications for drug development. Trends Pharmacol Sci. 41:110–122. 2020. View Article : Google Scholar : PubMed/NCBI

22 

Papadopoulos V, Aghazadeh Y, Fan J, Campioli E, Zirkin B and Midzak A: Translocator protein-mediated pharmacology of cholesterol transport and steroidogenesis. Mol Cell Endocrinol. 408:90–98. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Jaremko M, Jaremko L, Jaipuria G, Becker S and Zweckstetter M: Structure of the mammalian TSPO/PBR protein. Biochem Soc Trans. 43:566–571. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Rupprecht R, Papadopoulos V, Rammes G, Baghai TC, Fan J, Akula N, Groyer G, Adams D and Schumacher M: Translocator protein (18 kDa) (TSPO) as a therapeutic target for neurological and psychiatric disorders. Nat Rev Drug Discov. 9:971–988. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Biswas L, Farhan F, Reilly J, Bartholomew C and Shu X: TSPO ligands promote cholesterol efflux and suppress oxidative stress and inflammation in choroidal endothelial cells. Int J Mol Sci. 19:37402018. View Article : Google Scholar : PubMed/NCBI

26 

Jaremko L, Jaremko M, Giller K, Becker S and Zweckstetter M: Structure of the mitochondrial translocator protein in complex with a diagnostic ligand. Science. 343:1363–1366. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Morohaku K, Pelton SH, Daugherty DJ, Butler WR, Deng W and Selvaraj V: Translocator protein/peripheral benzodiazepine receptor is not required for steroid hormone biosynthesis. Endocrinology. 155:89–97. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Tu LN, Morohaku K, Manna PR, Pelton SH, Butler WR, Stocco DM and Selvaraj V: Peripheral benzodiazepine receptor/translocator protein global knock-out mice are viable with no effects on steroid hormone biosynthesis. J Biol Chem. 289:27444–27454. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Tu LN, Zhao AH, Stocco DM and Selvaraj V: PK11195 effect on steroidogenesis is not mediated through the translocator protein (TSPO). Endocrinology. 156:1033–1039. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Zhao AH, Tu LN, Mukai C, Sirivelu MP, Pillai VV, Morohaku K, Cohen R and Selvaraj V: Mitochondrial translocator protein (TSPO) function is not essential for heme biosynthesis. J Biol Chem. 291:1591–1603. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Casellas P, Galiegue S and Basile AS: Peripheral benzodiazepine receptors and mitochondrial function. Neurochem Int. 40:475–486. 2002. View Article : Google Scholar : PubMed/NCBI

32 

Corsi L, Geminiani E and Baraldi M: Peripheral benzodiazepine receptor (PBR) new insight in cell proliferation and cell differentiation review. Curr Clin Pharmacol. 3:38–45. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Gatliff J, East D, Crosby J, Abeti R, Harvey R, Craigen W, Parker P and Campanella M: TSPO interacts with VDAC1 and triggers a ROS-mediated inhibition of mitochondrial quality control. Autophagy. 10:2279–2296. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Varga B, Marko K, Hadinger N, Jelitai M, Demeter K, Tihanyi K, Vas A and Madarász E: Translocator protein (TSPO 18kDa) is expressed by neural stem and neuronal precursor cells. Neurosci Lett. 462:257–262. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Veenman L and Gavish M: The role of 18 kDa mitochondrial translocator protein (TSPO) in programmed cell death, and effects of steroids on TSPO expression. Curr Mol Med. 12:398–412. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Bernardi P, Carraro M and Lippe G: The mitochondrial permeability transition: Recent progress and open questions. FEBS J. 289:7051–7074. 2022. View Article : Google Scholar : PubMed/NCBI

37 

Loth MK, Guariglia SR, Re DB, Perez J, de Paiva VN, Dziedzic JL, Chambers JW, Azzam DJ and Guilarte TR: A novel interaction of translocator protein 18 kDa (TSPO) with NADPH oxidase in microglia. Mol Neurobiol. 57:4467–4487. 2020. View Article : Google Scholar : PubMed/NCBI

38 

Gatliff J, East DA, Singh A, Alvarez MS, Frison M, Matic I, Ferraina C, Sampson N, Turkheimer F and Campanella M: A role for TSPO in mitochondrial Ca2+ homeostasis and redox stress signaling. Cell Death Dis. 8:e28962017. View Article : Google Scholar : PubMed/NCBI

39 

Zeno S, Veenman L, Katz Y, Bode J, Gavish M and Zaaroor M: The 18 kDa mitochondrial translocator protein (TSPO) prevents accumulation of protoporphyrin IX. Involvement of reactive oxygen species (ROS). Curr Mol Med. 12:494–501. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Wolf A, Herb M, Schramm M and Langmann T: The TSPO-NOX1 axis controls phagocyte-triggered pathological angiogenesis in the eye. Nat Commun. 11:27092020. View Article : Google Scholar : PubMed/NCBI

