Skip to main content

Differential distribution of steroid hormone signaling networks in the human choroid-retinal pigment epithelial complex

Abstract

Background

The retinal pigment epithelium (RPE), a layer of pigmented cells that lies between the neurosensory retina and the underlying choroid, plays a critical role in maintaining the functional integrity of photoreceptor cells and in mediating communication between the neurosensory retina and choroid. Prior studies have demonstrated neurotrophic effects of select steroids that mitigate the development and progression of retinal degenerative diseases via an array of distinct mechanisms of action.

Methods

Here, we identified major steroid hormone signaling pathways and their key functional protein constituents controlling steroid hormone signaling, which are potentially involved in the mitigation or propagation of retinal degenerative processes, from human proteome datasets with respect to their relative abundances in the retinal periphery, macula, and fovea.

Results

Androgen, glucocorticoid, and progesterone signaling networks were identified and displayed differential distribution patterns within these three anatomically distinct regions of the choroid-retinal pigment epithelial complex. Classical and non-classical estrogen and mineralocorticoid receptors were not identified.

Conclusion

Identified differential distribution patterns suggest both selective susceptibility to chronic neurodegenerative disease processes, as well as potential substrates for drug target discovery and novel drug development focused on steroid signaling pathways in the choroid-RPE.

Peer Review reports

Background

The retinal pigment epithelium (RPE) is a layer of pigmented cells that lies between the neurosensory retina and the underlying choroid comprised of the fenestrated choriocapillaris. The location of the RPE allows it to interact with both the neurosensory retina and extra-ocular systems of the body. Given this location, the RPE plays a critical role in mediating the functional communication between the neurosensory retina and the choroid and is also critical to maintaining the functional integrity of photoreceptors [1].

The RPE secretes protective cytokines and growth factors onto the neurosensory retina and conveys waste products to the blood via the choriocapillaris, protecting photoreceptors from ambient insult and also contributes to establishing the immune privilege of the eye by creating a blood-retinal barrier through its tight junctions [1, 2]. The photoreceptors endure daily photo-oxidative damage, which deteriorates the lipid, protein, and opsin contents of the disks. The RPE mediates the essential process of receptor turnover through the phagocytosis of aged photoreceptor disks residing in the photoreceptor outer segments in a diurnal fashion. Nascent phagosomes mature and are resolved within a few hours of the entry of light by healthy RPE, with nutrients and metabolic waste recycled back to photoreceptors or eliminated through the fenestrated choriocapillaris to maintain homeostasis of the retina [3].

The proximally associated choriocapillaris is also critical to maintaining the functional integrity of photoreceptors. These specialized capillaries supply the highly metabolically active photoreceptors and are responsible for removing waste products of metabolism [4].

Given the critical roles these structures play in the visual system, loss of functional integrity of either of these structures could lead to the development of retinal disease.

Understanding the proteomic landscape of the choroid-RPE could provide insight into the pathogenesis of retinal disease and also shed light onto the differential regional susceptibility of the choroid-RPE to the development of disease [5].

Steroids exert neurotrophic effects and may mitigate the development and progression of retinal degenerative disease via an array of mechanisms. Previous studies have shown that ocular tissues express steroid receptors and have demonstrated the neuroprotective effects of several steroids including those of estrogen and glucocorticoids [6]. Here we survey the choroid-RPE proteome dataset for neurosteroids and their pathway constituents from the Skeie and Mahajan, 2014 study, highlighting select pathways potentially involved in neuroprotection and discuss the clinical implications of their relative abundance in regions of the choroid-RPE [5].

Methods

An initial literature review of androgen, estrogen, mineralocorticoid, glucocorticoid, and progesterone signaling in the central nervous system was conducted in order to develop a list of steroid hormone receptors and proteins involved in steroid signaling in the central nervous system (CNS) that could then be surveyed for from prior proteomic analysis datasets of the choroid-RPE complex. This initial protein list consisted primarily of steroid receptors of the classical and non-classical signaling pathways. Identification of mediators in androgen, estrogen, mineralocorticoid, glucocorticoid, or progesterone signaling in the choroid-RPE complex in proteomic analysis datasets then guided a second and more focused literature review. The aim of the second and more focused literature review was to develop a more comprehensive list of proteins involved in the steroid signaling pathways previously identified in the first screen and to identify other related signaling pathways for which to survey for in the selected proteome dataset.

The proteome dataset from which steroid proteins would be identified during the initial screen of steroid protein receptors was from the Skeie and Mahajan, 2014 study [5]. Skeie and Mahajan characterized the proteomic landscape of the human choroid-RPE complex, identifying proteins expressed in the fovea, macula, and periphery. The study identified over four thousand unique proteins in the choroid-RPE complex from the eyes of one man and two women with no known history of ocular disease and whom were all at least in their eighth decade of life. The ocular tissue was obtained within five hours of death. Skeie and Mahajan exploited a multidimensional chromatographic method to obtain deep proteomic coverage, strong anion exchange fractionation followed by online acidic reversed-phase liquid chromatography (LC)-tandem mass spectrometry (MS). Protein identifications were made using X!!Hunter (against the reference library available in 2010 in the global proteome machine), and X!!tandem. For quantitation, the relative abundance of each identified protein in the fovea, macula, and periphery was based on MS/MS spectral counting in the fovea, macula, and periphery datasets, normalized on the basis on total spectral counts (hits) per sample.

Once a classical or non-classical receptor of a steroid was identified in the Skeie and Mahajan [5], dataset, a literature review was conducted to create a more comprehensive list of protein constituents involved in the respective steroid’s classical and non-classical signaling pathways. The final protein list for each steroid hormone is not intended to be an all-encompassing list of proteins involved in all previously identified steroid-mediated signaling pathways, but rather is a list highlighting select pathways and pathway mediators. A guiding set of criteria was used when choosing steroid signaling pathways to highlight in this study. While some identified pathways may not meet all the criteria, they were highlighted in order to either stimulate further discussion and/or because future research may suggest their relevance to steroid signaling in ocular tissue. The following guiding criteria consisted of: (1) the signaling pathway has been demonstrated to be mediated by steroid proteins or are signaling pathways that mediate the effects of steroid hormones, (2) the signaling pathway was identified in the retina or other central nervous system tissue, and (3) priority was given to steroid signaling pathways that have been demonstrated to be involved in either neuroprotective or degenerative processes.

Once the second literature review was completed and a list of steroid signaling pathways and their constituents was completed, these identified proteins were then cross-referenced with the tandem mass spectrometry (MS/MS) data provided by the Skeie and Mahajan [5], study. For the purposes of discussion, some pathway members may be included since it is possible that absent proteins may exist at an abundance below detection threshold in the current datasets.

Proteins that are involved in steroid hormone signaling and that are indeed localized to the choroid-RPE complex are not discussed here if they were either not identified in the proteome dataset or if they were not identified from this study’s literature review.

Here, we highlight proteins that have been previously identified as mediators of steroid hormone signaling that were also identified in the Skeie and Mahajan [5], choroid-RPE complex tissue proteome analysis. We further highlight the relative abundance of these proteins in the choroid-RPE complex and discuss their potential implications in the pathogenesis and treatment of various retinopathies.

Results

After conducting a literature review of androgen, estrogen, mineralocorticoid, progesterone, and glucocorticoid signaling, an initial list of proteins to survey for in the proteomic dataset of the Skeie and Mahajan [5], study was developed. This list consisted primarily of classical and non-classical receptors of steroids: nuclear receptor subfamily 3 group C member 2 (NR3C2), estrogen receptor 1 (ESR1), estrogen receptor 2 (ESR2), estrogen related receptor alpha (ESRRA), estrogen related receptor beta (ESRRB), estrogen related receptor gamma (ESRRG), G protein-coupled estrogen receptor 1 (GPER1), progesterone receptor (PGR), progesterone receptor membrane component 1 (PGRMC1), progesterone receptor membrane component 2 (PGRMC2), progestin and adipoQ receptor family member 7 (PAQR7), progestin and adipoQ receptor family member 8 (PAQR8), progestin and adipoQ receptor family member 5 (PAQR5), progestin and adipoQ receptor family member 6 (PAQR6), progestin and adipoQ receptor family member 9 (PAQR9), nuclear receptor subfamily 3 group C member 1 (NR3C1), androgen receptor (AR), G protein-coupled receptor class C group 6 member A (GPRC6A), solute carrier family 39 member 9 (SLC39A9), cytochrome B5 domain containing 2 (CYB5D2), gamma-aminobutyric acid type A receptor subunit alpha1 (GABRA1), gamma-aminobutyric acid type A receptor subunit alpha2 (GABRA2), gamma-aminobutyric acid type A receptor subunit alpha3 (GABRA3), gamma-aminobutyric acid type A receptor subunit alpha4 (GABRA4), gamma-aminobutyric acid type A receptor subunit alpha5 (GABRA5), gamma-aminobutyric acid type A receptor subunit alpha6 (GABRA6), and neudesin neurotrophic factor (NENF) (refer to Table 1 for further protein information and references).

Table 1 Summary of proteins surveyed for and identified in the Skeie and Mahajan [5], choroid-RPE proteome analysis. The relative abundance of proteins in regions of the choroid-RPE complex is represented by the average peptide hits in the periphery, macula, and fovea. Note, this MS/MS data is provided by the Skeie and Mahajan [5], study (Key: P: Involved in progesterone signaling; A: Involved in androgen signaling; G: Involved in glucocorticoid signaling; E: Involved in estrogen signaling; M: Involved in mineralocorticoid signaling; Y: Yes, identified in dataset; N: No, not identified in dataset). Gene names and abbreviations were verified with the GeneCards database [7]

Of these proteins, the following were identified in the Skeie and Mahajan [5], dataset: AR, NR3C1, PGRMC1, PGRMC2, GABRA1, GABRA2, and NENF (refer to Table 1 for further protein information and references). These identified proteins are involved in androgen, glucocorticoid, and progesterone signaling. This initial survey then guided the second and more focused literature review of androgen, glucocorticoid, and progesterone signaling in the retina and other CNS tissues to develop a more comprehensive list of pathways involved in steroid signaling and their respective constituents. As previously described, select pathways were identified in this study, using the criteria described in the methods section. The list of proteins that was surveyed for in the Skeie and Mahajan [5], dataset can be found in Table 1 and consists of one-hundred-and-one unique proteins. While this list does not include all proteins involved in all identified pathways, it aims to identify key pathway mediators.

The proteome dataset from the Skeie and Mahajan [5], study was then surveyed to identify proteins from the second protein list. Of the one-hundred-and-one unique proteins surveyed for, 39 were identified in the Skeie and Mahajan [5], proteome dataset. The average number of peptides identified in the periphery, macula, and fovea from the MS/MS data of the Skeie and Mahajan [5], study were noted for each protein.