41 

Rupprecht R, Rammes G, Eser D, Baghai TC, Schüle C, Nothdurfter C, Troxler T, Gentsch C, Kalkman HO, Chaperon F, et al: Translocator protein (18 kD) as target for anxiolytics without benzodiazepine-like side effects. Science. 325:490–493. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Barresi E, Robello M, Costa B, Da Pozzo E, Baglini E, Salerno S, Da Settimo F, Martini C and Taliani S: An update into the medicinal chemistry of translocator protein (TSPO) ligands. Eur J Med Chem. 209:1129242021. View Article : Google Scholar : PubMed/NCBI

43 

Gut P: Targeting mitochondrial energy metabolism with TSPO ligands. Biochem Soc Trans. 43:537–542. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Taketani S, Kohno H, Furukawa T and Tokunaga R: Involvement of peripheral-type benzodiazepine receptors in the intracellular transport of heme and porphyrins. J Biochem. 117:875–880. 1995. View Article : Google Scholar : PubMed/NCBI

45 

Snyder SH, Verma A and Trifiletti RR: The peripheral-type benzodiazepine receptor: A protein of mitochondrial outer membranes utilizing porphyrins as endogenous ligands. FASEB J. 1:282–288. 1987. View Article : Google Scholar : PubMed/NCBI

46 

Veenman L, Vainshtein A, Yasin N, Azrad M and Gavish M: Tetrapyrroles as endogenous TSPO ligands in eukaryotes and prokaryotes: Comparisons with synthetic ligands. Int J Mol Sci. 17:8802016. View Article : Google Scholar : PubMed/NCBI

47 

Tonon MC, Vaudry H, Chuquet J, Guillebaud F, Fan J, Masmoudi-Kouki O, Vaudry D, Lanfray D, Morin F, Prevot V, et al: Endozepines and their receptors: Structure, functions and pathophysiological significance. Pharmacol Ther. 208:1073862020. View Article : Google Scholar : PubMed/NCBI

48 

Ma Z, An R, Chen M, Wang X and Zhu M: Random versus Block Glycopolymers bearing betulin and porphyrin for enhanced photodynamic therapy. Biomacromolecules. 23:5074–5083. 2022. View Article : Google Scholar : PubMed/NCBI

49 

Yamamoto M, Arimura H, Fukushige T, Minami K, Nishizawa Y, Tanimoto A, Kanekura T, Nakagawa M, Akiyama S and Furukawa T: Abcb10 role in heme biosynthesis in vivo: Abcb10 knockout in mice causes anemia with protoporphyrin IX and iron accumulation. Mol Cell Biol. 34:1077–1084. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Batoko H, Veljanovski V and Jurkiewicz P: Enigmatic Translocator protein (TSPO) and cellular stress regulation. Trends Biochem Sci. 40:497–503. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Korolnek T and Hamza I: Like iron in the blood of the people: The requirement for heme trafficking in iron metabolism. Front Pharmacol. 5:1262014. View Article : Google Scholar : PubMed/NCBI

52 

Chauveau F, Boutin H, Van Camp N, Dolle F and Tavitian B: Nuclear imaging of neuroinflammation: A comprehensive review of [11C]PK11195 challengers. Eur J Nucl Med Mol Imaging. 35:2304–2319. 2008. View Article : Google Scholar : PubMed/NCBI

53 

Dolle F, Luus C, Reynolds A and Kassiou M: Radiolabelled molecules for imaging the translocator protein (18 kDa) using positron emission tomography. Curr Med Chem. 16:2899–2923. 2009. View Article : Google Scholar : PubMed/NCBI

54 

Fan Z, Calsolaro V, Atkinson RA, Femminella GD, Waldman A, Buckley C, Trigg W, Brooks DJ, Hinz R and Edison P: Flutriciclamide (18F-GE180) PET: First-in-Human PET study of novel third-generation in vivo marker of human translocator protein. J Nucl Med. 57:1753–1759. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Chiabrando D, Marro S, Mercurio S, Giorgi C, Petrillo S, Vinchi F, Fiorito V, Fagoonee S, Camporeale A, Turco E, et al: The mitochondrial heme exporter FLVCR1b mediates erythroid differentiation. J Clin Invest. 122:4569–4579. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Ni R, Rojdner J, Voytenko L, Dyrks T, Thiele A, Marutle A and Nordberg A: In vitro characterization of the regional binding distribution of amyloid PET tracer florbetaben and the glia tracers deprenyl and PK11195 in Autopsy Alzheimer's brain tissue. J Alzheimers Dis. 80:1723–1737. 2021. View Article : Google Scholar : PubMed/NCBI

57 

Ishikawa M, Yoshitomi T, Covey DF, Zorumski CF and Izumi Y: TSPO activation modulates the effects of high pressure in a rat ex vivo glaucoma model. Neuropharmacology. 111:142–159. 2016. View Article : Google Scholar : PubMed/NCBI

58 

Papadopoulos V and Lecanu L: Translocator protein (18 kDa) TSPO: An emerging therapeutic target in neurotrauma. Exp Neurol. 219:53–57. 2009. View Article : Google Scholar : PubMed/NCBI

59 

Girard C, Liu S, Adams D, Lacroix C, Sinéus M, Boucher C, Papadopoulos V, Rupprecht R, Schumacher M and Groyer G: Axonal regeneration and neuroinflammation: Roles for the translocator protein 18 kDa. J Neuroendocrinol. 24:71–81. 2012. View Article : Google Scholar : PubMed/NCBI

60 

Ravikumar B, Crawford D, Dellovade T, Savinainen A, Graham D, Liere P, Oudinet JP, Webb M and Hering H: Differential efficacy of the TSPO ligands etifoxine and XBD-173 in two rodent models of Multiple Sclerosis. Neuropharmacology. 108:229–237. 2016. View Article : Google Scholar : PubMed/NCBI

61 

Choi HB, Khoo C, Ryu JK, van Breemen E, Kim SU and McLarnon JG: Inhibition of lipopolysaccharide-induced cyclooxygenase-2, tumor necrosis factor-alpha and [Ca2+]i responses in human microglia by the peripheral benzodiazepine receptor ligand PK11195. J Neurochem. 83:546–555. 2002. View Article : Google Scholar : PubMed/NCBI

62 

Lebedeva IV, Su ZZ, Sarkar D, Kitada S, Dent P, Waxman S, Reed JC and Fisher PB: Melanoma differentiation associated gene-7, mda-7/interleukin-24, induces apoptosis in prostate cancer cells by promoting mitochondrial dysfunction and inducing reactive oxygen species. Cancer Res. 63:8138–8144. 2003.PubMed/NCBI

63 

Seneviratne MS, Faccenda D, De Biase V and Campanella M: PK11195 inhibits mitophagy targeting the F1Fo-ATPsynthase in Bcl-2 knock-down cells. Curr Mol Med. 12:476–482. 2012. View Article : Google Scholar : PubMed/NCBI

64 

Bonora M, Giorgi C and Pinton P: Molecular mechanisms and consequences of mitochondrial permeability transition. Nat Rev Mol Cell Biol. 23:266–285. 2022. View Article : Google Scholar : PubMed/NCBI

65 

Gonzalez-Polo RA, Carvalho G, Braun T, Decaudin D, Fabre C, Larochette N, Perfettini JL, Djavaheri-Mergny M, Youlyouz-Marfak I, Codogno P, et al: PK11195 potently sensitizes to apoptosis induction independently from the peripheral benzodiazepin receptor. Oncogene. 24:7503–7513. 2005. View Article : Google Scholar : PubMed/NCBI

66 

Choi J, Ifuku M, Noda M and Guilarte TR: Translocator protein (18 kDa)/peripheral benzodiazepine receptor specific ligands induce microglia functions consistent with an activated state. Glia. 59:219–230. 2011. View Article : Google Scholar : PubMed/NCBI

67 

Awad M and Gavish M: Binding of [3H]Ro 5-4864 and [3H]PK 11195 to cerebral cortex and peripheral tissues of various species: Species differences and heterogeneity in peripheral benzodiazepine binding sites. J Neurochem. 49:1407–1414. 1987. View Article : Google Scholar : PubMed/NCBI

68 

Marangos PJ, Patel J, Boulenger JP and Clark-Rosenberg R: Characterization of peripheral-type benzodiazepine binding sites in brain using [3H]Ro 5-4864. Mol Pharmacol. 22:26–32. 1982.PubMed/NCBI

69 

Kreisl WC, Kim MJ, Coughlin JM, Henter ID, Owen DR and Innis RB: PET imaging of neuroinflammation in neurological disorders. Lancet Neurol. 19:940–950. 2020. View Article : Google Scholar : PubMed/NCBI

70 

Gut P, Baeza-Raja B, Andersson O, Hasenkamp L, Hsiao J, Hesselson D, Akassoglou K, Verdin E, Hirschey MD and Stainier DY: Whole-organism screening for gluconeogenesis identifies activators of fasting metabolism. Nat Chem Biol. 9:97–104. 2013. View Article : Google Scholar : PubMed/NCBI

71 

Da Pozzo E, Giacomelli C, Barresi E, Costa B, Taliani S, Passetti Fda S and Martini C: Targeting the 18-kDa translocator protein: recent perspectives for neuroprotection. Biochem Soc Trans. 43:559–565. 2015. View Article : Google Scholar : PubMed/NCBI

72 

Gavioli EC, Duarte FS, Bressan E, Ferrara P, Farges RC and De Lima TC: Antidepressant-like effect of Ro5-4864, a peripheral-type benzodiazepine receptor ligand, in forced swimming test. Eur J Pharmacol. 471:21–26. 2003. View Article : Google Scholar : PubMed/NCBI