Androgen signaling

Androgen signaling exerts both neuroprotective and neurotoxic effects in CNS tissue via classical and non-classical signaling pathways (as described in the “Androgen signaling” section of the discussion). The classical androgen receptor, AR, was identified in the fovea but not in the macula or periphery. Sex hormone binding globulin was identified in the periphery, macula, and fovea. Androgen metabolites interact with the γ-Aminobutyric acid type A (GABAA) receptor [8]. GABAA receptor subunits were identified in the periphery, macula, and fovea. Solute Carrier Family 39 Member 9 (SLC39A9) and G Protein-Coupled Receptor Class C Group 6 Member A (GPRC6A), non-classical androgen receptors, were not identified in the proteome dataset. Further information regarding protein constituents of androgen signaling can be found in Table 1. The above-mentioned signaling pathways are depicted in Figs. 1 and 2.

Fig. 1
figure 1

A Classical androgen receptor (AR) signaling pathway. When unbound, AR is localized in the cytoplasm associated with molecular chaperones. Binding of AR to its appropriate ligand induces a conformational change that leads to the dissociation of AR from the protein chaperones. AR then interacts with co-regulators that facilitate nuclear targeting and dimerization. AR typically forms a homodimer and is then translocated across the nuclear membrane where it binds to androgen response elements (AREs) and induces the transcription of  androgen-dependent genes [9, 10]. B Sex hormone binding globulin (SHBG) binds androgens and estrogens and transports these steroids in the systemic circulation. SHBG is believed to also be involved in steroid hormone signaling [11]. C 3α-Androstanediol has been shown to interact with the γ-aminobutyric acid (GABA)-A receptor and increase GABA receptor (GBR) function, mediating the GABA-A chloride ion flux demonstrated in Long Evans rats [8]. GABA-A receptor activation has been shown to inhibit retinal regeneration in damaged retina in zebrafish [12]. Pie charts adjacent to proteins represent the protein’s relative abundance in the periphery (blue), macula (orange), and fovea (gray) based on the mass spectrometry data provided by the Skeie and Mahajan [5] study

Fig. 2
figure 2

Evidence suggest that androgens can mediate either neuroprotective or neurotoxic effects. Classical intracellular AR activation mediated phosphorylation of effectors of the MAPK and PI3K-AKT pathways while activation of putative membrane androgen receptors mediated suppression of phosphorylation of MAPK and PI3K-AKT pathway effectors in C6 cells and cortical astrocytes [13, 14]. These studies suggest that consideration of androgen receptor type predominance in select tissues may be warranted when developing androgen pathway targeted interventions. Pie charts adjacent to proteins represent the protein’s relative abundance in the periphery (blue), macula (orange), and fovea (gray) based on the mass spectrometry data provided by the Skeie and Mahajan [5] study

Glucocorticoid signaling

Glucocorticoids modulate inflammation via both genomic and non-genomic signaling (as described in the “Glucocorticoid signaling” section of the discussion). The classical glucocorticoid (GC) receptor, NR3C1, was identified in the fovea but not in the periphery or macula. Glucocorticoid-induced leucine zipper protein (GILZ), a protein product of the classical GC transcription pathway, was not identified in the proteome dataset. GC metabolites regulate GABAA receptor activity [15, 16]. As previously mentioned, GABAA receptor subunits were identified in the periphery, macula, and fovea. Further information regarding protein constituents of glucocorticoid signaling can be found in Table 1. The above-mentioned signaling pathways are depicted in Figs. 3, 4, 5, 6 and 7.

Fig. 3
figure 3

When the classical glucocorticoid receptor (GR) is unbound to its ligand, it is bound to a multiprotein complex of molecular chaperones. Upon binding to its appropriate ligand, GR undergoes conformational changes, dissociating from the molecular chaperones. As a result, its nuclear localization signal is exposed and the GR is translocated to the nucleus [17, 18]. The genomic actions of glucocorticoids include either transcriptional activation of genes or the transcriptional repression of genes. Transactivation consists of activation of glucocorticoid response elements (GREs) and transcription of anti-inflammatory and regulator proteins [19, 20]. The GR complex can also bind to GREs within the GILZ promoter, initiating the transcription of the Glucocorticoid-induced leucine zipper protein (GILZ), a mediator of glucocorticoid effects [21]. Transrepression consists of the GR complex binding to transcription factor subunits, preventing their association to DNA and their co-activators [19, 20];. Glucocorticoids (GCs) are also involved in non-genomic signaling. GCs have been shown to inhibit the GRB2-RAF-MEK1 pathway in A549 cells by blocking GRB2 recruitment [22]. The pathway leads to the downstream phosphorylation of cytosolic phospholipase A2 and consequent liberation of arachidonic acid and pro-inflammatory proteins [22, 23]. Pie charts adjacent to proteins represent the protein’s relative abundance in the periphery (blue), macula (orange), and fovea (gray) based on the mass spectrometry data provided by the Skeie and Mahajan [5] study

Fig. 4
figure 4

VEGF has been demonstrated to be produced and secreted by RPE cells. VEGF-induced angiogenesis is involved in the pathogenesis of several retinopathies including AMD, retinopathy of prematurity, and neovascular glaucoma [24]. VEGFR-1 and VEGFR-2 both bind to VEGF-A [25]. VEGFR-2 exhibits broad pro-angiogenic effects while VEGFR-1 has a greater affinity to its ligand but is more tightly regulated with less potent activity [26]. Pie charts adjacent to proteins represent the protein’s relative abundance in the periphery (blue), macula (orange), and fovea (gray) based on the mass spectrometry data provided by the Skeie and Mahajan [5] study

Fig. 5
figure 5

The GILZ protein is an immune-modulating protein that exerts numerous effects. GILZ inactivates several, transcription factors that regulate T-cell activation and differentiation, including NF-kB [21, 27]. GILZ was also shown to reduce Th1 transcription factor activity and activates Th2 transcription factors in T-cells from GILZ transgenic mice [28]. GILZ was shown to suppress lipopolysaccharide (LPS)-induced expression of several interleukins and chemokines in the retina in male Sprague-Dawley rats with endotoxin-induced uveitis [29]. Suppression of these cytokines inhibited macrophage Inflammatory protein-1 alpha (MIP-1a) which is expressed in the hypoxic inner retina in a C57BL/6 model for ischemia-induced retina neovascularization and may attract microglia that induce angiogenesis, suppressed monocyte chemoattractant protein-1 activity which has also been shown to mediate retinal neovascularization, and as well as supressed interleukin-beta [30,32,33,34]. GILZ suppresses dendritic cell (DC) activation markers and promoted tolerance markers, inhibiting T-cell activation and differentiation [21, 35, 36]. Finally, GILZ has been shown to inhibit interleukin 17 alpha production and subsequent activation of Th17 cells in a murine model for streptozotocin-induced diabetes which increases retinal inflammation [37]. Pie charts adjacent to proteins represent the protein’s relative abundance in the periphery (blue), macula (orange), and fovea (gray) based on the mass spectrometry data provided by the Skeie and Mahajan [5] study

Fig. 6
figure 6

GILZ synergistically acts with TGF-beta to induce SMAD2 phosphorylation and subsequently induce FOXP3 expression [38]. FOXP3 is a transcription factor expressed in a subset of naïve CD4+ T-cells that is necessary for Treg differentiation [39, 40]. Regulatory T-cells (Treg) upregulation may serve as an intervention for posterior uveitis as a study demonstrated that Treg depletion leads to increased susceptibility of the ipsilateral retina to experimental autoimmune uveitis [41, 42]. Tregs were also shown to mediate retinal glial regeneration via production of insulin-like growth factor [43]. Pie charts adjacent to proteins represent the protein’s relative abundance in the periphery (blue), macula (orange), and fovea (gray) based on the mass spectrometry data provided by the Skeie and Mahajan [5] study

Fig. 7
figure 7

Retinal regeneration model. Inhibition of GABA-A receptors in the zebrafish retina induced muller glial (MG)-dependent regeneration [12, 44]. The Kent et al., 2021 [45] study results further support decreasing GABA levels as a potential mechanism for retinal regeneration via MG de-differentiation and generation of progenitor cells. This regenerative pathway, however, may be inhibited by the hippo pathway in mammals. Inhibition of the hippo pathway may induce potential retinal regenerative capacity by adult human MG [46]. Select steroid metabolites have been demonstrated to inhibit GABA-A activity while others exhibited barbiturate-like activity [15, 16]. GABA-A rho has also been shown to be highly expressed in the retina and was thus surveyed for in this study [47]. Pie charts adjacent to proteins represent the protein’s relative abundance in the periphery (blue), macula (orange), and fovea (gray) based on the mass spectrometry data provided by the Skeie and Mahajan [5] study

Progesterone signaling

Progesterone-mediated signaling occurs via both classical and non-classical pathways in CNS tissue (as described in the “Progesterone signaling” section of the discussion). Of all the identified classical and non-classical steroid receptors, PGRMC1 was most highly expressed in the periphery, macula, and fovea. PGRMC1 binding partners, PGRMC2 and SERPINE1 MRNA Binding Protein 1 (SERBP1), were also identified in the proteome dataset [48, 49]. The classical progesterone receptor, PGR, was not identified in the proteome dataset. Progesterone membrane receptors that are members of the progestin and adipoQ receptor family were also not identified in the proteome dataset. Progesterone interacts with the GABAA receptor [50]. GABAA receptor subunits were identified in the periphery, macula, and fovea. Cytochrome B5 domain containing 2 was not identified in the proteome dataset. NENF suggested to be a non-classical progesterone receptor, was only identified in the fovea [51]. Further information regarding protein constituents of progesterone signaling can be found in Table 1. The above-mentioned signaling pathways are depicted in Figs. 8 and 9.

Fig. 8
figure 8

A Non-classical progestin signaling. PGRMC1 has been found to be highly expressed in the 661 W photoreceptor cell line, RPE, and MG [52, 53]. Norgestrel has been shown to exhibit antioxidant properties, preventing photoreceptor damage in a model for RP via increasing the expression of Nrf2, which in response to oxidative stress, binds to DNA anti-oxidant response elements and initiates the transcription of cytoprotective genes [54,55,56]. Huang et al. [57] suggested that it is the phosphorylation of Nrf2 by protein kinase C that induces Nrf2’s nuclear translocation and anti-oxidant effects. B Norgestrel exhibits neuroprotective properties in stressed photoreceptor-like cells and retinal explants via upregulating basic fibroblast growth factor (bFGF) activity via a protein kinase A pathway-dependent mechanism. bFGF phosphorylates and inactivates glycogen synthase kinase 3-beta (GSK3B), preventing the dysregulation of the Nrf2 defense system via preventing the phosphorylating FYN which induces the nuclear export and degradation of Nrf2 [56, 58, 59]. C Norgestrel upregulates fractalkine-CX3CR1 signaling in 661 W cells and C57 explants and fractalkine signaling which mediates norgestrel cytoprotection via reduction of inflammatory cytokine production in rd10 microglia [60]. Pie charts adjacent to proteins represent the protein’s relative abundance in the periphery (blue), macula (orange), and fovea (gray) based on the mass spectrometry data provided by the Skeie and Mahajan [5] study

Fig. 9
figure 9

PGRMC1 signaling. SERBP1 and PGRMC2 may serve as binding partners for PGRMC1 [48, 49]. Progesterone induces BDNF expression via PGRMC1-dependent ERK5 signaling which mediates progesterone’s cytoprotective effects in C6 glial cells and primary astrocytes [61]. The PI3K and ERK pathways have been shown to mediate BDNF neuroprotective effects in response to serum deprivation and DNA damaging agents such as camptothecin in cortical neurons [62]. Pie charts adjacent to proteins represent the protein’s relative abundance in the periphery (blue), macula (orange), and fovea (gray) based on the mass spectrometry data provided by the Skeie and Mahajan [5] study

Estrogen signaling

Estrogens exert variable retinal neuroprotective effects via nuclear estrogen receptor signaling, membrane receptor signaling, as well as through interactions with other intracellular cytoprotective signaling pathways (as described in the in the “Estrogen signaling” section of the discussion). The estrogen receptors, estrogen receptor 1, estrogen receptor 2, estrogen related receptor alpha, estrogen related receptor beta, estrogen related receptor gamma, and G protein-coupled estrogen receptor 1 were not identified in the proteome dataset.

Mineralocorticoid signaling

The mineralocorticoid receptor has been identified previously in the retina and can exert its effects after aldosterone-mediated activation (as described in the “Mineralocorticoid signaling” section of the discussion). The classical mineralocorticoid receptor, NR3C2, was not identified in the proteome dataset.

Discussion

The study identified mediators of the progesterone, androgen, and glucocorticoid signaling pathways, all which may have implications in the pathogenesis or treatment of retinopathies. Classical and non-classical estrogen or mineralocorticoid receptors were not identified in the Skeie and Mahajan [5], proteome dataset, while androgen, glucocorticoid, and progesterone receptors and associated proteins were identified.

Although the present study did not identify estrogen or mineralocorticoid receptors, this lack of identification does not indicate the absence of these receptors in the choroid-RPE complex as the analyzed proteome datasets focused only on identified protein mediators of steroid signaling pathways. Previous studies, for example, have reported the presence of estrogen receptors in the choroid-RPE complex, including estrogen receptor (ER)-alpha and ER-beta [63]. Estrogen’s neuroprotective effects in the RPE and in other CNS tissues has also been well studied [63,64,65,66].

Androgen signaling

The classical androgen receptor, AR, was identified in the fovea, but not in the macula or periphery. AR has been previously identified in the choroid-RPE complex [67]. Androgens are important modulators of neuronal viability, demonstrating both neuroprotective and neurotoxic effects [68,69,70,71].

Androgen-induced neurotoxic effects have been shown to be mediated by AR. Cunningham and colleagues demonstrated testosterone induced apoptosis of N27 neurons, a dopaminergic cell line, via activation of the protein kinase C-delta pathway and that this was dependent upon AR activation [70]. Chronic exposure to androgens has also been suggested to promote mitochondrial dysfunction. Testosterone increases mitochondrial dysfunction in N27 neurons, increasing the levels of reactive oxygen species (ROS) and reducing levels of thiol in N27 cells. These effects were suggested to be dependent upon AR activation [70]. Mitochondrial functioning is important for the regulation of cell death. Mitochondria both generate ROS, which is important for activation of the apoptosis pathway, and generates antioxidant enzymes to protect cells from apoptosis [72]. Appropriate mitochondrial function is necessary to sustain RPE function. Choroid-RPE complex mitochondria are especially vulnerable to oxidative damage and is believed to be intimately involved in the pathogenesis of retinal degeneration [73]. While androgen induced mitochondrial damage has been demonstrated in N27 cells, further research is needed to determine if this activity would be present in the retina. If such an interaction were to be identified in the RPE, this may have implications for the pathogenesis of retinopathies mediated by mitochondrial damage that affect the fovea. Evidence suggests that mitochondrial dysfunction in the choroid-RPE is intimately associated with the development and progression of age-related macular degeneration (AMD) [73].

AR has also played an important role in mediating androgens’ neuroprotective effects [69, 71]. Hammond and colleagues demonstrated that serum deprivation-mediated neuronal apoptosis of human primary neurons was either eliminated entirely or significantly reduced at physiological concentrations of testosterone enanthate. This neuroprotective effect did not require aromatization into estrogens and the neuroprotective effects were prevented when treated with flutamide, suggesting that androgen neuroprotection is mediated via AR [74].

AR has also been involved in mediating non-genomic signaling via activation of second messengers. Activation of AR activates phospholipase C (PLC) via activation of an inhibitory G-protein (GPi). PLC activation may then lead to the rise in intracellular inositol 1,4,5-triphosphate, release of calcium from the sarcoplasmic reticulum, and subsequent regulation of the RAS/MEK/ERK pathway (RAS = “rat sarcoma virus,” a family of GTPases that regulate cell growth and differentiation; MEK = MAPK/ERK kinase; ERK = extracellular-signal regulated kinase) [71]. If this specific interaction between AR and GPi were to be identified in the RPE, it may have important implications for choroid-RPE integrity in the setting of AR dysfunction. Calcium has been demonstrated to regulate the RAS/MEK/ERK pathway, which in many studied tissues, mediates cell survival by regulating the activity of the pro-apoptotic molecules Bad and Bcl-2 (BCL2 = BCL2 apoptosis regulator; bad = BCL2 associated agonist of cell death) [75,76,77]. ERK pathway activation appears to inhibit apoptosis in most tissues, although it has been reported that calcium mediated ERK pathway activation is involved in stress-activated apoptosis of lens epithelial cells [77]. Nguyen and colleagues also demonstrated that androgen-induced neuroprotection in hippocampal neurons was mediated via the androgen-dependent ERK-Rsk (ribosomal s6 kinase)-bad pathway [78].

Gatson and colleagues showed that in addition to phosphorylation of ERK, an effector of the ERK neuroprotective pathway, AR activation also mediates the phosphorylation of AKT, an effector of the phosphoinositide-3-kinase–protein kinase B/Akt (PI3K-PKB/Akt) signaling pathway. The PI3K-Akt signaling pathway is important in regulating cell growth and viability [13].

Androgens interact with non-classical androgen receptors [71]. Expression of sex hormone binding globulin (SHBG), which binds circulating androgens and estrogens was identified in the fovea, macula, and periphery in the proteome dataset. SHBG not only regulates androgen and estrogen bioavailability but also acts as a modulator of unique signaling pathways.

SHBG, for example, inhibits estrogen’s anti-apoptotic effects in MCF-7 cells, a breast cancer cell line. Estrogen phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) mediates estrogen induced anti-apoptotic effects. SHBG, when bound to the SHBG membrane receptor (SHBG-R) and subsequently to its steroid ligand inhibits estrogen’s anti-apoptotic effects through the elevation of cyclic adenosine monophosphate (cAMP) levels. Elevated cAMP, which has been previously demonstrated to suppress ERK1/2 activity, was suggested to reduce the ERK-dependent anti-apoptotic effects of estrogen [79, 80]. Both androgens and estrogens are involved in the formation of the SHBG-receptor(R)—SHBG—steroid complex and subsequent induction of cAMP synthesis and resultant downstream signaling. Estrogens and androgens, however, participate in this complex formation in a cell-type specific manner [81].

The SHBG receptor has also been identified in endometrial cell membranes, placental tissue, and prostatic membranes. To our knowledge, the SHBG-R gene has not yet been characterized [81]. It is possible that the SHBG-R may be present in the RPE but further research is needed to elucidate this. If the SHBG-R is identified in the choroid-RPE, its anti-estrogenic actions could inhibit estrogen-mediated neuroprotective effects in the retina. Again, further research would be needed to demonstrate this interaction in the retina.

Androgen metabolites interact with the γ-Aminobutyric acid type A (GABAA) receptor. GABAA receptor subunits were identified in the proteome dataset. γ-aminobutyric acid type A receptor subunit alpha1 (GABRA1) was identified only in the periphery, while γ-aminobutyric acid type A receptor subunit alpha2 (GABRA2) and γ-aminobutyric acid type A receptor subunit beta2 (GABRB2) were identified in the in the macula and fovea but not in the periphery. Identification of these subunits is important in determining the structure of the GABAA receptor(s) in the RPE and subsequent studying of its specific activity. GABAA receptors are composed of five subunits around a central ion pore [82]. There are fifteen different protein subunits that can form different isoforms of the GABAA receptor. Homomeric isoforms as well as isoforms with different subunits are possible. By identifying GABAA receptors subunits, one may be able to deduce the possible structure of GABAA receptor(s) in regions of the choroid-RPE.

The androgen metabolite, 3α-Androstanediol, interacts with the GABAA receptor and increases the activity of the GABA-receptor complexes (GBRs), resulting in increased chloride ion flux [8]. This may have significance in the development of future therapeutics aimed at treating retinal degeneration as demonstrated by a study that explored the mechanism of GABA-regulated retinal regeneration in zebrafish [12]. GABAA activation was demonstrated to inhibit retinal regeneration in damaged retinas of zebrafish while inhibition of GABAA stimulated regeneration in undamaged retinas via induction of Müller glia (MG) proliferation. MG are able to detect decreased levels of GABA and respond by undergoing differentiation, serving as progenitors in retinal regeneration. Humans have MG in the retina, however, MG-mediated retinal regeneration is inhibited in mammals [12]. Rueda and colleagues demonstrated that the Hippo pathway, by repressing the activity of a transcriptional co-factor called YAP (yes-associated protein), blocked the mammalian MG-mediated retinal regeneration process and that inhibition of the Hippo pathway could stimulate the potentially latent regenerative activity of mammalian MG [46].

Gatson and colleagues provided insight for the differential actions of androgens, those being either mediating neuroprotection or neurotoxicity. It was suggested that the differential actions of androgens in various tissues may be due to the predominance of either AR or membrane androgen receptors [13]. AR activation leads to the activation of both ERK and PI3K-AKT neuroprotective pathways while activation of a putative androgen membrane receptor suppressed ERK and PI3K-AKT pathway activation in primary cortical astrocytes [14]. While the findings of Gatson and colleagues accurately describe the androgen activity in certain neural tissues, it is important to note that AR activation also has injurious effects on cells, as demonstrated by Cunningham and colleagues [70].

Further research elucidating the predominance of certain androgen receptors, as well as research that further explores androgen signaling in the retina may be helpful in the development of androgen-targeted therapeutics aimed at treating retinal degeneration.

Glucocorticoid signaling

Glucocorticoids are widely used as therapeutics for their anti-inflammatory effects [83]. Excessive inflammation is involved in the development and progression of a wide array of diseases including ocular diseases such as AMD, retinitis pigmentosa (RP), and glaucoma [30, 84, 85].

The glucocorticoid classical receptor, NR3C1, was detected in the fovea but not in the macula or periphery. While the lack of detection of NR3C1 in the macula and periphery does not necessitate its absence in these regions, it may suggest that even if NR3C1 is present in these regions, it may be more abundant in the fovea.

Glucocorticoids (GCs) exert their effects via the classical or genomic pathway, as well as via non-genomic (non-classical) pathways. In the classical pathway GCs either transcriptionally activate genes (transactivation) or transcriptionally repress genes (transrepression) [17, 86]. The transactivation pathway activates anti-inflammatory and regulatory proteins [19, 20, 86]. Glucocorticoid-induced leucine zipper (GILZ) is an especially important protein that mediates GC anti-inflammatory effects [21]. GILZ was not identified in the proteome dataset but this may be due to the fact that the samples of ocular tissue were taken from individuals with no known ocular disease and therefore, glucocorticoid receptor activation and resultant increase in GILZ levels would not have been observed.