73 

Kita A, Kohayakawa H, Kinoshita T, Ochi Y, Nakamichi K, Kurumiya S, Furukawa K and Oka M: Antianxiety and antidepressant-like effects of AC-5216, a novel mitochondrial benzodiazepine receptor ligand. Br J Pharmacol. 142:1059–1072. 2004. View Article : Google Scholar : PubMed/NCBI

74 

Baez E, Guio-Vega GP, Echeverria V, Sandoval-Rueda DA and Barreto GE: 4′-chlorodiazepam protects mitochondria in T98G astrocyte cell line from glucose deprivation. Neurotox Res. 32:163–171. 2017. View Article : Google Scholar : PubMed/NCBI

75 

Nothdurfter C, Rammes G, Baghai TC, Schüle C, Schumacher M, Papadopoulos V and Rupprecht R: Translocator protein (18 kDa) as a target for novel anxiolytics with a favourable side-effect profile. J Neuroendocrinol. 24:82–92. 2012. View Article : Google Scholar : PubMed/NCBI

76 

Kita A, Kinoshita T, Kohayakawa H, Furukawa K and Akaike A: Lack of tolerance to anxiolysis and withdrawal symptoms in mice repeatedly treated with AC-5216, a selective TSPO ligand. Prog Neuropsychopharmacol Biol Psychiatry. 33:1040–1045. 2009. View Article : Google Scholar : PubMed/NCBI

77 

Karlstetter M, Nothdurfter C, Aslanidis A, Moeller K, Horn F, Scholz R, Neumann H, Weber BH, Rupprecht R and Langmann T: Translocator protein (18 kDa) (TSPO) is expressed in reactive retinal microglia and modulates microglial inflammation and phagocytosis. J Neuroinflammation. 11:32014. View Article : Google Scholar : PubMed/NCBI

78 

Scholz R, Caramoy A, Bhuckory MB, Rashid K, Chen M, Xu H, Grimm C and Langmann T: Targeting translocator protein (18 kDa) (TSPO) dampens pro-inflammatory microglia reactivity in the retina and protects from degeneration. J Neuroinflammation. 12:2012015. View Article : Google Scholar : PubMed/NCBI

79 

Schlichter R, Rybalchenko V, Poisbeau P, Verleye M and Gillardin J: Modulation of GABAergic synaptic transmission by the non-benzodiazepine anxiolytic etifoxine. Neuropharmacology. 39:1523–1535. 2000. View Article : Google Scholar : PubMed/NCBI

80 

Choi YM and Kim KH: Etifoxine for pain patients with anxiety. Korean J Pain. 28:4–10. 2015. View Article : Google Scholar : PubMed/NCBI

81 

Girard C, Liu S, Cadepond F, Adams D, Lacroix C, Verleye M, Gillardin JM, Baulieu EE, Schumacher M and Schweizer-Groyer G: Etifoxine improves peripheral nerve regeneration and functional recovery. Proc Natl Acad Sci USA. 105:20505–20510. 2008. View Article : Google Scholar : PubMed/NCBI

82 

Olsen RW and Li GD: GABA(A) receptors as molecular targets of general anesthetics: Identification of binding sites provides clues to allosteric modulation. Can J Anaesth. 58:206–215. 2011. View Article : Google Scholar : PubMed/NCBI

83 

Gunn BG, Brown AR, Lambert JJ and Belelli D: Neurosteroids and GABA(A) Receptor Interactions: A focus on stress. Front Neurosci. 5:1312011. View Article : Google Scholar : PubMed/NCBI

84 

Biswas L, Ibrahim KS, Li X, Zhou X, Zeng Z, Craft J and Shu X: Effect of a TSPO ligand on retinal pigment epithelial cholesterol homeostasis in high-fat fed mice, implication for age-related macular degeneration. Exp Eye Res. 208:1086252021. View Article : Google Scholar : PubMed/NCBI

85 

Ibrahim KS, Craft JA, Biswas L, Spencer J and Shu X: Etifoxine reverses weight gain and alters the colonic bacterial community in a mouse model of obesity. Biochem Pharmacol. 180:1141512020. View Article : Google Scholar : PubMed/NCBI

86 

Costa B, Cavallini C, Da Pozzo E, Taliani S, Da Settimo F and Martini C: The anxiolytic etifoxine binds to TSPO Ro5-4864 binding site with long residence time showing a high neurosteroidogenic activity. ACS Chem Neurosci. 8:1448–1454. 2017. View Article : Google Scholar : PubMed/NCBI

87 

Kondo D, Saegusa H, Yabe R, Takasaki I, Kurihara T, Zong S and Tanabe T: Peripheral-type benzodiazepine receptor antagonist is effective in relieving neuropathic pain in mice. J Pharmacol Sci. 110:55–63. 2009. View Article : Google Scholar : PubMed/NCBI