GILZ, a major effector of the GC transactivation pathway, mediates anti-inflammatory effects via numerous mechanisms. GILZ suppresses the activity of pro-inflammatory transcription factors including NFkB-p65 (subunit of NF-kappa-B transcription complex), activator protein-1 (AP-1), NF-κB (nuclear factor-kappa B), and NFATC2 [21, 27]. These transcription factors were identified in the proteome dataset in macula and fovea, but not in the periphery.

GILZ-mediated glucocorticoid effects suppress lipopolysaccharide (LPS)-induced retinal inflammation, inhibit T-cell differentiation, and increase T-regulatory (Treg) cell expression [21, 29, 35, 38,39,40]. Further, GILZ may suppress T-cell interleukin (IL)-17A production [87]. This is may be clinically important as IL-17A signaling exacerbates vascular permeability, retinal inflammation, and oxidative stress. This was demonstrated in a murine model of streptozotocin-diabetes. It is worth noting that while the IL-17A receptor (IL17RC) was not identified in the proteome dataset, it is constitutively expressed in neural and vascular cells of the retina and has been identified in photoreceptors [37].

A potentially valuable aspect of GILZ is that while it mediates GC effects, its activity may not produce the unfavorable GC-associated metabolic effects [88]. A better understanding of GILZ activity would be useful in the potential future development of therapeutics that mitigate inflammation in ocular tissue while avoiding the GC-induced metabolic effects.

While GILZ activation, as well as activation of other anti-inflammatory and regulatory proteins is hallmark of the transactivation route of GC classical signaling, many GC effects are also mediated by the transrepression pathway. In transrepression, transcription factor activity is inhibited, suppressing the synthesis of pro-inflammatory cytokines including IL-1, IL2, IL-6, IL-8, vascular endothelial growth factor (VEGF), cyclooxygenase-2 (COX-2), prostaglandins, tumor necrosis factor (TNF), and interferon (IFN)-gamma. Transrepression mediated GC anti-inflammatory effects are more clinically desirable and produce less unfavorable metabolic effects [19, 20]. The transrepression mediated activity of GC has important implications in a variety of retinopathies as inflammation is involved in the pathogenesis of AMD, diabetic retinopathy (DR), retinal vein occlusion (RVO), diabetic macular edema, and RP [89,90,91]. Transrepression-dependent GC activity may also have therapeutic effects in retinopathies that exhibit excessive angiogenesis such as AMD, DR, retinopathy of prematurity, sickle cell retinopathy, neovascular glaucoma, and inherited retinopathies [24]. GC can suppress the transcription of VEGF, which has been demonstrated to be secreted and produced by RPE cells, astrocytes, MG, vascular endothelium, and ganglion cells [19, 24]. In the proteome dataset, receptors to VEGF-A, kinase insert domain receptor (KDR), and Fms related receptor tyrosine kinase 1 (FLT1) were primarily identified in the periphery. FLT1, however, was also identified in the macula and fovea. The localization of these VEGF receptors in the retina may suggest that different regions of the choroid-RPE complex may be more susceptible to pathologic angiogenesis.

NR3C1 may also mediate anti-inflammatory effects via non-genomic mechanisms. GR inhibits the GRB2-RAF-MEK1 pathway (GRB2 = growth factor receptor bound protein 2; RAF = RAF Proto-Oncogene Serine/Threonine-Protein Kinase; MEK1 = Mitogen-Activated Protein Kinase Kinase 1) in A549 cells, a human lung adenocarcinoma cell line, by blocking GRB2 recruitment, preventing its association to phosphorylated EGFR and subsequent downstream signaling [22]. Blockage of this signaling inhibits the liberation of arachidonic acid and subsequent synthesis of pro-inflammatory mediators [92].

Different glucocorticoid metabolites interact with the GABAA receptor and either inhibit or stimulate GABA-mediated chloride ion uptake. The glucocorticoid metabolites, allotetrahydrocortisol, tetrahydrocortisol, allotetrahydrocortisone, tetrahydrocortisone, were demonstrated to inhibit GABAA activity in cortical micro sacs of adult male Wistar rats [15]. 3alpha,5alpha-tetrahydrodeoxycorticosterone exhibits barbiturate like activity, potentiating GABAA-mediated increased chloride ion uptake. This was demonstrated in hippocampal and spinal cord neurons in a murine model [16]. As previously discussed, the GABAA subunits that were identified in the proteome dataset include the GABRA1, GABRA2, and GABRB2 subunits. GABRA1 was only identified in the periphery while GABRA2 and GABRB2 were identified in the macula and fovea. These identifications may provide insight to the structure of the GABAA receptor(s) localized in the choroid-RPE complex. Also as previously mentioned, modulation of GABAA activity may have significance in the future development of therapeutics that stimulate MG-dependent retinal regeneration.

Progesterone signaling

The classical progesterone receptor, PGR, was not identified in the proteome dataset but has been previously identified in the retina of rd10 mice (rd10, retinal degeneration 10), a model for retinitis pigmentosa, and C57 wild type mice [58]. Upon binding to its steroid ligand, PGR dimerizes, is translocated into the nucleus, and then modulates gene transcription [93]. PGR upregulates the transcription of the neurotrophic protein, brain-derived neurotrophic factor (BDNF) [94]. BDNF serves as a mediator of progesterone induced neuroprotection against a variety of insults and unfavorable ambient conditions including ischemia, trauma, and glutamate toxicity [94,95,96].

In addition to regulating the transcription of genes through the classical or genomic signaling pathway, progesterone can also interact with non-classical receptors and interact with protein kinases. The non-classical progesterone membrane receptors, PAQR5, PAQR7, PAQR8, were not identified in the dataset, but are expressed in the 661 W photoreceptor cell line [52, 58].

A well-studied progesterone membrane receptor is progesterone receptor membrane component 1 (PGRMC1) and has been previously shown to be expressed in the RPE [53]. Regulation of PGRMC1 activity is crucial for cell viability and has been identified as a regulator of apoptosis. PGRMC1 expression has been demonstrated to be upregulated in the setting of retinal degeneration in the rd10 mouse retina [58]. PGRMC1 has been identified to be co-localized with binding partners progesterone receptor membrane component 2 (PGRMC2) and plasminogen activator inhibitor 1 (SERBP1) [48, 49]. PGRMC1 and SERBP1 were identified in the periphery, macula, and fovea in the proteome dataset. PGRMC2, however, was identified in the macula and fovea but not in the periphery. PGRMC2 does not bind progesterone and its function in progesterone signaling is not as well understood [48, 49].

PGRMC1 was identified in the proteomic dataset and was found to be expressed in the periphery, macula, and fovea. PGRMC1 mediates progesterone induced neuroprotection via numerous mechanisms. One such studied mechanism is PGRMC1-mediated norgestrel induced transcription of antioxidants [54, 97]. In this pathway, PGRMC1, activated by norgestrel (a progestin), induces protein kinase C (PKC) phosphorylation of nuclear factor erythroid 2–related factor 2 (Nrf2) [54]. Phosphorylation of Nrf2 then allows for dissociation of Nrf2 from its cytoplasmic repressor, Kelch-like ECH-associated protein 1 (KEAP1), allowing for Nrf2 to then be translocated into the nucleus, form a heterodimer with a small Maf (sMAF) protein, and then upregulate the transcription of cytoprotective genes [54, 57, 98]. Although Nrf2 and KEAP1 were not identified in the dataset, studies have demonstrated that Nrf2 signaling is present in the RPE. Previous studies have also shown that Nrf2 signaling is impaired in the aging RPE and that compromised Nrf2 antioxidant effects may render the aging RPE to be more vulnerable to oxidative stress-induced damage to the RPE and subsequently develop AMD [99]. Norgestrel also promotes neuroprotection by inhibiting glycogen synthase kinase 3b (GSK3B) signaling [100]. Downstream GSK3B signaling induces the nuclear export and ubiquitination of Nrf2 [59]. Norgestrel inhibits GSK3B signaling via basic fibroblast growth factor and protein kinase A (PKA) [100]. This led to a reduction of inflammatory cytokine production in rd10 microglia [60].

Su and colleagues also demonstrated that PGRMC1 mediated the progesterone-induced release of neurotrophic BDNF in rat C6 glioma cells via activation of the ERK5 signaling pathway [61]. The PI3K-AKT and ERK1/2 pathways may then mediate BDNF neuroprotection as demonstrated in the Harlan Sprague-Dawley rat brain [62, 101]. Members of the PI3K-AKT pathway were identified in the proteome dataset and were found to be expressed in the periphery and macula. Members of the ERK1/2 pathway were identified in the periphery, macula, and fovea.

PGRMC1 also has pro-angiogenic effects. Lange and colleagues demonstrated that PGRMC1 can induce VEGF expression in MG. PGRMC1 was demonstrated to induce calcium influx in MG, leading to PI3K-dependent phosphorylation of protein kinase C and ERK1/2 and downstream transcription of VEGF [102]. While physiologic stimulation of angiogenesis in response to oxygen deprivation may be protective, dysregulation of this VEGF-A expression pathway may lead to pathologic neovascularization in the choroid-RPE.

Progesterone can also interact with another non-classical receptor, the GABAA receptor; Callachan and colleagues demonstrated that select progesterone metabolites can activate the GABAA receptor. This was demonstrated in bovine adrenomedullary chromaffin cells [50]. These findings are consistent with the findings of the Majewska [16] study that showed that progesterone metabolites may be barbiturate-like regulators of GABAA receptor activity.

Visual representations of the identified androgen, glucocorticoid, and progesterone signaling pathways [9, 10, 12,13,14,15, 17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,47, 52,53,54,55,56,57,58,59,60,61,62, 67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,102] are depicted in Figs. 1, 2, 3, 4, 5, 6, 7, 8 and 9.

Lack of identification of classical and non-classical estrogen and mineralocorticoid receptors

While the present study did not identify estrogen and mineralocorticoid receptors in the dataset, these receptors have been previously identified in ocular tissue [63,64,65,66, 103,104,105,106,107,108,109,110,111,112,113,114,115,116].

Estrogen signaling

Estrogen receptor expression in ocular tissue as well as the local production of estrogens in the retina via androgen aromatization and cholesterol-based synthesis has been described extensively [63,64,65,66, 104,105,106,107,108,109,110,111,112,113,114]. ESR1 and ESR2 have been identified in the human RPE-choroid complex and GPER1, a G-protein coupled estrogen receptor, has been localized to the endoplasmic reticulum of CNS cell types including retinal ganglion cells [106, 107, 111,112,113,114].

Studies have provided evidence that estrogens may exert neuroprotective effects in the retina, including promoting RPE cell survival during periods of oxidative stress and exerting neuroprotective effects in the setting of glutamate and N-methyl-D-aspartate (NMDA) toxicity [63,64,65,66, 104,105,106,107,108,109,110,111,112,113,114]. Estrogens may exert neuroprotective effects via nuclear estrogen receptor signaling, membrane receptor signaling, as well as via its interactions with other intracellular pathways [63]. Jiang and colleagues demonstrated that administration GPER1 agonists mitigated murine Müller cell gliosis and retinal ganglion cell apoptosis in the setting of NMDA neurotoxicity and that these effects may be mediated by the PI3K-AKT signaling pathway [110].

Studies have suggested that estrogen receptor expression levels as well as retinal functioning may be positively associated with higher estrogenic states [113, 115]. This apparent association could possibly explain the lack of identification of estrogen receptors from the proteome dataset. The Skeie and Mahajan [5], study utilized ocular tissue from the eyes of one man and two presumably postmenopausal women who were all at least in their eighth decade of life and may have had estrogen receptor expression levels below the threshold for detection. This, however, is only speculative as further studies would be needed to further explore this apparent association.

Mineralocorticoid signaling

While the mineralocorticoid nuclear receptor (MR), NR3C2, was not identified in the dataset, it has been previously identified in the retina, including in ganglion cells, RPE cells, and in vascular cells [103].

Studies have provided evidence for the deleterious effects of high aldosterone levels and the overactivation of MR in the retina. Wilkinson-Berka and colleagues identified aldosterone and its associated receptor, NR3C2, in the pathogenesis of retinal vasculopathy in animal models [103]. The study demonstrated that MR antagonism mitigated angiogenesis, reported MR antagonism’s reduction of retinal leukostasis and aldosterone-mediated tubulogenesis, and provided evidence that increased aldosterone levels may compromise the functioning of the antioxidant glutathione system. Studies have also provided evidence that aldosterone-mediated MR overactivation may be implicated in the pathogenesis of retinal edema via upregulation of ion and water channels involved in retinal fluid homeostasis [116].

Further, Wagner and colleagues provided evidence of an intraocular renin-angiotensin system (RAS) that is distinct from the RAS in systemic circulation and found that the renin gene was most highly expressed in the RPE choroidal layer of enucleated human eyes [117]. This system may be an important regulator of intraocular MR activity.

Reasons as to why the nuclear mineralocorticoid receptor was not identified in the Skeie and Mahajan [5], proteome dataset could include protein levels below the detection threshold, selective degradation of the protein, or both.

Future studies that further elucidate the steroid signaling pathways in the choroid-RPE, may provide valuable insight into both regional differences in disease susceptibility and responsiveness to steroid pathway targeted therapies.

Availability of data and materials

The datasets analyzed during the current study are available in reference [5], Skeie and Mahajan, 2014.

Abbreviations

ANOVA:

Analysis of variance

AMD:

Age-Related Macular Degeneration

AR:

Androgen receptor

bFGF:

Basic fibroblast growth factor

cAMP:

Cyclic adenosine monophosphate

CNS:

Central nervous system

DR:

Diabetic retinopathy

ER:

Estrogen receptor

GC:

Glucocorticoid

GILZ:

Glucocorticoid-induced leucine zipper

GSK3B:

Glycogen synthase kinase 3-beta

LC:

Liquid chromatography

MG:

Müller glia

MS:

Mass spectrometry

Nrf2:

Nuclear factor erythroid 2-related factor 2

PLC:

Phospholipase C

PR:

Progesterone receptor

PGRMC1:

Progesterone receptor membrane component 1

PGRMC2:

Progesterone receptor membrane component 2

ROS:

Reactive oxygen species

RP:

Retinitis pigmentosa

RPE:

Retinal pigment epithelium

RVO:

Retinal vein occlusion

SERBP1:

Plasminogen activator inhibitor 1

References

  1. Strauß O. Anatomy and Pathophysiology of Retinal Pigment Epithelial Detachment (H. Helbig & J. Wachtlin, Eds.). In 2036893326 1412883038 M. A. Gamulescu (Ed.), Retinal Pigment Epithelial Detachment (pp. 1-12). Cham: Springer; 2017.

  2. Forrester JV, Xu H. Good news-bad news: the yin and Yang of immune privilege in the eye. Front Immunol. 2012;3:338. https://doi.org/10.3389/fimmu.2012.00338 PMID: 23230433; PMCID: PMC3515883.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Kwon W, Freeman SA. Phagocytosis by the retinal pigment epithelium: recognition, resolution, recycling. Front Immunol. 2020;11:604205. https://doi.org/10.3389/fimmu.2020.604205 PMID: 33281830; PMCID: PMC7691529.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Zouache MA, Eames I, Klettner CA, Luthert PJ. Form, shape and function: segmented blood flow in the choriocapillaris. Sci Rep. 2016;6:35754. https://doi.org/10.1038/srep35754 PMID: 27779198; PMCID: PMC5078844.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Skeie JM, Mahajan VB. Proteomic landscape of the human choroid-retinal pigment epithelial complex. JAMA Ophthalmol. 2014;132(11):1271–81. https://doi.org/10.1001/jamaophthalmol.2014.2065 PMID: 25058583.

    Article  PubMed  Google Scholar 

  6. Guarneri P, Cascio C, Russo D, D'Agostino S, Drago G, Galizzi G, et al. Neurosteroids in the retina: neurodegenerative and neuroprotective agents in retinal degeneration. Ann N Y Acad Sci. 2003;1007:117–28. https://doi.org/10.1196/annals.1286.012 PMID: 14993046.

    Article  CAS  PubMed  Google Scholar 

  7. Genecards®: The Human Gene Database. GeneCards (Database). Retrieved from https://www.genecards.org

  8. Frye CA, Van Keuren KR, Erskine MS. Behavioral effects of 3 alpha-androstanediol. I: modulation of sexual receptivity and promotion of GABA-stimulated chloride flux. Behav Brain Res. 1996;79(1-2):109–18. https://doi.org/10.1016/0166-4328(96)00004-6 PMID: 8883822.

    Article  CAS  PubMed  Google Scholar 

  9. Lucas-Herald AK, Alves-Lopes R, Montezano AC, Ahmed SF, Touyz RM. Genomic and non-genomic effects of androgens in the cardiovascular system: clinical implications. Clin Sci (Lond). 2017;131(13):1405–18. https://doi.org/10.1042/CS20170090 PMID: 28645930; PMCID: PMC5736922.

    Article  CAS  Google Scholar 

  10. Bennett NC, Gardiner RA, Hooper JD, Johnson DW, Gobe GC. Molecular cell biology of androgen receptor signalling. Int J Biochem Cell Biol. 2010;42(6):813–27. https://doi.org/10.1016/j.biocel.2009.11.013 Epub 2009 Nov 30. PMID: 19931639.

    Article  CAS  PubMed  Google Scholar 

  11. U.S. National Library of Medicine. (n.d.). National Center for Biotechnology Information. https://www.ncbi.nlm.nih.gov/.

  12. Rao MB, Didiano D, Patton JG. Neurotransmitter-Regulated Regeneration in the Zebrafish Retina. Stem Cell Rep. 2017;8(4):831–42. https://doi.org/10.1016/j.stemcr.2017.02.007 Epub 2017 Mar 9. PMID: 28285877; PMCID: PMC5390103.

    Article  CAS  Google Scholar 

  13. Gatson JW, Kaur P, Singh M. Dihydrotestosterone differentially modulates the mitogen-activated protein kinase and the phosphoinositide 3-kinase/Akt pathways through the nuclear and novel membrane androgen receptor in C6 cells. Endocrinology. 2006;147(4):2028–34. https://doi.org/10.1210/en.2005-1395 Epub 2006 Jan 12. Erratum in: Endocrinology. 2008 Jan;149(1):366. PMID: 16410299.

    Article  CAS  PubMed  Google Scholar 

  14. Gatson JW, Singh M. Activation of a membrane-associated androgen receptor promotes cell death in primary cortical astrocytes. Endocrinology. 2007;148(5):2458–64. https://doi.org/10.1210/en.2006-1443 Epub 2007 Feb 15. PMID: 17303658.

    Article  CAS  PubMed  Google Scholar 

  15. Strömberg J, Bäckström T, Lundgren P. Rapid non-genomic effect of glucocorticoid metabolites and neurosteroids on the gamma-aminobutyric acid-a receptor. Eur J Neurosci. 2005;21(8):2083–8. https://doi.org/10.1111/j.1460-9568.2005.04047.x PMID: 15869504.

    Article  PubMed  Google Scholar 

  16. Majewska MD, Harrison NL, Schwartz RD, Barker JL, Paul SM. Steroid hormone metabolites are barbiturate-like modulators of the GABA receptor. Science. 1986;232(4753):1004–7. https://doi.org/10.1126/science.2422758 PMID: 2422758.

    Article  CAS  PubMed  Google Scholar 

  17. Ramamoorthy S, Cidlowski JA. Corticosteroids: Mechanisms of Action in Health and Disease. Rheum Dis Clin N Am. 2016;42(1):15–31. https://doi.org/10.1016/j.rdc.2015.08.002 PMID: 26611548; PMCID: PMC4662771.

    Article  Google Scholar 

  18. Grad I, Picard D. The glucocorticoid responses are shaped by molecular chaperones. Mol Cell Endocrinol. 2007;275(1-2):2–12. https://doi.org/10.1016/j.mce.2007.05.018 Epub 2007 Jun 2. PMID: 17628337.

    Article  CAS  PubMed  Google Scholar 

  19. Stahn C, Buttgereit F. Genomic and nongenomic effects of glucocorticoids. Nat Clin Pract Rheumatol. 2008;4(10):525–33. https://doi.org/10.1038/ncprheum0898 Epub 2008 Sep 2. PMID: 18762788.

    Article  CAS  PubMed  Google Scholar 

  20. Schäcke H, Schottelius A, Döcke WD, Strehlke P, Jaroch S, Schmees N, et al. Dissociation of transactivation from transrepression by a selective glucocorticoid receptor agonist leads to separation of therapeutic effects from side effects. Proc Natl Acad Sci U S A. 2004;101(1):227–32. https://doi.org/10.1073/pnas.0300372101 Epub 2003 Dec 23. PMID: 14694204; PMCID: PMC314167.

    Article  CAS  PubMed  Google Scholar 

  21. Cheng Q, Morand E, Yang YH. Development of novel treatment strategies for inflammatory diseases-similarities and divergence between glucocorticoids and GILZ. Front Pharmacol. 2014;5:169. https://doi.org/10.3389/fphar.2014.00169 PMID: 25100999; PMCID: PMC4102084.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Croxtall JD, Choudhury Q, Flower RJ. Glucocorticoids act within minutes to inhibit recruitment of signalling factors to activated EGF receptors through a receptor-dependent, transcription-independent mechanism. Br J Pharmacol. 2000;130(2):289–98. https://doi.org/10.1038/sj.bjp.0703272 PMID: 10807665; PMCID: PMC1572055.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Haller J, Mikics E, Makara GB. The effects of non-genomic glucocorticoid mechanisms on bodily functions and the central neural system. A critical evaluation of findings. Front Neuroendocrinol. 2008;29(2):273–91. https://doi.org/10.1016/j.yfrne.2007.10.004 Epub 2007 Oct 24. PMID: 18054070.