88 

Tsukagoshi E, Kawaguchi M, Shinomiya T, Yoshikawa M, Kawano T, Okubo M and Sawaki K: Diazepam enhances production of diazepam-binding inhibitor (DBI), a negative saliva secretion regulator, localized in rat salivary gland. J Pharmacol Sci. 115:221–229. 2011. View Article : Google Scholar

89 

Morin D, Musman J, Pons S, Berdeaux A and Ghaleh B: Mitochondrial translocator protein (TSPO): From physiology to cardioprotection. Biochem Pharmacol. 105:1–13. 2016. View Article : Google Scholar : PubMed/NCBI

90 

Leducq N, Bono F, Sulpice T, Vin V, Janiak P, Fur GL, O'Connor SE and Herbert JM: Role of peripheral benzodiazepine receptors in mitochondrial, cellular, and cardiac damage induced by oxidative stress and ischemia-reperfusion. J Pharmacol Exp Ther. 306:828–837. 2003. View Article : Google Scholar : PubMed/NCBI

91 

Santoro A, Mattace Raso G, Taliani S, Da Pozzo E, Simorini F, Costa B, Martini C, Laneri S, Sacchi A, Cosimelli B, et al: TSPO-ligands prevent oxidative damage and inflammatory response in C6 glioma cells by neurosteroid synthesis. Eur J Pharm Sci. 88:124–131. 2016. View Article : Google Scholar : PubMed/NCBI

92 

Klee K, Storti F, Barben M, Samardzija M, Langmann T, Dunaief J and Grimm C: Systemic knockout of Tspo in mice does not affect retinal morphology, function and susceptibility to degeneration. Exp Eye Res. 188:1078162019. View Article : Google Scholar : PubMed/NCBI

93 

Storti F, Klee K, Todorova V, Steiner R, Othman A, van der Velde-Visser S, Samardzija M, Meneau I, Barben M, Karademir D, et al: Impaired ABCA1/ABCG1-mediated lipid efflux in the mouse retinal pigment epithelium (RPE) leads to retinal degeneration. Elife. 8:e451002019. View Article : Google Scholar : PubMed/NCBI

94 

Santos AM, Calvente R, Tassi M, Carrasco MC, Martín-Oliva D, Marín-Teva JL, Navascués J and Cuadros MA: Embryonic and postnatal development of microglial cells in the mouse retina. J Comp Neurol. 506:224–239. 2008. View Article : Google Scholar : PubMed/NCBI

95 

Lee MA, Sitko AA, Khalid S, Shirasu-Hiza M and Mason CA: Spatiotemporal distribution of glia in and around the developing mouse optic tract. J Comp Neurol. 527:508–521. 2019. View Article : Google Scholar : PubMed/NCBI

96 

Stevens B, Allen NJ, Vazquez LE, Howell GR, Christopherson KS, Nouri N, Micheva KD, Mehalow AK, Huberman AD, Stafford B, et al: The classical complement cascade mediates CNS synapse elimination. Cell. 131:1164–1178. 2007. View Article : Google Scholar : PubMed/NCBI

97 

Prinz M and Mildner A: Microglia in the CNS: Immigrants from another world. Glia. 59:177–187. 2011. View Article : Google Scholar : PubMed/NCBI

98 

Banati RB, Middleton RJ, Chan R, Hatty CR, Kam WW, Quin C, Graeber MB, Parmar A, Zahra D, Callaghan P, et al: Positron emission tomography and functional characterization of a complete PBR/TSPO knockout. Nat Commun. 5:54522014. View Article : Google Scholar : PubMed/NCBI

99 

Barron AM, Ji B, Kito S, Suhara T and Higuchi M: Steroidogenic abnormalities in translocator protein knockout mice and significance in the aging male. Biochem J. 475:75–85. 2018. View Article : Google Scholar : PubMed/NCBI

100 

Grassmann F, Fauser S and Weber BH: The genetics of age-related macular degeneration (AMD)-Novel targets for designing treatment options? Eur J Pharm Biopharm. 95:194–202. 2015. View Article : Google Scholar : PubMed/NCBI

101 

Csader S, Korhonen S, Kaarniranta K and Schwab U: The effect of dietary supplementations on delaying the progression of age-related macular degeneration: A systematic review and meta-analysis. Nutrients. 14:42732022. View Article : Google Scholar : PubMed/NCBI

102 

Fleckenstein M, Keenan TDL, Guymer RH, Chakravarthy U, Schmitz-Valckenberg S, Klaver CC, Wong WT and Chew EY: Age-related macular degeneration. Nat Rev Dis Primers. 7:312021. View Article : Google Scholar : PubMed/NCBI

103 

Pikuleva IA and Curcio CA: Cholesterol in the retina: The best is yet to come. Prog Retin Eye Res. 41:64–89. 2014. View Article : Google Scholar : PubMed/NCBI