    Article  CAS  PubMed  Google Scholar 

  24. Penn JS, Madan A, Caldwell RB, Bartoli M, Caldwell RW, Hartnett ME. Vascular endothelial growth factor in eye disease. Prog Retin Eye Res. 2008;27(4):331–71.

    Article  CAS  Google Scholar 

  25. Stuttfeld E, Ballmer-Hofer K. Structure and function of VEGF receptors. IUBMB Life. 2009;61(9):915–22. https://doi.org/10.1002/iub.234 PMID: 19658168.

    Article  CAS  PubMed  Google Scholar 

  26. Rahimi N. Vascular endothelial growth factor receptors: molecular mechanisms of activation and therapeutic potentials. Exp Eye Res. 2006;83(5):1005–16. https://doi.org/10.1016/j.exer.2006.03.019 Epub 2006 May 19. PMID: 16713597; PMCID: PMC1576298.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Srinivasan M, Lahiri N, Thyagarajan A, Witek E, Hickman D, Lahiri DK. Nuclear factor-kappa B: glucocorticoid-induced leucine zipper interface analogs suppress pathology in an Alzheimer's disease model. Alzheimers Dement (N Y). 2018;(4):488–98. https://doi.org/10.1016/j.trci.2018.04.004 PMID: 30338290; PMCID: PMC6186959.

  28. Cannarile L, Fallarino F, Agostini M, Cuzzocrea S, Mazzon E, Vacca C, et al. Increased GILZ expression in transgenic mice up-regulates Th-2 lymphokines. Blood. 2006;107(3):1039–47. https://doi.org/10.1182/blood-2005-05-2183 Epub 2005 Oct 4. PMID: 16204313.

    Article  CAS  PubMed  Google Scholar 

  29. Gu R, Lei B, Shu Q, Li G, Xu G. Glucocorticoid-induced leucine zipper overexpression inhibits lipopolysaccharide-induced retinal inflammation in rats. Exp Eye Res. 2017;165:151–63. https://doi.org/10.1016/j.exer.2017.02.011 Epub 2017 Feb 24. PMID: 28238753.

    Article  CAS  PubMed  Google Scholar 

  30. Yoshida S, Yoshida A, Ishibashi T, Elner SG, Elner VM. Role of MCP-1 and MIP-1alpha in retinal neovascularization during postischemic inflammation in a mouse model of retinal neovascularization. J Leukoc Biol. 2003;73(1):137–44. https://doi.org/10.1189/jlb.0302117 PMID: 12525571.

    Article  CAS  PubMed  Google Scholar 

  31. Dong N, Li X, Xiao L, Yu W, Wang B, Chu L. Upregulation of retinal neuronal MCP-1 in the rodent model of diabetic retinopathy and its function in vitro. Invest Ophthalmol Vis Sci. 2012;53(12):7567–75. https://doi.org/10.1167/iovs.12-9446 PMID: 23010641.

    Article  CAS  PubMed  Google Scholar 

  32. Tuaillon N, Shen DF, Berger RB, Lu B, Rollins BJ, Chan CC. MCP-1 expression in endotoxin-induced uveitis. Invest Ophthalmol Vis Sci. 2002;43(5):1493–8 PMID: 11980865.

    PubMed  Google Scholar 

  33. Jonas JB, Tao Y, Neumaier M, Findeisen P. Monocyte chemoattractant protein 1, intercellular adhesion molecule 1, and vascular cell adhesion molecule 1 in exudative age-related macular degeneration. Arch Ophthalmol. 2010;128(10):1281–6. https://doi.org/10.1001/archophthalmol.2010.227 PMID: 20937997.

    Article  CAS  PubMed  Google Scholar 

  34. Davies MH, Stempel AJ, Powers MR. MCP-1 deficiency delays regression of pathologic retinal neovascularization in a model of ischemic retinopathy. Invest Ophthalmol Vis Sci. 2008;49(9):4195–202. https://doi.org/10.1167/iovs.07-1491 Epub 2008 May 16. PMID: 18487365; PMCID: PMC2679417.

    Article  PubMed  Google Scholar 

  35. Cohen N, Mouly E, Hamdi H, Maillot MC, Pallardy M, Godot V, et al. GILZ expression in human dendritic cells redirects their maturation and prevents antigen-specific T lymphocyte response. Blood. 2006;107(5):2037–44. https://doi.org/10.1182/blood-2005-07-2760 Epub 2005 Nov 17. PMID: 16293609.

    Article  CAS  PubMed  Google Scholar 

  36. Kitajima T, Ariizumi K, Bergstresser PR, Takashima A. A novel mechanism of glucocorticoid-induced immune suppression: the inhibiton of T cell-mediated terminal maturation of a murine dendritic cell line. J Clin Invest. 1996;98(1):142–7. https://doi.org/10.1172/JCI118759 PMID: 8690786; PMCID: PMC507410.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Sigurdardottir S, Zapadka TE, Lindstrom SI, Liu H, Taylor BE, Lee CA, et al. Diabetes-mediated IL-17A enhances retinal inflammation, oxidative stress, and vascular permeability. Cell Immunol. 2019;341:103921. https://doi.org/10.1016/j.cellimm.2019.04.009 Epub 2019 May 2. PMID: 31076079; PMCID: PMC6599623.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Bereshchenko O, Coppo M, Bruscoli S, Biagioli M, Cimino M, Frammartino T, et al. GILZ promotes production of peripherally induced Treg cells and mediates the crosstalk between glucocorticoids and TGF-β signaling. Cell Rep. 2014;7(2):464–75.

    Article  CAS  Google Scholar 

  39. Lee W, Lee GR. Transcriptional regulation and development of regulatory T cells. Exp Mol Med. 2018;50(3):e456.

    Article  CAS  Google Scholar 

  40. Li Z, Li D, Tsun A, Li B. FOXP3+ regulatory T cells and their functional regulation. Cell Mol Immunol. 2015;12(5):558–65.

    Article  Google Scholar 

  41. McPherson SW, Heuss ND, Gregerson DS. Regulation of CD8(+) T cell responses to retinal antigen by local FoxP3(+) regulatory T cells. Front Immunol. 2012;3:166. https://doi.org/10.3389/fimmu.2012.00166 PMID: 22737153; PMCID: PMC3380377.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Ruggieri S, Frassanito MA, Dammacco R, Guerriero S. Treg lymphocytes in autoimmune uveitis. Ocul Immunol Inflamm. 2012;20(4):255–61. https://doi.org/10.3109/09273948.2012.681830 Epub 2012 May 7. PMID: 22564107.

    Article  CAS  PubMed  Google Scholar 

  43. Jahn C, Weidinger G. Regulatory T cells know what is needed to regenerate. Dev Cell. 2017;43(6):651–2. https://doi.org/10.1016/j.devcel.2017.12.010 PMID: 29257945.

    Article  CAS  PubMed  Google Scholar 

  44. Bernardos RL, Barthel LK, Meyers JR, Raymond PA. Late-stage neuronal progenitors in the retina are radial Müller glia that function as retinal stem cells. J Neurosci. 2007;27(26):7028–40. https://doi.org/10.1523/JNEUROSCI.1624-07.2007 PMID: 17596452; PMCID: PMC6672216.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Kent MR, Kara N, Patton JG. Inhibition of GABAA-ρ receptors induces retina regeneration in zebrafish. Neural Regen Res. 2021;16(2):367–74. https://doi.org/10.4103/1673-5374.286972 PMID: 32859800; PMCID: PMC7896201.

    Article  CAS  PubMed  Google Scholar 

  46. Rueda EM, Hall BM, Hill MC, Swinton PG, Tong X, Martin JF, et al. The hippo pathway blocks mammalian retinal Müller glial cell reprogramming. Cell Rep. 2019;27(6):1637–1649.e6. https://doi.org/10.1016/j.celrep.2019.04.047 PMID: 31067451; PMCID: PMC6521882.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Cutting GR, Lu L, O'Hara BF, Kasch LM, Montrose-Rafizadeh C, Donovan DM, et al. Cloning of the gamma-aminobutyric acid (GABA) rho 1 cDNA: a GABA receptor subunit highly expressed in the retina. Proc Natl Acad Sci U S A. 1991;88(7):2673–7. https://doi.org/10.1073/pnas.88.7.2673 PMID: 1849271; PMCID: PMC51300.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Petersen SL, Intlekofer KA, Moura-Conlon PJ, Brewer DN, Del Pino SJ, Lopez JA. Nonclassical progesterone signalling molecules in the nervous system. J Neuroendocrinol. 2013;25(11):991–1001. https://doi.org/10.1111/jne.12060 PMID: 23763432.

    Article  CAS  PubMed  Google Scholar 

  49. Petersen SL, Intlekofer KA, Moura-Conlon PJ, Brewer DN, Del Pino SJ, Lopez JA. Novel progesterone receptors: neural localization and possible functions. Front Neurosci. 2013;7:164. https://doi.org/10.3389/fnins.2013.00164 PMID: 24065878; PMCID: PMC3776953.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Callachan H, Cottrell GA, Hather NY, Lambert JJ, Nooney JM, Peters JA. Modulation of the GABAA receptor by progesterone metabolites. Proceedings of the royal society of London. Series B. Biol Sci. 1987;231(1264):359–69.

    CAS  Google Scholar 

  51. Kimura I, Nakayama Y, Zhao Y, Konishi M, Itoh N. Neurotrophic effects of neudesin in the central nervous system. Front Neurosci. 2013;(7):111. https://doi.org/10.3389/fnins.2013.00111 PMID: 23805070; PMCID: PMC3691553.

  52. Wheway G, Nazlamova L, Turner D, Cross S. 661W photoreceptor cell line as a cell model for studying retinal Ciliopathies. Front Genet. 2019;10:308. https://doi.org/10.3389/fgene.2019.00308 PMID: 31024622; PMCID: PMC6459963.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Shanmugam AK, Mysona BA, Wang J, Zhao J, Tawfik A, Sanders A, et al. Progesterone Receptor Membrane Component 1 (PGRMC1) Expression in Murine Retina. Curr Eye Res. 2016;41(8):1105–12. https://doi.org/10.3109/02713683.2015.1085579 Epub 2015 Dec 7. PMID: 26642738; PMCID: PMC4896862.

    Article  CAS  PubMed  Google Scholar 

  54. Byrne AM, Ruiz-Lopez AM, Roche SL, Moloney JN, Wyse-Jackson AC, Cotter TG. The synthetic progestin norgestrel modulates Nrf2 signaling and acts as an antioxidant in a model of retinal degeneration. Redox Biol. 2016;10:128–39. https://doi.org/10.1016/j.redox.2016.10.002 Epub 2016 Oct 4. PMID: 27744118; PMCID: PMC5065647.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Itoh K, Wakabayashi N, Katoh Y, Ishii T, Igarashi K, Engel JD, et al. Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain. Genes Dev. 1999;13(1):76–86. https://doi.org/10.1101/gad.13.1.76 PMID: 9887101; PMCID: PMC316370.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Sandberg M, Patil J, D'Angelo B, Weber SG, Mallard C. NRF2-regulation in brain health and disease: implication of cerebral inflammation. Neuropharmacology. 2014;79:298–306. https://doi.org/10.1016/j.neuropharm.2013.11.004 Epub 2013 Nov 19. PMID: 24262633; PMCID: PMC3958930.

    Article  CAS  PubMed  Google Scholar 

  57. Huang HC, Nguyen T, Pickett CB. Phosphorylation of Nrf2 at Ser-40 by protein kinase C regulates antioxidant response element-mediated transcription. J Biol Chem. 2002;277(45):42769–74. https://doi.org/10.1074/jbc.M206911200 Epub 2002 Aug 26. PMID: 12198130.

    Article  CAS  PubMed  Google Scholar 

  58. Jackson AC, Roche SL, Byrne AM, Ruiz-Lopez AM, Cotter TG. Progesterone receptor signalling in retinal photoreceptor neuroprotection. J Neurochem. 2016;136(1):63–77. https://doi.org/10.1111/jnc.13388 Epub 2015 Oct 30. PMID: 26447367.

    Article  PubMed  Google Scholar 

  59. Jain AK, Jaiswal AK. GSK-3beta acts upstream of Fyn kinase in regulation of nuclear export and degradation of NF-E2 related factor 2. J Biol Chem. 2007;282(22):16502–10. https://doi.org/10.1074/jbc.M611336200 Epub 2007 Apr 2. PMID: 17403689.

    Article  CAS  PubMed  Google Scholar 

  60. Roche SL, Wyse-Jackson AC, Ruiz-Lopez AM, Byrne AM, Cotter TG. Fractalkine-CX3CR1 signaling is critical for progesterone-mediated neuroprotection in the retina. Sci Rep. 2017;7:43067. https://doi.org/10.1038/srep43067 PMID: 28216676; PMCID: PMC5316933.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Su C, Cunningham RL, Rybalchenko N, Singh M. Progesterone increases the release of brain-derived neurotrophic factor from glia via progesterone receptor membrane component 1 (Pgrmc1)-dependent ERK5 signaling. Endocrinology. 2012;153(9):4389–400. https://doi.org/10.1210/en.2011-2177 Epub 2012 Jul 9. PMID: 22778217; PMCID: PMC3423611.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Hetman M, Kanning K, Cavanaugh JE, Xia Z. Neuroprotection by brain-derived neurotrophic factor is mediated by extracellular signal-regulated kinase and phosphatidylinositol 3-kinase. J Biol Chem. 1999;274(32):22569–80. https://doi.org/10.1074/jbc.274.32.22569 PMID: 10428835.

    Article  CAS  PubMed  Google Scholar 

  63. Yu X, Tang Y, Li F, Frank MB, Huang H, Dozmorov I, et al. Protection against hydrogen peroxide-induced cell death in cultured human retinal pigment epithelial cells by 17beta-estradiol: a differential gene expression profile. Mech Ageing Dev. 2005;126(11):1135–45. https://doi.org/10.1016/j.mad.2005.05.005 PMID: 16029884.

    Article  CAS  PubMed  Google Scholar 

  64. Behl C, Holsboer F. The female sex hormone oestrogen as a neuroprotectant. Trends Pharmacol Sci. 1999;20(11):441–4. https://doi.org/10.1016/s0165-6147(99)01392-9 PMID: 10542441.

    Article  CAS  PubMed  Google Scholar 

  65. Brann DW, Dhandapani K, Wakade C, Mahesh VB, Khan MM. Neurotrophic and neuroprotective actions of estrogen: basic mechanisms and clinical implications. Steroids. 2007;72(5):381–405. https://doi.org/10.1016/j.steroids.2007.02.003 Epub 2007 Feb 21. PMID: 17379265; PMCID: PMC2048656.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Nixon E, Simpkins JW. Neuroprotective effects of nonfeminizing estrogens in retinal photoreceptor neurons. Invest Ophthalmol Vis Sci. 2012;53(8):4739–47. https://doi.org/10.1167/iovs.12-9517 PMID: 22700711; PMCID: PMC4625827.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Rocha EM, Wickham LA, da Silveira LA, Krenzer KL, Yu FS, Toda I, et al. Identification of androgen receptor protein and 5alpha-reductase mRNA in human ocular tissues. Br J Ophthalmol. 2000;84(1):76–84. https://doi.org/10.1136/bjo.84.1.76 PMID: 10611104; PMCID: PMC1723240.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Biaek M, Zaremba P, Borowicz KK, Czuczwar SJ. Neuroprotective role of testosterone in the nervous system. Pol J Pharmacol. 2004;56:509–18.

    Google Scholar 

  69. Avsar O. Is testosterone perspective available for neurodegenerative diseases. Neuropsychiatry. 2018;8(5):1482–9.

    Article  Google Scholar 

  70. Cunningham RL, Giuffrida A, Roberts JL. Androgens induce dopaminergic neurotoxicity via caspase-3-dependent activation of protein kinase Cdelta. Endocrinology. 2009;150(12):5539–48. https://doi.org/10.1210/en.2009-0640 Epub 2009 Oct 16. PMID: 19837873; PMCID: PMC2795716.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Pomara C, Neri M, Bello S, Fiore C, Riezzo I, Turillazzi E. Neurotoxicity by synthetic androgen steroids: oxidative stress, apoptosis, and neuropathology: a review. Curr Neuropharmacol. 2015;13(1):132–45. https://doi.org/10.2174/1570159X13666141210221434 PMID: 26074748; PMCID: PMC4462038.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Ott M, Gogvadze V, Orrenius S, Zhivotovsky B. Mitochondria, oxidative stress and cell death. Apoptosis. 2007;12(5):913–22.

    Article  CAS  Google Scholar 

  73. Barot M, Gokulgandhi MR, Mitra AK. Mitochondrial dysfunction in retinal diseases. Curr Eye Res. 2011;36(12):1069–77. https://doi.org/10.3109/02713683.2011.607536 Epub 2011 Oct 6. PMID: 21978133; PMCID: PMC4516173.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Hammond J, Le Q, Goodyer C, Gelfand M, Trifiro M, LeBlanc A. Testosterone-mediated neuroprotection through the androgen receptor in human primary neurons. J Neurochem. 2001;77(5):1319–26. https://doi.org/10.1046/j.1471-4159.2001.00345.x PMID: 11389183.

    Article  CAS  PubMed  Google Scholar 

  75. Chang F, Steelman LS, Shelton JG, Lee JT, Navolanic PM, Blalock WL, et al. Regulation of cell cycle progression and apoptosis by the Ras/Raf/MEK/ERK pathway (review). Int J Oncol. 2003;22(3):469–80 PMID: 12579299.

    CAS  PubMed  Google Scholar 

  76. Chuderland D, Seger R. Calcium regulates ERK signaling by modulating its protein-protein interactions. Commun Integr Biol. 2008;1(1):4–5. https://doi.org/10.4161/cib.1.1.6107 PMID: 19704446; PMCID: PMC2633783.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Li DW, Liu JP, Mao YW, Xiang H, Wang J, Ma WY, et al. Calcium-activated RAF/MEK/ERK signaling pathway mediates p53-dependent apoptosis and is abrogated by alpha B-crystallin through inhibition of RAS activation. Mol Biol Cell. 2005;16(9):4437–53. https://doi.org/10.1091/mbc.e05-01-0010 Epub 2005 Jul 6. PMID: 16000378; PMCID: PMC1196350.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Nguyen TV, Yao M, Pike CJ. Androgens activate mitogen-activated protein kinase signaling: role in neuroprotection. J Neurochem. 2005;94(6):1639–51. https://doi.org/10.1111/j.1471-4159.2005.03318.x Epub 2005 Jul 11. PMID: 16011741.

    Article  CAS  PubMed  Google Scholar 

  79. Catalano MG, Frairia R, Boccuzzi G, Fortunati N. Sex hormone-binding globulin antagonizes the anti-apoptotic effect of estradiol in breast cancer cells. Mol Cell Endocrinol. 2005;230(1-2):31–7. https://doi.org/10.1016/j.mce.2004.11.005 PMID: 15664449.

    Article  CAS  PubMed  Google Scholar 

  80. Nakhla AM, Khan MS, Rosner W. Biologically active steroids activate receptor-bound human sex hormone-binding globulin to cause LNCaP cells to accumulate adenosine 3′,5′-monophosphate. J Clin Endocrinol Metab. 1990;71(2):398–404. https://doi.org/10.1210/jcem-71-2-398 PMID: 2166070.

    Article  CAS  PubMed  Google Scholar 

  81. Kahn SM, Hryb DJ, Nakhla AM, Romas NA, Rosner W. Sex hormone-binding globulin is synthesized in target cells. J Endocrinol. 2002;175(1):113–20. https://doi.org/10.1677/joe.0.1750113 PMID: 12379495.

    Article  CAS  PubMed  Google Scholar 

  82. Sigel E, Steinmann ME. Structure, function, and modulation of GABA(A) receptors. J Biol Chem. 2012;287(48):40224–31. https://doi.org/10.1074/jbc.R112.386664 Epub 2012 Oct 4. PMID: 23038269; PMCID: PMC3504738.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Barnes PJ. Anti-inflammatory actions of glucocorticoids: molecular mechanisms. Clin Sci (Lond). 1998;94(6):557–72. https://doi.org/10.1042/cs0940557 PMID: 9854452.

    Article  CAS  Google Scholar 

  84. Kim SY, Kambhampati SP, Bhutto IA, McLeod DS, Lutty GA, Kannan RM. Evolution of oxidative stress, inflammation and neovascularization in the choroid and retina in a subretinal lipid induced age-related macular degeneration model. Exp Eye Res. 2021;203:108391. https://doi.org/10.1016/j.exer.2020.108391 Epub 2020 Dec 8. PMID: 33307075.

    Article  CAS  PubMed  Google Scholar 

  85. Vohra R, Tsai JC, Kolko M. The role of inflammation in the pathogenesis of glaucoma. Surv Ophthalmol. 2013;58(4):311–20. https://doi.org/10.1016/j.survophthal.2012.08.010 PMID: 23768921.

    Article  PubMed  Google Scholar 

  86. Sulaiman RS, Kadmiel M, Cidlowski JA. Glucocorticoid receptor signaling in the eye. Steroids. 2018;133:60–6. https://doi.org/10.1016/j.steroids.2017.11.002 Epub 2017 Nov 10. PMID: 29129720; PMCID: PMC5875721.

    Article  CAS  PubMed  Google Scholar 

  87. Ngo D, Beaulieu E, Gu R, Leaney A, Santos L, Fan H, et al. Divergent effects of endogenous and exogenous glucocorticoid-induced leucine zipper in animal models of inflammation and arthritis. Arthritis Rheum. 2013;65(5):1203–12. https://doi.org/10.1002/art.37858 PMID: 23335223.