104 

Curcio CA, Presley JB, Malek G, Medeiros NE, Avery DV and Kruth HS: Esterified and unesterified cholesterol in drusen and basal deposits of eyes with age-related maculopathy. Exp Eye Res. 81:731–741. 2005. View Article : Google Scholar : PubMed/NCBI

105 

Bhutto I and Lutty G: Understanding age-related macular degeneration (AMD): Relationships between the photoreceptor/retinal pigment epithelium/Bruch's membrane/choriocapillaris complex. Mol Aspects Med. 33:295–317. 2012. View Article : Google Scholar : PubMed/NCBI

106 

Biesemeier A, Taubitz T, Julien S, Yoeruek E and Schraermeyer U: Choriocapillaris breakdown precedes retinal degeneration in age-related macular degeneration. Neurobiol Aging. 35:2562–2573. 2014. View Article : Google Scholar : PubMed/NCBI

107 

Fritsche LG, Igl W, Bailey JN, Grassmann F, Sengupta S, Bragg-Gresham JL, Burdon KP, Hebbring SJ, Wen C, Gorski M, et al: A large genome-wide association study of age-related macular degeneration highlights contributions of rare and common variants. Nat Genet. 48:134–143. 2016. View Article : Google Scholar : PubMed/NCBI

108 

Toomey CB, Johnson LV and Bowes Rickman C: Complement factor H in AMD: Bridging genetic associations and pathobiology. Prog Retin Eye Res. 62:38–57. 2018. View Article : Google Scholar : PubMed/NCBI

109 

Malek G, Johnson LV, Mace BE, Saloupis P, Schmechel DE, Rickman DW, Toth CA, Sullivan PM and Bowes Rickman C: Apolipoprotein E allele-dependent pathogenesis: A model for age-related retinal degeneration. Proc Natl Acad Sci USA. 102:11900–11905. 2005. View Article : Google Scholar : PubMed/NCBI

110 

Toomey CB, Kelly U, Saban DR and Bowes Rickman C: Regulation of age-related macular degeneration-like pathology by complement factor H. Proc Natl Acad Sci USA. 112:E3040–E3049. 2015. View Article : Google Scholar : PubMed/NCBI

111 

Langmann T: Microglia activation in retinal degeneration. J Leukoc Biol. 81:1345–1351. 2007. View Article : Google Scholar : PubMed/NCBI

112 

Zeng H, Ding M, Chen XX and Lu Q: Microglial NADPH oxidase activation mediates rod cell death in the retinal degeneration in rd mice. Neuroscience. 275:54–61. 2014. View Article : Google Scholar : PubMed/NCBI

113 

Zhao L, Zabel MK, Wang X, Ma W, Shah P, Fariss RN, Qian H, Parkhurst CN, Gan WB and Wong WT: Microglial phagocytosis of living photoreceptors contributes to inherited retinal degeneration. EMBO Mol Med. 7:1179–1197. 2015. View Article : Google Scholar : PubMed/NCBI

114 

Farhan F, Almarhoun M, Wong A, Findlay AS, Bartholomew C, Williams MTS, Hurd TW and Shu X: Deletion of TSPO causes dysregulation of cholesterol metabolism in mouse retina. Cells. 10:30662021. View Article : Google Scholar : PubMed/NCBI

115 

Luckoff A, Scholz R, Sennlaub F, Xu H and Langmann T: Comprehensive analysis of mouse retinal mononuclear phagocytes. Nat Protoc. 12:1136–1150. 2017. View Article : Google Scholar : PubMed/NCBI

116 

Nagineni CN, Kommineni VK, William A, Detrick B and Hooks JJ: Regulation of VEGF expression in human retinal cells by cytokines: implications for the role of inflammation in age-related macular degeneration. J Cell Physiol. 227:116–126. 2012. View Article : Google Scholar : PubMed/NCBI

117 

Osborne NN, Casson RJ, Wood JP, Chidlow G, Graham M and Melena J: Retinal ischemia: Mechanisms of damage and potential therapeutic strategies. Prog Retin Eye Res. 23:91–147. 2004. View Article : Google Scholar : PubMed/NCBI

118 

Osborne NN, Ugarte M, Chao M, Chidlow G, Bae JH, Wood JP and Nash MS: Neuroprotection in relation to retinal ischemia and relevance to glaucoma. Surv Ophthalmol. 43 (Suppl 1):S102–S128. 1999. View Article : Google Scholar : PubMed/NCBI

119 

Youngblood H, Robinson R, Sharma A and Sharma S: Proteomic biomarkers of retinal inflammation in diabetic retinopathy. Int J Mol Sci. 20:47552019. View Article : Google Scholar : PubMed/NCBI

120 

Khayat M, Williams M and Lois N: Ischemic retinal vein occlusion: Characterizing the more severe spectrum of retinal vein occlusion. Surv Ophthalmol. 63:816–850. 2018. View Article : Google Scholar : PubMed/NCBI

121 

Heyck M, Bonsack B, Zhang H, Sadanandan N, Cozene B, Kingsbury C, Lee JY and Borlongan CV: The brain and eye: Treating cerebral and retinal ischemia through mitochondrial transfer. Exp Biol Med (Maywood). 244:1485–1492. 2019. View Article : Google Scholar : PubMed/NCBI

122 

Liu GJ, Middleton RJ, Kam WW, Chin DY, Hatty CR, Chan RH and Banati RB: Functional gains in energy and cell metabolism after TSPO gene insertion. Cell Cycle. 16:436–447. 2017. View Article : Google Scholar : PubMed/NCBI

123 

Lieth E, Barber AJ, Xu B, Dice C, Ratz MJ, Tanase D and Strother JM: Glial reactivity and impaired glutamate metabolism in short-term experimental diabetic retinopathy. Penn State Retina Research Group. Diabetes. 47:815–820. 1998. View Article : Google Scholar : PubMed/NCBI

124 

Wagner L, Pannicke T, Rupprecht V, Frommherz I, Volz C, Illes P, Hirrlinger J, Jägle H, Egger V, Haydon PG, et al: Suppression of SNARE-dependent exocytosis in retinal glial cells and its effect on ischemia-induced neurodegeneration. Glia. 65:1059–1071. 2017. View Article : Google Scholar : PubMed/NCBI

125 

Dkhissi O, Chanut E, Wasowicz M, Savoldelli M, Nguyen-Legros J, Minvielle F and Versaux-Botteri C: Retinal TUNEL-positive cells and high glutamate levels in vitreous humor of mutant quail with a glaucoma-like disorder. Invest Ophthalmol Vis Sci. 40:990–995. 1999.PubMed/NCBI

126 

Delyfer MN, Forster V, Neveux N, Picaud S, Leveillard T and Sahel JA: Evidence for glutamate-mediated excitotoxic mechanisms during photoreceptor degeneration in the rd1 mouse retina. Mol Vis. 11:688–696. 2005.PubMed/NCBI

127 

Vujosevic S, Aldington SJ, Silva P, Hernández C, Scanlon P, Peto T and Simó R: Screening for diabetic retinopathy: New perspectives and challenges. Lancet Diabetes Endocrinol. 8:337–347. 2020. View Article : Google Scholar : PubMed/NCBI

128 

Wang W and Lo ACY: Diabetic retinopathy: Pathophysiology and treatments. Int J Mol Sci. 19:18162018. View Article : Google Scholar : PubMed/NCBI

129 

Fung TH, Patel B, Wilmot EG and Amoaku WM: Diabetic retinopathy for the non-ophthalmologist. Clin Med (Lond). 22:112–116. 2022. View Article : Google Scholar : PubMed/NCBI

130 

Brownlee M: The pathobiology of diabetic complications: A unifying mechanism. Diabetes. 54:1615–1625. 2005. View Article : Google Scholar : PubMed/NCBI

131 

Naruse K, Nakamura J, Hamada Y, Nakayama M, Chaya S, Komori T, Kato K, Kasuya Y, Miwa K and Hotta N: Aldose reductase inhibition prevents glucose-induced apoptosis in cultured bovine retinal microvascular pericytes. Exp Eye Res. 71:309–315. 2000. View Article : Google Scholar : PubMed/NCBI

132 

Romeo G, Liu WH, Asnaghi V, Kern TS and Lorenzi M: Activation of nuclear factor-kappaB induced by diabetes and high glucose regulates a proapoptotic program in retinal pericytes. Diabetes. 51:2241–2248. 2002. View Article : Google Scholar : PubMed/NCBI

133 

Abcouwer SF: Muller cell-microglia cross talk drives neuroinflammation in diabetic retinopathy. Diabetes. 66:261–263. 2017. View Article : Google Scholar : PubMed/NCBI

134 

Sorrentino FS, Allkabes M, Salsini G, Bonifazzi C and Perri P: The importance of glial cells in the homeostasis of the retinal microenvironment and their pivotal role in the course of diabetic retinopathy. Life Sci. 162:54–59. 2016. View Article : Google Scholar : PubMed/NCBI

135 

Podesta F, Romeo G, Liu WH, Krajewski S, Reed JC, Gerhardinger C and Lorenzi M: Bax is increased in the retina of diabetic subjects and is associated with pericyte apoptosis in vivo and in vitro. Am J Pathol. 156:1025–1032. 2000. View Article : Google Scholar : PubMed/NCBI

136 

Tien T, Zhang J, Muto T, Kim D, Sarthy VP and Roy S: High glucose induces mitochondrial dysfunction in retinal muller cells: Implications for diabetic retinopathy. Invest Ophthalmol Vis Sci. 58:2915–2921. 2017. View Article : Google Scholar : PubMed/NCBI

137 

Sasaki M, Ozawa Y, Kurihara T, Kubota S, Yuki K, Noda K, Kobayashi S, Ishida S and Tsubota K: Neurodegenerative influence of oxidative stress in the retina of a murine model of diabetes. Diabetologia. 53:971–979. 2010. View Article : Google Scholar : PubMed/NCBI