    Article  CAS  PubMed  Google Scholar 

  88. Flynn JK, Dankers W, Morand EF. Could GILZ be the answer to glucocorticoid toxicity in lupus? Front Immunol. 2019;10:1684. https://doi.org/10.3389/fimmu.2019.01684 PMID: 31379872; PMCID: PMC6652235.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Whitcup SM, Nussenblatt RB, Lightman SL, Hollander DA. Inflammation in retinal disease. Int J Inf Secur. 2013;2013:724648. https://doi.org/10.1155/2013/724648 Epub 2013 Sep 10. PMID: 24109539; PMCID: PMC3784265.

    Article  Google Scholar 

  90. Kauppinen A, Paterno JJ, Blasiak J, Salminen A, Kaarniranta K. Inflammation and its role in age-related macular degeneration. Cell Mol Life Sci. 2016;73(9):1765–86. https://doi.org/10.1007/s00018-016-2147-8 Epub 2016 Feb 6. PMID: 26852158; PMCID: PMC4819943.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Rangasamy S, McGuire PG, Das A. Diabetic retinopathy and inflammation: novel therapeutic targets. Middle East Afr J Ophthalmol. 2012;19(1):52–9. https://doi.org/10.4103/0974-9233.92116 PMID: 22346115; PMCID: PMC3277025.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Higgins AJ, Lees P. The acute inflammatory process, arachidonic acid metabolism and the mode of action of anti-inflammatory drugs. Equine Vet J. 1984;16(3):163–75. https://doi.org/10.1111/j.2042-3306.1984.tb01893.x PMID: 6428879.

    Article  CAS  PubMed  Google Scholar 

  93. Pedroza DA, Subramani R, Lakshmanaswamy R. Classical and non-classical progesterone signaling in breast cancers. Cancers (Basel). 2020;12(9):2440. https://doi.org/10.3390/cancers12092440 PMID: 32867363; PMCID: PMC7563480.

    Article  CAS  Google Scholar 

  94. Jodhka PK, Kaur P, Underwood W, Lydon JP, Singh M. The differences in neuroprotective efficacy of progesterone and medroxyprogesterone acetate correlate with their effects on brain-derived neurotrophic factor expression. Endocrinology. 2009;150(7):3162–8. https://doi.org/10.1210/en.2008-1247 Epub 2009 Mar 26. PMID: 19325006; PMCID: PMC2703540.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Kaur P, Jodhka PK, Underwood WA, Bowles CA, de Fiebre NC, de Fiebre CM, et al. Progesterone increases brain-derived neuroptrophic factor expression and protects against glutamate toxicity in a mitogen-activated protein kinase- and phosphoinositide-3 kinase-dependent manner in cerebral cortical explants. J Neurosci Res. 2007;85(11):2441–9. https://doi.org/10.1002/jnr.21370 PMID: 17549730; PMCID: PMC2693123.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Singh M, Su C. Progesterone, brain-derived neurotrophic factor and neuroprotection. Neuroscience. 2013;239:84–91. https://doi.org/10.1016/j.neuroscience.2012.09.056 Epub 2012 Oct 2. PMID: 23036620; PMCID: PMC3582842.

    Article  CAS  PubMed  Google Scholar 

  97. Nguyen T, Nioi P, Pickett CB. The Nrf2-antioxidant response element signaling pathway and its activation by oxidative stress. J Biol Chem. 2009;284(20):13291–5. https://doi.org/10.1074/jbc.R900010200 Epub 2009 Jan 30. PMID: 19182219; PMCID: PMC2679427.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Doonan F, O'Driscoll C, Kenna P, Cotter TG. Enhancing survival of photoreceptor cells in vivo using the synthetic progestin Norgestrel. J Neurochem. 2011;118(5):915–27. https://doi.org/10.1111/j.1471-4159.2011.07354.x Epub 2011 Jul 18. PMID: 21689103.

    Article  CAS  PubMed  Google Scholar 

  99. Sachdeva MM, Cano M, Handa JT. Nrf2 signaling is impaired in the aging RPE given an oxidative insult. Exp Eye Res. 2014;119:111–4. https://doi.org/10.1016/j.exer.2013.10.024 Epub 2013 Nov 8. PMID: 24216314; PMCID: PMC3946784.

    Article  CAS  PubMed  Google Scholar 

  100. Wyse Jackson AC, Cotter TG. The synthetic progesterone Norgestrel is neuroprotective in stressed photoreceptor-like cells and retinal explants, mediating its effects via basic fibroblast growth factor, protein kinase a and glycogen synthase kinase 3β signalling. Eur J Neurosci. 2016;43(7):899–911. https://doi.org/10.1111/ejn.13166 Epub 2016 Feb 11. PMID: 26750157.

    Article  PubMed  Google Scholar 

  101. Han BH, Holtzman DM. BDNF protects the neonatal brain from hypoxic-ischemic injury in vivo via the ERK pathway. J Neurosci. 2000;20(15):5775–81. https://doi.org/10.1523/JNEUROSCI.20-15-05775.2000 PMID: 10908618; PMCID: PMC6772561.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Swiatek-De Lange M, Stampfl A, Hauck SM, Zischka H, Gloeckner CJ, Deeg CA, et al. Membrane-initiated effects of progesterone on calcium dependent signaling and activation of VEGF gene expression in retinal glial cells. Glia. 2007;55(10):1061–73. https://doi.org/10.1002/glia.20523 PMID: 17551930.

    Article  PubMed  Google Scholar 

  103. Wilkinson-Berka, J. L., Tan, G., Jaworski, K., Harbig, J., & Miller, A. G. (2009). Identification of a retinal aldosterone system and the protective effects of mineralocorticoid receptor antagonism on retinal vascular pathology. Circ Res, 104(1), 124–133. https://doi.org/10.1161/circresaha.108.176008.

  104. Guarneri, P., Guarneri, R., Cascio, C., Pavasant, P., Piccoli, F., & Papadopoulos, V. (2002). Neurosteroidogenesis in rat retinas. J Neurochem, 63(1), 86–96. https://doi.org/10.1046/j.1471-4159.1994.63010086.x.

  105. Neumann, F., Wurm, A., Linnertz, R., Pannicke, T., Iandiev, I., Wiedemann, P., Reichenbach, A., & Bringmann, A. (2009). Sex steroids inhibit osmotic swelling of retinal glial cells. Neurochem Res, 35(4), 522–530. https://doi.org/10.1007/s11064-009-0092-8.

  106. Gingerich, S., Kim, G. L., Chalmers, J. A., Koletar, M. M., Wang, X., Wang, Y., & Belsham, D. D. (2010). Estrogen receptor alpha and G-protein coupled receptor 30 mediate the neuroprotective effects of 17β-estradiol in novel murine hippocampal cell models. Neuroscience, 170(1), 54–66. https://doi.org/10.1016/j.neuroscience.2010.06.076.

  107. Liu S-B, Han J, Zhang N, Tian Z, Li X-B, Zhao M-G. Neuroprotective effects of oestrogen against oxidative toxicity through activation of G-protein-coupled receptor 30 receptor. Clin Exp Pharmacol Physiol. 2011;38(9):577–85. https://doi.org/10.1111/j.1440-1681.2011.05549.x.

    Article  CAS  PubMed  Google Scholar 

  108. Zhou X, Li F, Ge J, Sarkisian SR, Tomita H, Zaharia A, et al. Retinal ganglion cell protection by 17-β-estradiol in a mouse model of inherited glaucoma. Dev Neurobiol. 2007;67(5):603–16. https://doi.org/10.1002/dneu.20373.

    Article  CAS  PubMed  Google Scholar 

  109. Yamashita H, Sugihara K, Yamada C, Tsutsumi S, Iwaki Y. Effect of estrogen on electroretinographic responses in streptozotocin-induced diabetic female rats. Exp Eye Res. 2010;90(5):591–7. https://doi.org/10.1016/j.exer.2010.02.003.

    Article  CAS  PubMed  Google Scholar 

  110. Jiang M, Ma X, Zhao Q, Li Y, Xing Y, Deng Q, et al. The neuroprotective effects of novel estrogen receptor GPER1 in mouse retinal ganglion cell degeneration. Exp Eye Res. 2019;189:107826. https://doi.org/10.1016/j.exer.2019.107826.

    Article  CAS  PubMed  Google Scholar 

  111. Kobayashi K, Kobayashi H, Ueda M, Honda Y. Estrogen receptor expression in bovine and rat retinas. Invest Ophthalmol Vis Sci. 1998;39(11):2105–10.

    CAS  PubMed  Google Scholar 

  112. Wickham, L. A., Gao, J., Toda, I., Rocha, E. M., Ono, M., & Sullivan, D. A. (2000). Identification of androgen, estrogen and progesterone receptor mrnas in the eye. Acta Ophthalmol Scand, 78(2), 146–153. https://doi.org/10.1034/j.1600-0420.2000.078002146.x.

  113. Ogueta SB, Schwartz SD, Yamashita CK, Farber DB. Estrogen receptor in the human eye: influence of gender and age on gene expression. Invest Ophthalmol Vis Sci. 1999;40(9):1906–11.

    CAS  PubMed  Google Scholar 

  114. Munaut C. Presence of oestrogen receptor type beta in human retina. Br J Ophthalmol. 2001;85(7):877–82. https://doi.org/10.1136/bjo.85.7.877.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Ozawa GY, Bearse MA, Harrison WW, Bronson-Castain KW, Schneck ME, Barez S, et al. Differences in neuroretinal function between adult males and females. Optom Vis Sci. 2014;91(6):602–7. https://doi.org/10.1097/opx.0000000000000255.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Zhao M, Valamanesh F, Celerier I, Savoldelli M, Jonet L, Jeanny JC, et al. The neuroretina is a novel mineralocorticoid target: aldosterone up-regulates ion and water channels in Müller glial cells. FASEB J. 2010;24(9):3405–15. https://doi.org/10.1096/fj.09-154344.

    Article  CAS  PubMed  Google Scholar 

  117. Wagner J, Jan Danser AH, Derkx FH, de Jong TV, Paul M, Mullins JJ, et al. Demonstration of renin mrna, angiotensinogen mrna, and angiotensin converting enzyme mrna expression in the human eye: evidence for an intraocular renin-angiotensin system. Br J Ophthalmol. 1996;80(2):159–63. https://doi.org/10.1136/bjo.80.2.159.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Support by Nelson R. Sabates is gratefully acknowledged. The authors thank Margaret, Richard and Sara Koulen for generous support and encouragement.

Funding

Research reported in this publication was supported in part by a grant from the National Eye Institute (EY030747) of the National Institutes of Health (PK). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. Additional support by the Felix and Carmen Sabates Missouri Endowed Chair in Vision Research, a Challenge Grant from Research to Prevent Blindness and the Vision Research Foundation of Kansas City is gratefully acknowledged.

Author information

Authors and Affiliations

Authors

Contributions

PK conceived and designed the study. PK and SMG performed the experiments. All authors analyzed the data. SMG and PK wrote the paper. All authors edited and reviewed the paper. The author(s) read and approved the final manuscript.

Corresponding author

Correspondence to Peter Koulen.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Galindez, S.M., Keightley, A. & Koulen, P. Differential distribution of steroid hormone signaling networks in the human choroid-retinal pigment epithelial complex. BMC Ophthalmol 22, 406 (2022). https://doi.org/10.1186/s12886-022-02585-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12886-022-02585-7

Keywords