138 

Li J and Papadopoulos V: Translocator protein (18 kDa) as a pharmacological target in adipocytes to regulate glucose homeostasis. Biochem Pharmacol. 97:99–110. 2015. View Article : Google Scholar : PubMed/NCBI

139 

Musman J, Paradis S, Panel M, Pons S, Barau C, Caccia C, Leoni V, Ghaleh B and Morin D: A TSPO ligand prevents mitochondrial sterol accumulation and dysfunction during myocardial ischemia-reperfusion in hypercholesterolemic rats. Biochem Pharmacol. 142:87–95. 2017. View Article : Google Scholar : PubMed/NCBI

140 

Ciudin A, Simo-Servat O, Hernandez C, Arcos G, Diego S, Sanabria Á, Sotolongo Ó, Hernández I, Boada M and Simó R: Retinal Microperimetry: A new tool for identifying patients with type 2 diabetes at risk for developing Alzheimer disease. Diabetes. 66:3098–3104. 2017. View Article : Google Scholar : PubMed/NCBI

141 

Kinuthia UM, Wolf A and Langmann T: Microglia and inflammatory responses in diabetic retinopathy. Front Immunol. 11:5640772020. View Article : Google Scholar : PubMed/NCBI

142 

Flaxman SR, Bourne RRA, Resnikoff S, Ackland P, Braithwaite T, Cicinelli MV, Das A, Jonas JB, Keeffe J, Kempen JH, et al: Global causes of blindness and distance vision impairment 1990–2020: A systematic review and meta-analysis. Lancet Glob Health. 5:e1221–e1234. 2017. View Article : Google Scholar : PubMed/NCBI

143 

Tham YC, Li X, Wong TY, Quigley HA, Aung T and Cheng CY: Global prevalence of glaucoma and projections of glaucoma burden through 2040: A systematic review and meta-analysis. Ophthalmology. 121:2081–2090. 2014. View Article : Google Scholar : PubMed/NCBI

144 

Zukerman R, Harris A, Oddone F, Siesky B, Verticchio Vercellin A and Ciulla TA: Glaucoma heritability: Molecular mechanisms of disease. Genes (Basel). 12:11352021. View Article : Google Scholar : PubMed/NCBI

145 

Quigley HA: Glaucoma. Lancet. 377:1367–1377. 2011. View Article : Google Scholar : PubMed/NCBI

146 

Ishikawa M, Yoshitomi T, Zorumski CF and Izumi Y: Neurosteroids are endogenous neuroprotectants in an ex vivo glaucoma model. Invest Ophthalmol Vis Sci. 55:8531–8541. 2014. View Article : Google Scholar : PubMed/NCBI

147 

Belelli D and Lambert JJ: Neurosteroids: endogenous regulators of the GABA(A) receptor. Nat Rev Neurosci. 6:565–575. 2005. View Article : Google Scholar : PubMed/NCBI

148 

Weir CJ, Ling AT, Belelli D, Wildsmith JA, Peters JA and Lambert JJ: The interaction of anaesthetic steroids with recombinant glycine and GABAA receptors. Br J Anaesth. 92:704–711. 2004. View Article : Google Scholar : PubMed/NCBI

149 

Midzak A, Akula N, Lecanu L and Papadopoulos V: Novel androstenetriol interacts with the mitochondrial translocator protein and controls steroidogenesis. J Biol Chem. 286:9875–9887. 2011. View Article : Google Scholar : PubMed/NCBI

150 

Izumi Y, Benz AM, Kurby CO, Labruyere J, Zorumski CF, Price MT and Olney JW: An ex vivo rat retinal preparation for excitotoxicity studies. J Neurosci Methods. 60:219–225. 1995. View Article : Google Scholar : PubMed/NCBI

151 

Izumi Y, Kirby CO, Benz AM, Olney JW and Zorumski CF: Muller cell swelling, glutamate uptake, and excitotoxic neurodegeneration in the isolated rat retina. Glia. 25:379–389. 1999. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2024
Volume 29 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yu M and Yu M: Functional role of translocator protein and its ligands in ocular diseases (Review). Mol Med Rep 29: 33, 2024
APA
Yu, M., & Yu, M. (2024). Functional role of translocator protein and its ligands in ocular diseases (Review). Molecular Medicine Reports, 29, 33. https://doi.org/10.3892/mmr.2024.13157
MLA
Yu, M., Zhao, S."Functional role of translocator protein and its ligands in ocular diseases (Review)". Molecular Medicine Reports 29.2 (2024): 33.
Chicago
Yu, M., Zhao, S."Functional role of translocator protein and its ligands in ocular diseases (Review)". Molecular Medicine Reports 29, no. 2 (2024): 33. https://doi.org/10.3892/mmr.2024.13157